chapter N ucleotides have a variety of roles in cellular metab- olism. They are the energy currency in metabolic transactions, the essential chemical links in the re- sponse of cells to hormones and other extracellular stim- uli, and the structural components of an array of en- zyme cofactors and metabolic intermediates. And, last but certainly not least, they are the constituents of nu- cleic acids: deoxyribonucleic acid (DNA) and ribonu- cleic acid (RNA), the molecular repositories of genetic information. The structure of every protein, and ulti- mately of every biomolecule and cellular component, is a product of information programmed into the nu- cleotide sequence of a cell’s nucleic acids. The ability to store and transmit genetic information from one gener- ation to the next is a fundamental condition for life. This chapter provides an overview of the chemical nature of the nucleotides and nucleic acids found in most cells; a more detailed examination of the function of nucleic acids is the focus of Part III of this text. 8.1 Some Basics Nucleotides, Building Blocks of Nucleic Acids The amino acid sequence of every protein in a cell, and the nucleotide sequence of every RNA, is specified by a nucleotide se- quence in the cell’s DNA. A segment of a DNA molecule that contains the information required for the synthesis of a functional biological product, whether protein or RNA, is referred to as a gene. A cell typically has many thousands of genes, and DNA molecules, not surpris- ingly, tend to be very large. The storage and transmis- sion of biological information are the only known func- tions of DNA. RNAs have a broader range of functions, and sev- eral classes are found in cells. Ribosomal RNAs (rRNAs) are components of ribosomes, the complexes that carry out the synthesis of proteins. Messenger RNAs (mRNAs) are intermediaries, carrying genetic information from one or a few genes to a ribosome, where the corresponding proteins can be synthesized. Transfer RNAs (tRNAs) are adapter molecules that faithfully translate the information in mRNA into a specific sequence of amino acids. In addition to these major classes there is a wide variety of RNAs with spe- cial functions, described in depth in Part III. Nucleotides and Nucleic Acids Have Characteristic Bases and Pentoses Nucleotides have three characteristic components: (1) a nitrogenous (nitrogen-containing) base, (2) a pen- tose, and (3) a phosphate (Fig. 8–1). The molecule with- out the phosphate group is called a nucleoside. The nitrogenous bases are derivatives of two parent com- pounds, pyrimidine and purine. The bases and pentoses of the common nucleotides are heterocyclic compounds. The carbon and nitrogen atoms in the parent structures are conventionally numbered to facilitate the naming and identification of the many derivative compounds. The convention for the pentose ring follows rules out- lined in Chapter 7, but in the pentoses of nucleotides NUCLEOTIDES AND NUCLEIC ACIDS 8.1 Some Basics 273 8.2 Nucleic Acid Structure 279 8.3 Nucleic Acid Chemistry 291 8.4 Other Functions of Nucleotides 300 A structure this pretty just had to exist. —James Watson, The Double Helix, 1968 8 273 and nucleosides the carbon numbers are given a prime (H11032) designation to distinguish them from the numbered atoms of the nitrogenous bases. The base of a nucleotide is joined covalently (at N-1 of pyrimidines and N-9 of purines) in an N-H9252-glycosyl bond to the 1H11032 carbon of the pentose, and the phosphate is esterified to the 5H11032 carbon. The N-H9252-glycosyl bond is formed by removal of the elements of water (a hydroxyl group from the pentose and hydrogen from the base), as in O-glycosidic bond formation (see Fig. 7–31). Both DNA and RNA contain two major purine bases, adenine (A) and guanine (G), and two major pyrim- idines. In both DNA and RNA one of the pyrimidines is cytosine (C), but the second major pyrimidine is not the same in both: it is thymine (T) in DNA and uracil (U) in RNA. Only rarely does thymine occur in RNA or uracil in DNA. The structures of the five major bases are shown in Figure 8–2, and the nomenclature of their corresponding nucleotides and nucleosides is summa- rized in Table 8–1. Nucleic acids have two kinds of pentoses. The re- curring deoxyribonucleotide units of DNA contain 2H11032- deoxy-D-ribose, and the ribonucleotide units of RNA contain D-ribose. In nucleotides, both types of pentoses are in their H9252-furanose (closed five-membered ring) form. As Figure 8–3 shows, the pentose ring is not pla- nar but occurs in one of a variety of conformations gen- erally described as “puckered.” Figure 8–4 gives the structures and names of the four major deoxyribonucleotides (deoxyribonucleo- side 5H11032-monophosphates), the structural units of DNAs, and the four major ribonucleotides (ribonucleoside 5H11032- monophosphates), the structural units of RNAs. Specific long sequences of A, T, G, and C nucleotides in DNA are the repository of genetic information. Although nucleotides bearing the major purines and pyrimidines are most common, both DNA and RNA also Chapter 8 Nucleotides and Nucleic Acids274 FIGURE 8–1 Structure of nucleotides. (a) General structure showing the numbering convention for the pentose ring. This is a ribonu- cleotide. In deoxyribonucleotides the OOH group on the 2H11032 carbon (in red) is replaced with OH. (b) The parent compounds of the pyrim- idine and purine bases of nucleotides and nucleic acids, showing the numbering conventions. (b) (a) FIGURE 8–2 Major purine and pyrimidine bases of nucleic acids. Some of the common names of these bases reflect the circumstances of their discovery. Guanine, for example, was first isolated from guano (bird manure), and thymine was first isolated from thymus tissue. FIGURE 8–3 Conformations of ribose. (a) In solution, the straight- chain (aldehyde) and ring (H9252-furanose) forms of free ribose are in equi- librium. RNA contains only the ring form, H9252-D-ribofuranose. Deoxy- ribose undergoes a similar interconversion in solution, but in DNA exists solely as H9252-2H11032-deoxy-D-ribofuranose. (b) Ribofuranose rings in nucleotides can exist in four different puckered conformations. In all cases, four of the five atoms are in a single plane. The fifth atom (C-2H11032 or C-3H11032) is on either the same (endo) or the opposite (exo) side of the plane relative to the C-5H11032 atom. 8.1 Some Basics 275 CH 2 O H11002 O OH H P CH 3 O H11002 HN N H H H H O T, dT, dTMP Deoxythymidine Nucleotide: Deoxyadenylate (deoxyadenosine 5H11032-monophosphate) Deoxyguanylate (deoxyguanosine 5H11032-monophosphate) Deoxythymidylate (deoxythymidine 5H11032-monophosphate) Deoxycytidylate (deoxycytidine 5H11032-monophosphate) Symbols: A, dA, dAMP Nucleoside: Deoxyadenosine O G, dG, dGMP Deoxyguanosine O C, dC, dCMP Deoxycytidine (a) Deoxyribonucleotides O O CH 2 N O H11002 O OH H P NH 2 O H11002 N N N H H H H O O CH 2 O H11002 O OH H P HN H 2 N O H11002 N N N H H H H O O CH 2 O H11002 O OH H P NH 2 O H11002 N N H H H H O O O O CH 2 N O H11002 O OH H P NH 2 O H11002 N N N H H H O O CH 2 O H11002 O OH H P HN H 2 N O H11002 N N N H H H O O CH 2 O H11002 O OH H P O H11002 N N H H H O O (b) Ribonucleotides U, UMP C, CMP Uridine Nucleotide: Adenylate (adenosine 5H11032-monophosphate) Guanylate (guanosine 5H11032-monophosphate) Uridylate (uridine 5H11032-monophosphate) Cytidylate (cytidine 5H11032-monophosphate) Symbols: A, AMP Nucleoside: Adenosine G, GMP Guanosine Cytidine CH 2 O H11002 O OH H P NH 2 O H11002 N N H H H O O O OH OH OH OH H O O FIGURE 8–4 Deoxyribonucleotides and ribonucleotides of nucleic acids. All nucleotides are shown in their free form at pH 7.0. The nu- cleotide units of DNA (a) are usually symbolized as A, G, T, and C, sometimes as dA, dG, dT, and dC; those of RNA (b) as A, G, U, and C. In their free form the deoxyribonucleotides are commonly abbre- viated dAMP, dGMP, dTMP, and dCMP; the ribonucleotides, AMP, GMP, UMP, and CMP. For each nucleotide, the more common name is followed by the complete name in parentheses. All abbreviations assume that the phosphate group is at the 5H11032 position. The nucleoside portion of each molecule is shaded in light red. In this and the fol- lowing illustrations, the ring carbons are not shown. TABLE 8–1 Nucleotide and Nucleic Acid Nomenclature Base Nucleoside Nucleotide Nucleic acid Purines Adenine Adenosine Adenylate RNA Deoxyadenosine Deoxyadenylate DNA Guanine Guanosine Guanylate RNA Deoxyguanosine Deoxyguanylate DNA Pyrimidines Cytosine Cytidine Cytidylate RNA Deoxycytidine Deoxycytidylate DNA Thymine Thymidine or deoxythymidine Thymidylate or deoxythymidylate DNA Uracil Uridine Uridylate RNA Note: “Nucleoside” and “nucleotide” are generic terms that include both ribo- and deoxyribo- forms. Also, ribonucleosides and ribonucleotides are here designated simply as nucleosides and nucleotides (e.g., ribo- adenosine as adenosine), and deoxyribo- nucleosides and deoxyribonucleotides as deoxynucleosides and deoxynucleotides (e.g., deoxyriboadenosine as deoxyadeno- sine). Both forms of naming are accept- able, but the shortened names are more commonly used. Thymine is an exception; “ribothymidine” is used to describe its unusual occurrence in RNA. contain some minor bases (Fig. 8–5). In DNA the most common of these are methylated forms of the major bases; in some viral DNAs, certain bases may be hy- droxymethylated or glucosylated. Altered or unusual bases in DNA molecules often have roles in regulating or protecting the genetic information. Minor bases of many types are also found in RNAs, especially in tRNAs (see Fig. 26–24). The nomenclature for the minor bases can be con- fusing. Like the major bases, many have common names— hypoxanthine, for example, shown as its nucleoside ino- sine in Figure 8–5. When an atom in the purine or pyrimidine ring is substituted, the usual convention (used here) is simply to indicate the ring position of the sub- stituent by its number—for example, 5-methylcytosine, 7-methylguanine, and 5-hydroxymethylcytosine (shown as the nucleosides in Fig. 8–5). The element to which the substituent is attached (N, C, O) is not identified. The convention changes when the substituted atom is exocyclic (not within the ring structure), in which case the type of atom is identified and the ring position to which it is attached is denoted with a superscript. The amino nitrogen attached to C-6 of adenine is N 6 ; simi- larly, the carbonyl oxygen and amino nitrogen at C-6 and C-2 of guanine are O 6 and N 2 , respectively. Examples of this nomenclature are N 6 -methyladenosine and N 2 - methylguanosine (Fig. 8–5). Cells also contain nucleotides with phosphate groups in positions other than on the 5H11032 carbon (Fig. 8–6). Ribonucleoside 2H11541,3H11541-cyclic monophosphates are isolatable intermediates, and ribonucleoside 3H11541- monophosphates are end products of the hydrolysis of RNA by certain ribonucleases. Other variations are adenosine 3H11032,5H11032-cyclic monophosphate (cAMP) and guanosine 3H11032,5H11032-cyclic monophosphate (cGMP), consid- ered at the end of this chapter. Phosphodiester Bonds Link Successive Nucleotides in Nucleic Acids The successive nucleotides of both DNA and RNA are covalently linked through phosphate-group “bridges,” in which the 5H11032-phosphate group of one nucleotide unit is Chapter 8 Nucleotides and Nucleic Acids276 (a) (b) FIGURE 8–5 Some minor purine and pyrimidine bases, shown as the nucleosides. (a) Minor bases of DNA. 5-Methylcytidine occurs in the DNA of animals and higher plants, N 6 -methyladenosine in bacterial DNA, and 5-hydroxymethylcytidine in the DNA of bacteria infected with certain bacteriophages. (b) Some minor bases of tRNAs. Inosine contains the base hypoxanthine. Note that pseudouridine, like uridine, contains uracil; they are distinct in the point of attachment to the ribose—in uridine, uracil is attached through N-1, the usual attach- ment point for pyrimidines; in pseudouridine, through C-5. FIGURE 8–6 Some adenosine monophosphates. Adenosine 2H11032- monophosphate, 3H11032-monophosphate, and 2H11032,3H11032-cyclic monophosphate are formed by enzymatic and alkaline hydrolysis of RNA. joined to the 3H11032-hydroxyl group of the next nucleotide, creating a phosphodiester linkage (Fig. 8–7). Thus the covalent backbones of nucleic acids consist of al- ternating phosphate and pentose residues, and the ni- trogenous bases may be regarded as side groups joined to the backbone at regular intervals. The backbones of both DNA and RNA are hydrophilic. The hydroxyl groups of the sugar residues form hydrogen bonds with water. The phosphate groups, with a pK a near 0, are completely ionized and negatively charged at pH 7, and the negative charges are generally neutralized by ionic interactions with positive charges on proteins, metal ions, and polyamines. All the phosphodiester linkages have the same ori- entation along the chain (Fig. 8–7), giving each linear nucleic acid strand a specific polarity and distinct 5H11032 and 3H11032 ends. By definition, the 5H11541 end lacks a nucleotide at the 5H11032 position and the 3H11541 end lacks a nucleotide at the 3H11032 position. Other groups (most often one or more phos- phates) may be present on one or both ends. The covalent backbone of DNA and RNA is subject to slow, nonenzymatic hydrolysis of the phosphodiester bonds. In the test tube, RNA is hydrolyzed rapidly un- der alkaline conditions, but DNA is not; the 2H11032-hydroxyl groups in RNA (absent in DNA) are directly involved in the process. Cyclic 2H11032,3H11032-monophosphate nucleotides are the first products of the action of alkali on RNA and are rapidly hydrolyzed further to yield a mixture of 2H11032- and 3H11032-nucleoside monophosphates (Fig. 8–8). The nucleotide sequences of nucleic acids can be represented schematically, as illustrated on the follow- ing page by a segment of DNA with five nucleotide units. The phosphate groups are symbolized by PH22071, and each deoxyribose is symbolized by a vertical line, from C-1H11032 at the top to C-5H11032 at the bottom (but keep in mind that 8.1 Some Basics 277 O H11002 RNA CH 2 O H11002 O H P H OH H O 3H11032 5H11032 U H O CH 2 O H11002 O H P HH O O 3H11032 5H11032 G H O CH 2 O H11002 O H P HH O O 3H11032 5H11032 H O H O 5H11032 End O H11002 CH 2 O H11002 O H P H H H O 3H11032 5H11032 A H O CH 2 O H11002 O H P H H H O O 3H11032 5H11032 T H O CH 2 O H11002 O H P H H H O O 3H11032 5H11032 G H O H O 5H11032 End 3H11032 End3H11032 End C 5H11541 3H11541 DNA Phospho- diester linkage OH OH FIGURE 8–7 Phosphodiester linkages in the covalent backbone of DNA and RNA. The phosphodiester bonds (one of which is shaded in the DNA) link successive nucleotide units. The backbone of alternat- ing pentose and phosphate groups in both types of nucleic acid is highly polar. The 5H11032 end of the macromolecule lacks a nucleotide at the 5H11032 position, and the 3H11032 end lacks a nucleotide at the 3H11032 position. H P H H H O H11002 OH 2H11032,3H11032-Cyclic monophosphate derivative O O CH 2 O H P H H H O O Base 1 O O H11002 O H CH 2 O H P H H H O O Base 2 O H11002 O H OP H11002 O CH 2 H H H H O O Base 2 O H OP H11002 O OH H11001 Base 1 OP O H11002 O Mixture of 2H11032- and 3H11032-monophosphate derivatives CH 2 H11002 O O RNA Shortened RNA H 2 O O RNA Shortened RNA FIGURE 8–8 Hydrolysis of RNA under alkaline conditions. The 2H11032 hydroxyl acts as a nucleophile in an intramolecular displacement. The 2H11032,3H11032-cyclic monophosphate derivative is further hydrolyzed to a mixture of 2H11032- and 3H11032-monophosphates. DNA, which lacks 2H11032 hydroxyls, is stable under similar conditions. the sugar is always in its closed-ring H9252-furanose form in nucleic acids). The connecting lines between nucleotides (which pass through PH22071) are drawn diagonally from the middle (C-3H11032) of the deoxyribose of one nucleotide to the bottom (C-5H11032) of the next. By convention, the structure of a single strand of nu- cleic acid is always written with the 5H11032 end at the left and the 3H11032 end at the right—that is, in the 5H11032 n 3H11032 di- rection. Some simpler representations of this pentade- oxyribonucleotide are pA-C-G-T-A OH , pApCpGpTpA, and pACGTA. A short nucleic acid is referred to as an oligonu- cleotide. The definition of “short” is somewhat arbi- trary, but polymers containing 50 or fewer nucleotides are generally called oligonucleotides. A longer nucleic acid is called a polynucleotide. The Properties of Nucleotide Bases Affect the Three-Dimensional Structure of Nucleic Acids Free pyrimidines and purines are weakly basic com- pounds and are thus called bases. They have a variety of chemical properties that affect the structure, and ultimately the function, of nucleic acids. The purines and pyrimidines common in DNA and RNA are highly conjugated molecules (Fig. 8–2), a property with im- portant consequences for the structure, electron distri- bution, and light absorption of nucleic acids. Resonance among atoms in the ring gives most of the bonds par- tial double-bond character. One result is that pyrim- idines are planar molecules; purines are very nearly planar, with a slight pucker. Free pyrimidine and purine bases may exist in two or more tautomeric forms de- pending on the pH. Uracil, for example, occurs in lac- tam, lactim, and double lactim forms (Fig. 8–9). The structures shown in Figure 8–2 are the tautomers that predominate at pH 7.0. As a result of resonance, all nu- cleotide bases absorb UV light, and nucleic acids are characterized by a strong absorption at wavelengths near 260 nm (Fig. 8–10). The purine and pyrimidine bases are hydrophobic and relatively insoluble in water at the near-neutral pH of the cell. At acidic or alkaline pH the bases become charged and their solubility in water increases. Hy- drophobic stacking interactions in which two or more bases are positioned with the planes of their rings par- allel (like a stack of coins) are one of two important modes of interaction between bases in nucleic acids. The stacking also involves a combination of van der Waals and dipole-dipole interactions between the bases. Base stacking helps to minimize contact of the bases with wa- ter, and base-stacking interactions are very important in stabilizing the three-dimensional structure of nucleic acids, as described later. Chapter 8 Nucleotides and Nucleic Acids278 Uracil FIGURE 8–9 Tautomeric forms of uracil. The lactam form predomi- nates at pH 7.0; the other forms become more prominent as pH de- creases. The other free pyrimidines and the free purines also have tau- tomeric forms, but they are more rarely encountered. 14,000 12,000 10,000 8,000 6,000 4,000 2,000 280 Molar extinction coefficient, H9280 Wavelength (nm) 230 240 250 260 270 Molar extinction coefficient at 260 nm, H9280 260 (M H110021 cm H110021 ) AMP GMP UMP dTMP CMP 15,400 11,700 9,900 9,200 7,500 FIGURE 8–10 Absorption spectra of the common nucleotides. The spectra are shown as the variation in molar extinction coefficient with wavelength. The molar extinction coefficients at 260 nm and pH 7.0 (H9255 260 ) are listed in the table. The spectra of corresponding ribonucleotides and deoxyribonucleotides, as well as the nucleosides, are essentially identical. For mixtures of nucleotides, a wavelength of 260 nm (dashed vertical line) is used for absorption measurements. The most important functional groups of pyrim- idines and purines are ring nitrogens, carbonyl groups, and exocyclic amino groups. Hydrogen bonds involving the amino and carbonyl groups are the second impor- tant mode of interaction between bases in nucleic acid molecules. Hydrogen bonds between bases permit a complementary association of two (and occasionally three or four) strands of nucleic acid. The most impor- tant hydrogen-bonding patterns are those defined by James D. Watson and Francis Crick in 1953, in which A bonds specifically to T (or U) and G bonds to C (Fig. 8–11). These two types of base pairs predominate in double-stranded DNA and RNA, and the tautomers shown in Figure 8–2 are responsible for these patterns. It is this specific pairing of bases that permits the du- plication of genetic information, as we shall discuss later in this chapter. SUMMARY 8.1 Some Basics ■ A nucleotide consists of a nitrogenous base (purine or pyrimidine), a pentose sugar, and one or more phosphate groups. Nucleic acids are polymers of nucleotides, joined together by phosphodiester linkages between the 5H11032- hydroxyl group of one pentose and the 3H11032- hydroxyl group of the next. ■ There are two types of nucleic acid: RNA and DNA. The nucleotides in RNA contain ribose, and the common pyrimidine bases are uracil and cytosine. In DNA, the nucleotides contain 2H11032-deoxyribose, and the common pyrimidine bases are thymine and cytosine. The primary purines are adenine and guanine in both RNA and DNA. 8.2 Nucleic Acid Structure The discovery of the structure of DNA by Watson and Crick in 1953 was a momentous event in science, an event that gave rise to entirely new disciplines and in- fluenced the course of many established ones. Our pres- ent understanding of the storage and utilization of a cell’s genetic information is based on work made possi- ble by this discovery, and an outline of how genetic in- formation is processed by the cell is now a prerequisite for the discussion of any area of biochemistry. Here, we concern ourselves with DNA structure itself, the events 8.2 Nucleic Acid Structure 279 3H11032 C C C CG G G G A A A A A T T T T T 5H11032 5H11032 3H11032 10.8 ? N C O C N C H C C H C N C N C 11.1 ? 2.8 ? 3.0 ? H N C O CH 3 C O N H N C H C CH N C C N C H N C N O N H H H H 2.9 ? 3.0 ? 2.9 ? Adenine Thymine Guanine Cytosine N H C-1H11032 C-1H11032 C-1H11032 H H N C N C-1H11032 FIGURE 8–11 Hydrogen-bonding patterns in the base pairs defined by Watson and Crick. Here as elsewhere, hydrogen bonds are represented by three blue lines. James Watson Francis Crick that led to its discovery, and more recent refinements in our understanding. RNA structure is also introduced. As in the case of protein structure (Chapter 4), it is sometimes useful to describe nucleic acid structure in terms of hierarchical levels of complexity (primary, secondary, tertiary). The primary structure of a nucleic acid is its covalent structure and nucleotide sequence. Any regular, stable structure taken up by some or all of the nucleotides in a nucleic acid can be referred to as secondary structure. All structures considered in the re- mainder of this chapter fall under the heading of sec- ondary structure. The complex folding of large chro- mosomes within eukaryotic chromatin and bacterial nucleoids is generally considered tertiary structure; this is discussed in Chapter 24. DNA Stores Genetic Information The biochemical investigation of DNA began with Friedrich Miescher, who carried out the first systematic chemical studies of cell nuclei. In 1868 Miescher isolated a phosphorus-containing substance, which he called “nuclein,” from the nuclei of pus cells (leukocytes) ob- tained from discarded surgical bandages. He found nuclein to consist of an acidic portion, which we know today as DNA, and a basic portion, protein. Miescher later found a similar acidic substance in the heads of sperm cells from salmon. Although he partially purified nuclein and studied its properties, the covalent (pri- mary) structure of DNA (as shown in Fig. 8–7) was not known with certainty until the late 1940s. Miescher and many others suspected that nuclein (nucleic acid) was associated in some way with cell in- heritance, but the first direct evidence that DNA is the bearer of genetic information came in 1944 through a discovery made by Oswald T. Avery, Colin MacLeod, and Maclyn McCarty. These investigators found that DNA extracted from a virulent (disease-causing) strain of the bacterium Streptococcus pneumoniae, also known as pneumococcus, genetically transformed a nonvirulent strain of this organism into a virulent form (Fig. 8–12). Chapter 8 Nucleotides and Nucleic Acids280 (a) (b) (c) (d) (e) FIGURE 8–12 The Avery-MacLeod-McCarty experiment. (a) When injected into mice, the encapsulated strain of pneumococcus is lethal, (b) whereas the nonencapsulated strain, (c) like the heat-killed en- capsulated strain, is harmless. (d) Earlier research by the bacteriolo- gist Frederick Griffith had shown that adding heat-killed virulent bac- teria (harmless to mice) to a live nonvirulent strain permanently transformed the latter into lethal, virulent, encapsulated bacteria. (e) Avery and his colleagues extracted the DNA from heat-killed vir- ulent pneumococci, removing the protein as completely as possible, and added this DNA to nonvirulent bacteria. The DNA gained en- trance into the nonvirulent bacteria, which were permanently trans- formed into a virulent strain. Avery and his colleagues concluded that the DNA ex- tracted from the virulent strain carried the inheritable ge- netic message for virulence. Not everyone accepted these conclusions, because protein impurities present in the DNA could have been the carrier of the genetic informa- tion. This possibility was soon eliminated by the finding that treatment of the DNA with proteolytic enzymes did not destroy the transforming activity, but treatment with deoxyribonucleases (DNA-hydrolyzing enzymes) did. A second important experiment provided inde- pendent evidence that DNA carries genetic information. In 1952 Alfred D. Hershey and Martha Chase used ra- dioactive phosphorus ( 32 P) and radioactive sulfur ( 35 S) tracers to show that when the bacterial virus (bacterio- phage) T2 infects its host cell, Escherichia coli, it is the phosphorus-containing DNA of the viral particle, not the sulfur-containing protein of the viral coat, that en- ters the host cell and furnishes the genetic information for viral replication (Fig. 8–13). These important early experiments and many other lines of evidence have shown that DNA is the exclusive chromosomal compo- nent bearing the genetic information of living cells. DNA Molecules Have Distinctive Base Compositions A most important clue to the structure of DNA came from the work of Erwin Chargaff and his colleagues in the late 1940s. They found that the four nucleotide bases of DNA occur in different ratios in the DNAs of different organisms and that the amounts of certain bases are closely related. These data, collected from DNAs of a great many different species, led Chargaff to the following conclusions: 1. The base composition of DNA generally varies from one species to another. 2. DNA specimens isolated from different tissues of the same species have the same base composition. 3. The base composition of DNA in a given species does not change with an organism’s age, nutritional state, or changing environment. 4. In all cellular DNAs, regardless of the species, the number of adenosine residues is equal to the number of thymidine residues (that is, A H11005 T), and the number of guanosine residues is equal to the number of cytidine residues (G H11005 C). From these relationships it follows that the sum of the purine residues equals the sum of the pyrimidine residues; that is, A H11001 G H11005 T H11001 C. These quantitative relationships, sometimes called “Chargaff’s rules,” were confirmed by many subsequent researchers. They were a key to establishing the three- dimensional structure of DNA and yielded clues to how genetic information is encoded in DNA and passed from one generation to the next. 32 P experiment 35 S experiment Radioactive DNA Nonradioactive coat Nonradioactive DNA Radioactive coat Injection Blender treatment shears off viral heads Separation by centrifugation Radioactive Not radioactive Phage Radioactive Not radioactive Bacterial cell FIGURE 8–13 The Hershey-Chase experiment. Two batches of iso- topically labeled bacteriophage T2 particles were prepared. One was labeled with 32 P in the phosphate groups of the DNA, the other with 35 S in the sulfur-containing amino acids of the protein coats (capsids). (Note that DNA contains no sulfur and viral protein contains no phos- phorus.) The two batches of labeled phage were then allowed to in- fect separate suspensions of unlabeled bacteria. Each suspension of phage-infected cells was agitated in a blender to shear the viral cap- sids from the bacteria. The bacteria and empty viral coats (called “ghosts”) were then separated by centrifugation. The cells infected with the 32 P-labeled phage were found to contain 32 P, indicating that the labeled viral DNA had entered the cells; the viral ghosts contained no radioactivity. The cells infected with 35 S-labeled phage were found to have no radioactivity after blender treatment, but the viral ghosts con- tained 35 S. Progeny virus particles (not shown) were produced in both batches of bacteria some time after the viral coats were removed, in- dicating that the genetic message for their replication had been in- troduced by viral DNA, not by viral protein. DNA Is a Double Helix To shed more light on the structure of DNA, Rosalind Franklin and Maurice Wilkins used the powerful method of x-ray diffraction (see Box 4–4) to analyze DNA fibers. They showed in the early 1950s that DNA produces a characteristic x-ray diffraction pattern (Fig. 8–14). From this pattern it was deduced that DNA molecules are helical with two periodicities along their long axis, a primary one of 3.4 ? and a secondary one of 34 ?. The problem then was to formulate a three-dimensional model of the DNA molecule that could account not only for the x-ray diffraction data but also for the spe- cific A H11005 T and G H11005 C base equivalences discovered by Chargaff and for the other chemical properties of DNA. In 1953 Watson and Crick postulated a three- dimensional model of DNA structure that accounted for all the available data. It consists of two helical DNA chains wound around the same axis to form a right- handed double helix (see Box 4–1 for an explanation of the right- or left-handed sense of a helical structure). The hydrophilic backbones of alternating deoxyribose and phosphate groups are on the outside of the double helix, facing the surrounding water. The furanose ring of each deoxyribose is in the C-2H11032 endo conformation. The purine and pyrimidine bases of both strands are stacked inside the double helix, with their hydrophobic and nearly planar ring structures very close together and perpendicular to the long axis. The offset pairing of the two strands creates a major groove and minor groove on the surface of the duplex (Fig. 8–15). Each nucleotide base of one strand is paired in the same plane with a base of the other strand. Watson and Crick found that the hydrogen-bonded base pairs illustrated in Fig- ure 8–11, G with C and A with T, are those that fit best within the structure, providing a rationale for Chargaff’s rule that in any DNA, G H11005 C and A H11005 T. It is important to note that three hydrogen bonds can form between G and C, symbolized GqC, but only two can form between A and T, symbolized AUT. This is one reason for the finding that separation of paired DNA strands is more difficult the higher the ratio of GqC to AUT base pairs. Other pairings of bases tend (to varying degrees) to destabilize the double-helical structure. When Watson and Crick constructed their model, they had to decide at the outset whether the strands of DNA should be parallel or antiparallel—whether their 5H11032,3H11032-phosphodiester bonds should run in the same or opposite directions. An antiparallel orientation pro- duced the most convincing model, and later work with DNA polymerases (Chapter 25) provided experimental evidence that the strands are indeed antiparallel, a find- ing ultimately confirmed by x-ray analysis. To account for the periodicities observed in the x- ray diffraction patterns of DNA fibers, Watson and Crick manipulated molecular models to arrive at a structure Chapter 8 Nucleotides and Nucleic Acids282 FIGURE 8–14 X-ray diffraction pattern of DNA. The spots forming a cross in the center denote a helical structure. The heavy bands at the left and right arise from the recurring bases. FIGURE 8–15 Watson-Crick model for the structure of DNA. The original model proposed by Watson and Crick had 10 base pairs, or 34 ? (3.4 nm), per turn of the helix; subsequent measurements revealed 10.5 base pairs, or 36 ? (3.6 nm), per turn. (a) Schematic represen- tation, showing dimensions of the helix. (b) Stick representation show- ing the backbone and stacking of the bases. (c) Space-filling model. Rosalind Franklin, 1920–1958 Maurice Wilkins in which the vertically stacked bases inside the double helix would be 3.4 ? apart; the secondary repeat dis- tance of about 34 ? was accounted for by the presence of 10 base pairs in each complete turn of the double helix. In aqueous solution the structure differs slightly from that in fibers, having 10.5 base pairs per helical turn (Fig. 8–15). As Figure 8–16 shows, the two antiparallel polynu- cleotide chains of double-helical DNA are not identical in either base sequence or composition. Instead they are complementary to each other. Wherever adenine oc- curs in one chain, thymine is found in the other; simi- larly, wherever guanine occurs in one chain, cytosine is found in the other. The DNA double helix, or duplex, is held together by two forces, as described earlier: hydrogen bonding between complementary base pairs (Fig. 8–11) and base-stacking interactions. The complementarity be- tween the DNA strands is attributable to the hydrogen bonding between base pairs. The base-stacking interac- tions, which are largely nonspecific with respect to the identity of the stacked bases, make the major contribu- tion to the stability of the double helix. The important features of the double-helical model of DNA structure are supported by much chemical and biological evidence. Moreover, the model immediately suggested a mechanism for the transmission of genetic information. The essential feature of the model is the complementarity of the two DNA strands. As Watson and Crick were able to see, well before confirmatory data be- came available, this structure could logically be replicated by (1) separating the two strands and (2) synthesizing a complementary strand for each. Because nucleotides in each new strand are joined in a sequence specified by the base-pairing rules stated above, each preexisting strand functions as a template to guide the synthesis of one complementary strand (Fig. 8–17). These expecta- tions were experimentally confirmed, inaugurating a rev- olution in our understanding of biological inheritance. DNA Can Occur in Different Three-Dimensional Forms DNA is a remarkably flexible molecule. Considerable ro- tation is possible around a number of bonds in the sugar–phosphate (phosphodeoxyribose) backbone, and thermal fluctuation can produce bending, stretching, and unpairing (melting) of the strands. Many significant de- viations from the Watson-Crick DNA structure are found in cellular DNA, some or all of which may play impor- tant roles in DNA metabolism. These structural varia- tions generally do not affect the key properties of DNA defined by Watson and Crick: strand complementarity, 8.2 Nucleic Acid Structure 283 FIGURE 8–16 Complementarity of strands in the DNA double helix. The complementary antiparallel strands of DNA follow the pairing rules proposed by Watson and Crick. The base-paired antiparallel strands differ in base composition: the left strand has the composition A 3 T 2 G 1 C 3 ; the right, A 2 T 3 G 3 C 1 . They also differ in sequence when each chain is read in the 5H11032 n 3H11032 direction. Note the base equiva- lences: A H11005 T and G H11005 C in the duplex. FIGURE 8–17 Replication of DNA as suggested by Watson and Crick. The preexisting or “parent” strands become separated, and each is the template for biosynthesis of a complementary “daughter” strand (in red). antiparallel strands, and the requirement for APT and GqC base pairs. Structural variation in DNA reflects three things: the different possible conformations of the deoxyribose, rotation about the contiguous bonds that make up the phosphodeoxyribose backbone (Fig. 8–18a), and free rotation about the C-1H11032–N-glycosyl bond (Fig. 8–18b). Because of steric constraints, purines in purine nu- cleotides are restricted to two stable conformations with respect to deoxyribose, called syn and anti (Fig. 8–18b). Pyrimidines are generally restricted to the anti confor- mation because of steric interference between the sugar and the carbonyl oxygen at C-2 of the pyrimidine. The Watson-Crick structure is also referred to as B- form DNA, or B-DNA. The B form is the most stable structure for a random-sequence DNA molecule under physiological conditions and is therefore the standard point of reference in any study of the properties of DNA. Two structural variants that have been well character- ized in crystal structures are the A and Z forms. These three DNA conformations are shown in Figure 8–19, with a summary of their properties. The A form is fa- vored in many solutions that are relatively devoid of wa- ter. The DNA is still arranged in a right-handed double helix, but the helix is wider and the number of base pairs per helical turn is 11, rather than 10.5 as in B-DNA. The Chapter 8 Nucleotides and Nucleic Acids284 FIGURE 8–18 Structural variation in DNA. (a) The conformation of a nucleotide in DNA is affected by rotation about seven different bonds. Six of the bonds rotate freely. The limited rotation about bond 4 gives rise to ring pucker, in which one of the atoms in the five-membered furanose ring is out of the plane described by the other four. This conformation is endo or exo, depending on whether the atom is displaced to the same side of the plane as C-5H11032 or to the opposite side (see Fig. 8–3b). (b) For purine bases in nucleotides, only two conformations with respect to the attached ribose units are sterically permitted, anti or syn. Pyrimidines generally occur in the anti conformation. FIGURE 8–19 Comparison of A, B, and Z forms of DNA. Each struc- ture shown here has 36 base pairs. The bases are shown in gray, the phosphate atoms in yellow, and the riboses and phosphate oxygens in blue. Blue is the color used to represent DNA strands in later chap- ters. The table summarizes some properties of the three forms of DNA. A form B form Z form Helical sense Right handed Right handed Left handed Diameter H1101126 ? H1101120 ? H1101118 ? Base pairs per helical turn 11 10.5 12 Helix rise per base pair 2.6 ? 3.4 ? 3.7 ? Base tilt normal to the helix axis 20° 6° 7° Sugar pucker conformation C-3H11032 endo C-2H11032 endo C-2H11032 endo for pyrimidines; C-3H11032 endo for purines Glycosyl bond conformation Anti Anti Anti for pyrimidines; syn for purines plane of the base pairs in A-DNA is tilted about 20H11034 with respect to the helix axis. These structural changes deepen the major groove while making the minor groove shallower. The reagents used to promote crystallization of DNA tend to dehydrate it, and thus most short DNA molecules tend to crystallize in the A form. Z-form DNA is a more radical departure from the B structure; the most obvious distinction is the left- handed helical rotation. There are 12 base pairs per hel- ical turn, and the structure appears more slender and elongated. The DNA backbone takes on a zigzag ap- pearance. Certain nucleotide sequences fold into left- handed Z helices much more readily than others. Promi- nent examples are sequences in which pyrimidines alternate with purines, especially alternating C and G or 5-methyl-C and G residues. To form the left-handed helix in Z-DNA, the purine residues flip to the syn conformation, alternating with pyrimidines in the anti conformation. The major groove is barely apparent in Z-DNA, and the minor groove is narrow and deep. Whether A-DNA occurs in cells is uncertain, but there is evidence for some short stretches (tracts) of Z-DNA in both prokaryotes and eukaryotes. These Z-DNA tracts may play a role (as yet undefined) in regulating the ex- pression of some genes or in genetic recombination. Certain DNA Sequences Adopt Unusual Structures A number of other sequence-dependent structural vari- ations have been detected within larger chromosomes that may affect the function and metabolism of the DNA segments in their immediate vicinity. For example, bends occur in the DNA helix wherever four or more adenosine residues appear sequentially in one strand. Six adenosines in a row produce a bend of about 18H11034. The bending observed with this and other sequences may be important in the binding of some proteins to DNA. A rather common type of DNA sequence is a palin- drome. A palindrome is a word, phrase, or sentence that is spelled identically read either forward or back- ward; two examples are ROTATOR and NURSES RUN. The term is applied to regions of DNA with inverted repeats of base sequence having twofold symmetry over two strands of DNA (Fig. 8–20). Such sequences are self-complementary within each strand and there- fore have the potential to form hairpin or cruciform (cross-shaped) structures (Fig. 8–21). When the in- verted repeat occurs within each individual strand of the DNA, the sequence is called a mirror repeat. Mirror repeats do not have complementary sequences within the same strand and cannot form hairpin or cru- ciform structures. Sequences of these types are found 8.2 Nucleic Acid Structure 285 FIGURE 8–20 Palindromes and mirror repeats. Palindromes are se- quences of double-stranded nucleic acids with twofold symmetry. In order to superimpose one repeat (shaded sequence) on the other, it must be rotated 180H11034 about the horizontal axis then 180H11034 about the vertical axis, as shown by the colored arrows. A mirror repeat, on the other hand, has a symmetric sequence within each strand. Superim- posing one repeat on the other requires only a single 180H11034 rotation about the vertical axis. FIGURE 8–21 Hairpins and cruciforms. Palindromic DNA (or RNA) sequences can form alternative structures with intrastrand base pair- ing. (a) When only a single DNA (or RNA) strand is involved, the structure is called a hairpin. (b) When both strands of a duplex DNA are involved, it is called a cruciform. Blue shading highlights asym- metric sequences that can pair with the complementary sequence ei- ther in the same strand or in the complementary strand. in virtually every large DNA molecule and can encom- pass a few base pairs or thousands. The extent to which palindromes occur as cruciforms in cells is not known, although some cruciform structures have been demon- strated in vivo in E.coli. Self-complementary sequences cause isolated single strands of DNA (or RNA) in solu- tion to fold into complex structures containing multiple hairpins. Several unusual DNA structures involve three or even four DNA strands. These structural variations merit investigation because there is a tendency for many of them to appear at sites where important events in DNA metabolism (replication, recombination, transcription) are initiated or regulated. Nucleotides participating in a Watson-Crick base pair (Fig. 8–11) can form a number of additional hydrogen bonds, particularly with func- tional groups arrayed in the major groove. For example, a cytidine residue (if protonated) can pair with the guanosine residue of a GqC nucleotide pair, and a thymidine can pair with the adenosine of an AUT pair (Fig. 8–22). The N-7, O 6 , and N 6 of purines, the atoms that participate in the hydrogen bonding of triplex DNA, are often referred to as Hoogsteen positions, and the non-Watson-Crick pairing is called Hoogsteen pairing, after Karst Hoogsteen, who in 1963 first recognized the potential for these unusual pairings. Hoogsteen pairing allows the formation of triplex DNAs. The triplexes shown in Figure 8–22 (a, b) are most stable at low pH Chapter 8 Nucleotides and Nucleic Acids286 CH 3 CH 3 O N O H N N H H N H N N C-1H11032 1H11032-C C-1H11032 N NN O O TAT (a) N N H11001 O H N O H H H N H N N C-1H11032 1H11032-C C-1H11032 N H H N NN O H N CGC H11001 H Guanosine tetraplex (c) H H N N O C-1H11032 N N N H H H N N N N 1H11032-C N O C-1H11032 N N N N O NH H H O C-1H11032 N H H N NN N H Parallel Antiparallel (e) FIGURE 8–22 DNA structures containing three or four DNA strands. (a) Base-pairing patterns in one well-characterized form of triplex DNA. The Hoogsteen pair in each case is shown in red. (b) Triple- helical DNA containing two pyrimidine strands (poly(T)) and one purine strand (poly(A)) (derived from PDB ID 1BCE). The dark blue and light blue strands are antiparallel and paired by normal Watson- Crick base-pairing patterns. The third (all-pyrimidine) strand (purple) is parallel to the purine strand and paired through non-Watson-Crick hydrogen bonds. The triplex is viewed end-on, with five triplets shown. Only the triplet closest to the viewer is colored. (c) Base-pairing pat- tern in the guanosine tetraplex structure. (d) Two successive tetraplets from a G tetraplex structure (derived from PDB ID 1QDG), viewed end-on with the one closest to the viewer in color. (e) Possible vari- ants in the orientation of strands in a G tetraplex. because the CqG H11554 C H11001 triplet requires a protonated cy- tosine. In the triplex, the pK a of this cytosine is H110227.5, altered from its normal value of 4.2. The triplexes also form most readily within long sequences containing only pyrimidines or only purines in a given strand. Some triplex DNAs contain two pyrimidine strands and one purine strand; others contain two purine strands and one pyrimidine strand. Four DNA strands can also pair to form a tetraplex (quadruplex), but this occurs readily only for DNA se- quences with a very high proportion of guanosine residues (Fig. 8–22c, d). The guanosine tetraplex, or G tetraplex, is quite stable over a wide range of condi- tions. The orientation of strands in the tetraplex can vary as shown in Figure 8–22e. A particularly exotic DNA structure, known as H-DNA, is found in polypyrimidine or polypurine tracts that also incorporate a mirror repeat. A simple example is a long stretch of alternating T and C residues (Fig. 8–23). The H-DNA structure features the triple-stranded form illustrated in Figure 8–22 (a, b). Two of the three strands in the H-DNA triple helix contain pyrimidines and the third contains purines. In the DNA of living cells, sites recognized by many sequence-specific DNA-binding proteins (Chapter 28) are arranged as palindromes, and polypyrimidine or polypurine sequences that can form triple helices or even H-DNA are found within regions involved in the regulation of expression of some eukaryotic genes. In principle, synthetic DNA strands designed to pair with these sequences to form triplex DNA could disrupt gene expression. This approach to controlling cellular me- tabolism is of growing commercial interest for its po- tential application in medicine and agriculture. Messenger RNAs Code for Polypeptide Chains We now turn our attention briefly from DNA structure to the expression of the genetic information that it con- tains. RNA, the second major form of nucleic acid in cells, has many functions. In gene expression, RNA acts as an intermediary by using the information encoded in DNA to specify the amino acid sequence of a functional protein. Given that the DNA of eukaryotes is largely con- fined to the nucleus whereas protein synthesis occurs on ribosomes in the cytoplasm, some molecule other than DNA must carry the genetic message from the nu- cleus to the cytoplasm. As early as the 1950s, RNA was considered the logical candidate: RNA is found in both the nucleus and the cytoplasm, and an increase in pro- tein synthesis is accompanied by an increase in the amount of cytoplasmic RNA and an increase in its rate of turnover. These and other observations led several researchers to suggest that RNA carries genetic infor- mation from DNA to the protein biosynthetic machin- ery of the ribosome. In 1961 Fran?ois Jacob and Jacques Monod presented a unified (and essentially correct) pic- ture of many aspects of this process. They proposed the name “messenger RNA” (mRNA) for that portion of the total cellular RNA carrying the genetic information from DNA to the ribosomes, where the messengers provide the templates that specify amino acid sequences in polypeptide chains. Although mRNAs from different genes can vary greatly in length, the mRNAs from a par- ticular gene generally have a defined size. The process of forming mRNA on a DNA template is known as transcription. In prokaryotes, a single mRNA molecule may code for one or several polypeptide chains. If it carries the code for only one polypeptide, the mRNA is monocistronic; 8.2 Nucleic Acid Structure 287 FIGURE 8–23 H-DNA. (a) A sequence of alternating T and C residues can be considered a mirror repeat centered about a central T or C. (b) These sequences form an unusual structure in which the strands in one half of the mirror repeat are separated and the pyrimidine- containing strand (alternating T and C residues) folds back on the other half of the repeat to form a triple helix. The purine strand (alternating A and G residues) is left unpaired. This structure produces a sharp bend in the DNA. if it codes for two or more different polypeptides, the mRNA is polycistronic. In eukaryotes, most mRNAs are monocistronic. (For the purposes of this discussion, “cistron” refers to a gene. The term itself has historical roots in the science of genetics, and its formal genetic definition is beyond the scope of this text.) The mini- mum length of an mRNA is set by the length of the polypeptide chain for which it codes. For example, a polypeptide chain of 100 amino acid residues requires an RNA coding sequence of at least 300 nucleotides, be- cause each amino acid is coded by a nucleotide triplet (this and other details of protein synthesis are discussed in Chapter 27). However, mRNAs transcribed from DNA are always somewhat longer than the length needed sim- ply to code for a polypeptide sequence (or sequences). The additional, noncoding RNA includes sequences that regulate protein synthesis. Figure 8–24 summarizes the general structure of prokaryotic mRNAs. Many RNAs Have More Complex Three-Dimensional Structures Messenger RNA is only one of several classes of cellu- lar RNA. Transfer RNAs serve as adapter molecules in protein synthesis; covalently linked to an amino acid at one end, they pair with the mRNA in such a way that amino acids are joined to a growing polypeptide in the correct sequence. Ribosomal RNAs are components of ribosomes. There is also a wide variety of special-func- tion RNAs, including some (called ribozymes) that have enzymatic activity. All the RNAs are considered in de- tail in Chapter 26. The diverse and often complex func- tions of these RNAs reflect a diversity of structure much richer than that observed in DNA molecules. The product of transcription of DNA is always single-stranded RNA. The single strand tends to assume a right-handed helical conformation dominated by base- stacking interactions (Fig. 8–25), which are stronger be- tween two purines than between a purine and pyrimi- dine or between two pyrimidines. The purine-purine interaction is so strong that a pyrimidine separating two purines is often displaced from the stacking pattern so that the purines can interact. Any self-complementary sequences in the molecule produce more complex struc- tures. RNA can base-pair with complementary regions of either RNA or DNA. Base pairing matches the pat- tern for DNA: G pairs with C and A pairs with U (or with the occasional T residue in some RNAs). One difference is that base pairing between G and U residues—unusual in DNA—is fairly common in RNA (see Fig. 8–27). The paired strands in RNA or RNA-DNA duplexes are an- tiparallel, as in DNA. RNA has no simple, regular secondary structure that serves as a reference point, as does the double he- lix for DNA. The three-dimensional structures of many RNAs, like those of proteins, are complex and unique. Weak interactions, especially base-stacking interactions, play a major role in stabilizing RNA structures, just as they do in DNA. Where complementary sequences are present, the predominant double-stranded structure is an A-form right-handed double helix. Z-form helices have been made in the laboratory (under very high-salt or high-temperature conditions). The B form of RNA has not been observed. Breaks in the regular A-form he- lix caused by mismatched or unmatched bases in one or both strands are common and result in bulges or in- ternal loops (Fig. 8–26). Hairpin loops form between nearby self-complementary sequences. The potential for base-paired helical structures in many RNAs is exten- sive (Fig. 8–27), and the resulting hairpins are the most common type of secondary structure in RNA. Specific Chapter 8 Nucleotides and Nucleic Acids288 FIGURE 8–24 Prokaryotic mRNA. Schematic diagrams show (a) monocistronic and (b) polycistronic mRNAs of prokaryotes. Red seg- ments represent RNA coding for a gene product; gray segments rep- resent noncoding RNA. In the polycistronic transcript, noncoding RNA separates the three genes. FIGURE 8–25 Typical right-handed stacking pattern of single- stranded RNA. The bases are shown in gray, the phosphate atoms in yellow, and the riboses and phosphate oxygens in green. Green is used to represent RNA strands in succeeding chapters, just as blue is used for DNA. short base sequences (such as UUCG) are often found at the ends of RNA hairpins and are known to form par- ticularly tight and stable loops. Such sequences may act as starting points for the folding of an RNA molecule into its precise three-dimensional structure. Important additional structural contributions are made by hydro- gen bonds that are not part of standard Watson-Crick base pairs. For example, the 2H11032-hydroxyl group of ribose can hydrogen-bond with other groups. Some of these properties are evident in the structure of the phenyl- alanine transfer RNA of yeast—the tRNA responsible for inserting Phe residues into polypeptides—and in two RNA enzymes, or ribozymes, whose functions, like those of protein enzymes, depend on their three-dimensional structures (Fig. 8–28). The analysis of RNA structure and the relationship between structure and function is an emerging field of inquiry that has many of the same complexities as the analysis of protein structure. The importance of under- standing RNA structure grows as we become increas- ingly aware of the large number of functional roles for RNA molecules. 8.2 Nucleic Acid Structure 289 G U C A C C A G U G C A A C A G A G A G C A A C A G U G A 180 A G G C G C 120 A C G G G C G C C C A 240 A U G G C C A G C G C C G A U C C C G C C G G G G A U C G G U G G C A 160 A G G 140 A A A G C C C G G C G G U G G A 220 A A U G G C G G C U G U G C C G A C G G U A 200 A A C C G G 100 A A G G C A G G 80 C C C U A A G A A U G G G C C C A C G A U A A A G U C C G G G C A G G C U G C U U G U A G A U G A A G G A G G A G G C U U C G G G C A A CA U A C U G A C A G A C U G U C G G G A C G G C A G G C G C U U C G U G G G G C C CC G O ON H O NH 2 N N N N O N H O Guanine Uracil C C G G A A A U A G G C C C A A G G U U C A G U G C U A A C G U G C G C C 280 A260 G U G G G U A 300 C A A G C G U G C C G G G U A G U U G A C 330 U A C C AG U CG A A GG U CA G U U U C GAC C U 377 360 1 C U A U U C G G C C C A A G A C A G C A C 20 60 G C G U A U U GG G C U U 40 A C FIGURE 8–26 Secondary structure of RNAs. (a) Bulge, internal loop, and hairpin loop. (b) The paired regions generally have an A-form right-handed helix, as shown for a hairpin. FIGURE 8–27 Base-paired helical structures in an RNA. Shown here is the possible secondary structure of the M1 RNA component of the enzyme RNase P of E. coli, with many hairpins. RNase P, which also contains a protein component (not shown), functions in the processing of transfer RNAs (see Fig. 26–23). The two brackets indicate additional complementary sequences that may be paired in the three-dimensional structure. The blue dots indicate non-Watson- Crick GUU base pairs (boxed inset). Note that GUU base pairs are allowed only when presynthesized strands of RNA fold up or anneal with each other. There are no RNA polymerases (the enzymes that synthesize RNAs on a DNA template) that insert a U opposite a template G, or vice versa, during RNA synthesis. SUMMARY 8.2 Nucleic Acid Structure ■ Many lines of evidence show that DNA bears genetic information. In particular, the Avery- MacLeod-McCarty experiment showed that DNA isolated from one bacterial strain can enter and transform the cells of another strain, endowing it with some of the inheritable characteristics of the donor. The Hershey-Chase experiment showed that the DNA of a bacterial virus, but not its protein coat, carries the genetic message for replication of the virus in a host cell. ■ Putting together much published data, Watson and Crick postulated that native DNA consists of two antiparallel chains in a right-handed double-helical arrangement. Complementary base pairs, AUT and GqC, are formed by hydrogen bonding within the helix. The base Chapter 8 Nucleotides and Nucleic Acids290 FIGURE 8–28 Three-dimensional structure in RNA. (a) Three- dimensional structure of phenylalanine tRNA of yeast (PDB ID 1TRA). Some unusual base-pairing patterns found in this tRNA are shown. Note also the involvement of the oxygen of a ribose phosphodiester bond in one hydrogen-bonding arrangement, and a ribose 2H11032-hydroxyl group in another (both in red). (b) A hammerhead ribozyme (so named because the secondary structure at the active site looks like the head of a hammer), derived from certain plant viruses (derived from PDB ID 1MME). Ribozymes, or RNA enzymes, catalyze a variety of reac- tions, primarily in RNA metabolism and protein synthesis. The com- plex three-dimensional structures of these RNAs reflect the complexity inherent in catalysis, as described for protein enzymes in Chapter 6. (c) A segment of mRNA known as an intron, from the ciliated proto- zoan Tetrahymena thermophila (derived from PDB ID 1GRZ). This intron (a ribozyme) catalyzes its own excision from between exons in an mRNA strand (discussed in Chapter 26). pairs are stacked perpendicular to the long axis of the double helix, 3.4 ? apart, with 10.5 base pairs per turn. ■ DNA can exist in several structural forms. Two variations of the Watson-Crick form, or B-DNA, are A- and Z-DNA. Some sequence-dependent structural variations cause bends in the DNA molecule. DNA strands with appropriate se- quences can form hairpin/cruciform structures or triplex or tetraplex DNA. ■ Messenger RNA transfers genetic information from DNA to ribosomes for protein synthesis. Transfer RNA and ribosomal RNA are also involved in protein synthesis. RNA can be structurally complex; single RNA strands can be folded into hairpins, double-stranded re- gions, or complex loops. 8.3 Nucleic Acid Chemistry To understand how nucleic acids function, we must un- derstand their chemical properties as well as their struc- tures. The role of DNA as a repository of genetic infor- mation depends in part on its inherent stability. The chemical transformations that do occur are generally very slow in the absence of an enzyme catalyst. The long-term storage of information without alteration is so important to a cell, however, that even very slow reactions that alter DNA structure can be physiologically significant. Processes such as carcinogenesis and aging may be intimately linked to slowly accumulating, irreversible al- terations of DNA. Other, nondestructive alterations also occur and are essential to function, such as the strand separation that must precede DNA replication or tran- scription. In addition to providing insights into physio- logical processes, our understanding of nucleic acid chemistry has given us a powerful array of technologies that have applications in molecular biology, medicine, and forensic science. We now examine the chemical proper- ties of DNA and some of these technologies. Double-Helical DNA and RNA Can Be Denatured Solutions of carefully isolated, native DNA are highly viscous at pH 7.0 and room temperature (25 H11034C). When such a solution is subjected to extremes of pH or to tem- peratures above 80 H11034C, its viscosity decreases sharply, indicating that the DNA has undergone a physical change. Just as heat and extremes of pH denature glob- ular proteins, they also cause denaturation, or melting, of double-helical DNA. Disruption of the hydrogen bonds between paired bases and of base stacking causes unwinding of the double helix to form two single strands, completely separate from each other along the entire length or part of the length (partial denaturation) of the molecule. No covalent bonds in the DNA are broken (Fig. 8–29). Renaturation of a DNA molecule is a rapid one-step process, as long as a double-helical segment of a dozen or more residues still unites the two strands. When the temperature or pH is returned to the range in which most organisms live, the unwound segments of the two strands spontaneously rewind, or anneal, to yield the intact duplex (Fig. 8–29). However, if the two strands are completely separated, renaturation occurs in two steps. In the first, relatively slow step, the two strands “find” each other by random collisions and form a short segment of complementary double helix. The second step is much faster: the remaining unpaired bases suc- cessively come into register as base pairs, and the two strands “zipper” themselves together to form the dou- ble helix. The close interaction between stacked bases in a nucleic acid has the effect of decreasing its absorption of UV light relative to that of a solution with the same concentration of free nucleotides, and the absorption is decreased further when two complementary nucleic acids strands are paired. This is called the hypochromic effect. Denaturation of a double-stranded nucleic acid produces the opposite result: an increase in absorption 8.3 Nucleic Acid Chemistry 291 FIGURE 8–29 Reversible denaturation and annealing (renaturation) of DNA. called the hyperchromic effect. The transition from double-stranded DNA to the single-stranded, denatured form can thus be detected by monitoring the absorption of UV light. Viral or bacterial DNA molecules in solution dena- ture when they are heated slowly (Fig. 8–30). Each species of DNA has a characteristic denaturation tem- perature, or melting point (t m ): the higher its content of GqC base pairs, the higher the melting point of the DNA. This is because GqC base pairs, with three hy- drogen bonds, require more heat energy to dissociate than AUT base pairs. Careful determination of the melt- ing point of a DNA specimen, under fixed conditions of pH and ionic strength, can yield an estimate of its base composition. If denaturation conditions are carefully controlled, regions that are rich in AUT base pairs will specifically denature while most of the DNA remains double-stranded. Such denatured regions (called bub- bles) can be visualized with electron microscopy (Fig. 8–31). Strand separation of DNA must occur in vivo dur- ing processes such as DNA replication and transcrip- tion. As we shall see, the DNA sites where these processes are initiated are often rich in AUT base pairs. Duplexes of two RNA strands or of one RNA strand and one DNA strand (RNA-DNA hybrids) can also be denatured. Notably, RNA duplexes are more stable than DNA duplexes. At neutral pH, denaturation of a double- helical RNA often requires temperatures 20 H11034C or more higher than those required for denaturation of a DNA molecule with a comparable sequence. The stability of an RNA-DNA hybrid is generally intermediate between that of RNA and that of DNA. The physical basis for these differences in thermal stability is not known. Nucleic Acids from Different Species Can Form Hybrids The ability of two complementary DNA strands to pair with one another can be used to detect similar DNA se- quences in two different species or within the genome of a single species. If duplex DNAs isolated from human cells and from mouse cells are completely denatured by heating, then mixed and kept at 65 H11034C for many hours, much of the DNA will anneal. Most of the mouse DNA strands anneal with complementary mouse DNA strands to form mouse duplex DNA; similarly, most human DNA strands anneal with complementary human DNA strands. However, some strands of the mouse DNA will associate with human DNA strands to yield hybrid Chapter 8 Nucleotides and Nucleic Acids292 100 G H11001 C (% of total nucleotides) 80 0 70 80 90 t m (°C) 11060 100 60 40 20 100 Denaturation (%) 50 0 75 80 85 Temperature (°C) t m t m FIGURE 8–30 Heat denaturation of DNA. (a) The denaturation, or melting, curves of two DNA specimens. The temperature at the mid- point of the transition (t m ) is the melting point; it depends on pH and ionic strength and on the size and base composition of the DNA. (b) Relationship between t m and the GqC content of a DNA. (a) (b) FIGURE 8–31 Partially denatured DNA. This DNA was partially de- natured, then fixed to prevent renaturation during sample preparation. The shadowing method used to visualize the DNA in this electron mi- crograph increases its diameter approximately fivefold and obliterates most details of the helix. However, length measurements can be ob- tained, and single-stranded regions are readily distinguishable from double-stranded regions. The arrows point to some single-stranded bubbles where denaturation has occurred. The regions that denature are highly reproducible and are rich in AUT base pairs. duplexes, in which segments of a mouse DNA strand form base-paired regions with segments of a human DNA strand (Fig. 8–32). This reflects a common evolutionary heritage; different organisms generally have some pro- teins and RNAs with similar functions and, often, simi- lar structures. In many cases, the DNAs encoding these proteins and RNAs have similar sequences. The closer the evolutionary relationship between two species, the more extensively their DNAs will hybridize. For exam- ple, human DNA hybridizes much more extensively with mouse DNA than with DNA from yeast. The hybridization of DNA strands from different sources forms the basis for a powerful set of techniques essential to the practice of modern molecular genetics. A specific DNA sequence or gene can be detected in the presence of many other sequences, if one already has an appropriate complementary DNA strand (usually la- beled in some way) to hybridize with it (Chapter 9). The complementary DNA can be from a different species or from the same species, or it can be synthesized chemi- cally in the laboratory using techniques described later in this chapter. Hybridization techniques can be varied to detect a specific RNA rather than DNA. The isolation and identification of specific genes and RNAs rely on these hybridization techniques. Applications of this technology make possible the identification of an indi- vidual on the basis of a single hair left at the scene of a crime or the prediction of the onset of a disease decades before symptoms appear (see Box 9–1). Nucleotides and Nucleic Acids Undergo Nonenzymatic Transformations Purines and pyrimidines, along with the nucleotides of which they are a part, undergo a number of spontaneous alterations in their covalent structure. The rate of these reactions is generally very slow, but they are physio- logically significant because of the cell’s very low toler- ance for alterations in its genetic information. Alter- ations in DNA structure that produce permanent changes in the genetic information encoded therein are called mutations, and much evidence suggests an inti- mate link between the accumulation of mutations in an individual organism and the processes of aging and carcinogenesis. Several nucleotide bases undergo spontaneous loss of their exocyclic amino groups (deamination) (Fig. 8–33a). For example, under typical cellular conditions, deamination of cytosine (in DNA) to uracil occurs in about one of every 10 7 cytidine residues in 24 hours. This corresponds to about 100 spontaneous events per day, on average, in a mammalian cell. Deamination of adenine and guanine occurs at about 1/100th this rate. The slow cytosine deamination reaction seems in- nocuous enough, but is almost certainly the reason why DNA contains thymine rather than uracil. The product of cytosine deamination (uracil) is readily recognized as foreign in DNA and is removed by a repair system (Chapter 25). If DNA normally contained uracil, recog- nition of uracils resulting from cytosine deamination would be more difficult, and unrepaired uracils would lead to permanent sequence changes as they were paired with adenines during replication. Cytosine deam- ination would gradually lead to a decrease in GqC base pairs and an increase in AUU base pairs in the DNA of all cells. Over the millennia, cytosine deamination could eliminate GqC base pairs and the genetic code that de- pends on them. Establishing thymine as one of the four bases in DNA may well have been one of the crucial turning points in evolution, making the long-term stor- age of genetic information possible. Another important reaction in deoxyribonu- cleotides is the hydrolysis of the N-H9252-glycosyl bond be- tween the base and the pentose (Fig. 8–33b). This oc- curs at a higher rate for purines than for pyrimidines. As many as one in 10 5 purines (10,000 per mammalian cell) are lost from DNA every 24 hours under typical 8.3 Nucleic Acid Chemistry 293 FIGURE 8–32 DNA hybridization. Two DNA samples to be compared are completely denatured by heating. When the two solutions are mixed and slowly cooled, DNA strands of each sample associate with their normal complementary partner and anneal to form duplexes. If the two DNAs have significant sequence similarity, they also tend to form partial duplexes or hybrids with each other: the greater the se- quence similarity between the two DNAs, the greater the number of hybrids formed. Hybrid formation can be measured in several ways. One of the DNAs is usually labeled with a radioactive isotope to sim- plify the measurements. cellular conditions. Depurination of ribonucleotides and RNA is much slower and generally is not considered physiologically significant. In the test tube, loss of purines can be accelerated by dilute acid. Incubation of DNA at pH 3 causes selective removal of the purine bases, resulting in a derivative called apurinic acid. Other reactions are promoted by radiation. UV light induces the condensation of two ethylene groups to form a cyclobutane ring. In the cell, the same reaction between adjacent pyrimidine bases in nucleic acids forms cyclobutane pyrimidine dimers. This happens most frequently between adjacent thymidine residues on the same DNA strand (Fig. 8–34). A second type of pyrimidine dimer, called a 6-4 photoproduct, is also formed during UV irradiation. Ionizing radiation (x rays and gamma rays) can cause ring opening and fragmen- tation of bases as well as breaks in the covalent back- bone of nucleic acids. Virtually all forms of life are exposed to energy-rich radiation capable of causing chemical changes in DNA. Near-UV radiation (with wavelengths of 200 to 400 nm), which makes up a significant portion of the solar spec- trum, is known to cause pyrimidine dimer formation and other chemical changes in the DNA of bacteria and of human skin cells. We are subject to a constant field of ionizing radiation in the form of cosmic rays, which can penetrate deep into the earth, as well as radiation emit- ted from radioactive elements, such as radium, pluto- nium, uranium, radon, 14 C, and 3 H. X rays used in med- ical and dental examinations and in radiation therapy of cancer and other diseases are another form of ionizing radiation. It is estimated that UV and ionizing radiations are responsible for about 10% of all DNA damage caused by environmental agents. DNA also may be damaged by reactive chemicals in- troduced into the environment as products of industrial activity. Such products may not be injurious per se but may be metabolized by cells into forms that are. Two prominent classes of such agents (Fig. 8–35) are (1) deaminating agents, particularly nitrous acid (HNO 2 ) or compounds that can be metabolized to nitrous acid or nitrites, and (2) alkylating agents. Nitrous acid, formed from organic precursors such as nitrosamines and from nitrite and nitrate salts, is a potent accelerator of the deamination of bases. Bisulfite has similar effects. Both agents are used as preserva- tives in processed foods to prevent the growth of toxic bacteria. They do not appear to increase cancer risks Chapter 8 Nucleotides and Nucleic Acids294 3 (a) Deamination 3 2 2 2 2 CH N HN Hypoxanthine Uracil N Cytosine O CH NO N Xanthine NO N O HN NH NH N N N NH Thymine5-Methylcytosine O HN O O N N N N Adenine Guanine O N H N N O N N N H N O HNHN FIGURE 8–33 Some well-characterized nonenzymatic reactions of nucleotides. (a) Deamination reactions. Only the base is shown. (b) Depurination, in which a purine is lost by hydrolysis of the N-H9252- glycosyl bond. The deoxyribose remaining after depurination is readily converted from the H9252-furanose to the aldehyde form (see Fig. 8–3). Fur- ther nonenzymatic reactions are illustrated in Figures 8–34 and 8–35. significantly when used in this way, perhaps because they are used in small amounts and make only a minor contribution to the overall levels of DNA damage. (The potential health risk from food spoilage if these preser- vatives were not used is much greater.) Alkylating agents can alter certain bases of DNA. For example, the highly reactive chemical dimethylsul- fate (Fig. 8–35b) can methylate a guanine to yield O 6 - methylguanine, which cannot base-pair with cytosine. Many similar reactions are brought about by alkylating agents normally present in cells, such as S-adenosyl- methionine. Possibly the most important source of mutagenic al- terations in DNA is oxidative damage. Excited-oxygen species such as hydrogen peroxide, hydroxyl radicals, and superoxide radicals arise during irradiation or as a byproduct of aerobic metabolism. Of these species, the hydroxyl radicals are responsible for most oxidative DNA damage. Cells have an elaborate defense system to destroy reactive oxygen species, including enzymes such as catalase and superoxide dismutase that convert reactive oxygen species to harmless products. A frac- tion of these oxidants inevitably escape cellular de- fenses, however, and damage to DNA occurs through any of a large, complex group of reactions ranging from oxidation of deoxyribose and base moieties to strand breaks. Accurate estimates for the extent of this dam- age are not yet available, but every day the DNA of each human cell is subjected to thousands of damaging oxidative reactions. This is merely a sampling of the best-understood reactions that damage DNA. Many carcinogenic com- pounds in food, water, or air exert their cancer-causing effects by modifying bases in DNA. Nevertheless, the in- tegrity of DNA as a polymer is better maintained than that of either RNA or protein, because DNA is the only macromolecule that has the benefit of biochemical repair systems. These repair processes (described in Chapter 25) greatly lessen the impact of damage to DNA. 8.3 Nucleic Acid Chemistry 295 OH C N H OH C O C C H N O C N H CH 3 C O C C H N UV lightUV light Adjacent thymines Cyclobutane thymine dimer 6-4 Photoproduct O C N HCH 3 C O C C H N O C N H CH 3 C O C C H N O C N H CH 3 C O C C H N O C H CH 3 C O C C H N 4 6 CH 3 5 665 (a) N P P P FIGURE 8–34 Formation of pyrimidine dimers induced by UV light. (a) One type of reaction (on the left) results in the formation of a cyclobutyl ring involving C-5 and C-6 of adjacent pyrimidine residues. An alternative reaction (on the right) results in a 6-4 photoproduct, with a linkage between C-6 of one pyrimidine and C-4 of its neighbor. (b) Formation of a cyclobutane pyrimidine dimer introduces a bend or kink into the DNA. Some Bases of DNA Are Methylated Certain nucleotide bases in DNA molecules are enzy- matically methylated. Adenine and cytosine are methy- lated more often than guanine and thymine. Methyla- tion is generally confined to certain sequences or regions of a DNA molecule. In some cases the function of methylation is well understood; in others the func- tion remains unclear. All known DNA methylases use S- adenosylmethionine as a methyl group donor. E. coli has two prominent methylation systems. One serves as part of a defense mechanism that helps the cell to dis- tinguish its DNA from foreign DNA by marking its own DNA with methyl groups and destroying (foreign) DNA without the methyl groups (this is known as a restriction-modification system; see Chapter 9). The other system methylates adenosine residues within the sequence (5H11032)GATC(3H11032) to N 6 -methyladenosine (Fig. 8–5a). This is mediated by the Dam (DNA adenine methylation) methylase, a component of a system that repairs mismatched base pairs formed occasionally dur- ing DNA replication (see Fig. 25–20). In eukaryotic cells, about 5% of cytidine residues in DNA are methylated to 5-methylcytidine (Fig. 8–5a). Methylation is most common at CpG sequences, pro- ducing methyl-CpG symmetrically on both strands of the DNA. The extent of methylation of CpG sequences varies by molecular region in large eukaryotic DNA mol- ecules. Methylation suppresses the migration of seg- ments of DNA called transposons, described in Chapter 25. These methylations of cytosine also have structural significance. The presence of 5-methylcytosine in an al- ternating CpG sequence markedly increases the ten- dency for that segment of DNA to assume the Z form. The Sequences of Long DNA Strands Can Be Determined In its capacity as a repository of information, a DNA mol- ecule’s most important property is its nucleotide se- quence. Until the late 1970s, determining the sequence of a nucleic acid containing even five or ten nucleotides was difficult and very laborious. The development of two new techniques in 1977, one by Alan Maxam and Walter Gilbert and the other by Frederick Sanger, has made pos- sible the sequencing of ever larger DNA molecules with an ease unimagined just a few decades ago. The tech- niques depend on an improved understanding of nu- cleotide chemistry and DNA metabolism, and on elec- trophoretic methods for separating DNA strands differing in size by only one nucleotide. Electrophoresis of DNA is similar to that of proteins (see Fig. 3–19). Polyacrylamide is often used as the gel matrix in work with short DNA molecules (up to a few hundred nucleotides); agarose is generally used for longer pieces of DNA. In both Sanger and Maxam-Gilbert sequencing, the general principle is to reduce the DNA to four sets of la- beled fragments. The reaction producing each set is base-specific, so the lengths of the fragments correspond to positions in the DNA sequence where a certain base occurs. For example, for an oligonucleotide with the se- quence pAATCGACT, labeled at the 5H11032 end (the left end), a reaction that breaks the DNA after each C residue will generate two labeled fragments: a four-nucleotide and a seven-nucleotide fragment; a reaction that breaks the DNA after each G will produce only one labeled, five- nucleotide fragment. Because the fragments are radio- actively labeled at their 5H11032 ends, only the fragment to the 5H11032 side of the break is visualized. The fragment sizes cor- respond to the relative positions of C and G residues in the sequence. When the sets of fragments corresponding to each of the four bases are electrophoretically sepa- rated side by side, they produce a ladder of bands from which the sequence can be read directly (Fig. 8–36). We illustrate only the Sanger method, because it has proven to be technically easier and is in more widespread use. It requires the enzymatic synthesis of a DNA strand com- plementary to the strand under analysis, using a radio- actively labeled “primer” and dideoxynucleotides. Chapter 8 Nucleotides and Nucleic Acids296 FIGURE 8–35 Chemical agents that cause DNA damage. (a) Precursors of nitrous acid, which promotes deamination reactions. (b) Alkylating agents. 8.3 Nucleic Acid Chemistry 297 (a) P P dATP dGTP 3H11032 P P P OH A P P P P P P PP ATCGGC OH A OH G P C P C P G P T 5H11032 Primer strand Template strand PP TC FIGURE 8–36 DNA sequencing by the Sanger method. This method makes use of the mechanism of DNA synthesis by DNA polymerases (Chapter 25). (a) DNA polymerases require both a primer (a short oligonucleotide strand), to which nucleotides are added, and a template strand to guide selection of each new nucleotide. In cells, the 3H11032-hy- droxyl group of the primer reacts with an incoming deoxynucleoside triphosphate (dNTP) to form a new phosphodiester bond. (b) The Sanger sequencing procedure uses dideoxynucleoside triphosphate (ddNTP) analogs to interrupt DNA synthesis. (The Sanger method is also known as the dideoxy method.) When a ddNTP is inserted in place of a dNTP, strand elongation is halted after the analog is added, because it lacks the 3H11032-hydroxyl group needed for the next step. (c) The DNA to be sequenced is used as the tem- plate strand, and a short primer, radioactively or fluorescently labeled, is an- nealed to it. By addition of small amounts of a single ddNTP, for example ddCTP, to an otherwise normal reaction system, the synthesized strands will be prematurely terminated at some lo- cations where dC normally occurs. Given the excess of dCTP over ddCTP, the chance that the analog will be incorporated whenever a dC is to be added is small. However, ddCTP is present in sufficient amounts to ensure that each new strand has a high proba- bility of acquiring at least one ddC at some point during synthesis. The result is a solution containing a mixture of labeled fragments, each end- ing with a C residue. Each C residue in the sequence generates a set of fragments of a particular length, such that the different-sized frag- ments, separated by electrophoresis, reveal the location of C residues. This procedure is repeated separately for each of the four ddNTPs, and the sequence can be read directly from an autoradiogram of the gel. Because shorter DNA fragments migrate faster, the fragments near the bottom of the gel represent the nucleotide positions closest to the primer (the 5H11032 end), and the sequence is read (in the 5H11032 n 3H11032 direction) from bottom to top. Note that the sequence obtained is that of the strand complementary to the strand being analyzed. DNA sequencing is readily automated by a varia- tion of Sanger’s sequencing method in which the dideoxynucleotides used for each reaction are labeled with a differently colored fluorescent tag (Fig. 8–37). This technology allows DNA sequences containing thou- sands of nucleotides to be determined in a few hours. Entire genomes of many organisms have now been se- quenced (see Table 1–4), and many very large DNA- sequencing projects are in progress. Perhaps the most ambitious of these is the Human Genome Project, in which researchers have sequenced all 3.2 billion base pairs of the DNA in a human cell (Chapter 9). Dideoxy Sequencing of DNA The Chemical Synthesis of DNA Has Been Automated Another technology that has paved the way for many biochemical advances is the chemical synthesis of oligonucleotides with any chosen sequence. The chem- ical methods for synthesizing nucleic acids were devel- oped primarily by H. Gobind Khorana and his colleagues in the 1970s. Refinement and automation of these meth- ods have made possible the rapid and accurate synthe- sis of DNA strands. The synthesis is carried out with the growing strand attached to a solid support (Fig. 8–38), using principles similar to those used by Merrifield in peptide synthesis (see Fig. 3–29). The efficiency of each addition step is very high, allowing the routine labora- tory synthesis of polymers containing 70 or 80 nu- cleotides and, in some laboratories, much longer strands. The availability of relatively inexpensive DNA polymers with predesigned sequences is having a pow- erful impact on all areas of biochemistry (Chapter 9). Chapter 8 Nucleotides and Nucleic Acids298 FIGURE 8–37 Strategy for automating DNA sequencing reactions. Each dideoxynucleotide used in the Sanger method can be linked to a fluorescent molecule that gives all the fragments terminating in that nucleotide a particular color. All four labeled ddNTPs are added to a single tube. The resulting colored DNA fragments are then separated by size in a single electrophoretic gel contained in a capillary tube (a refinement of gel electrophoresis that allows for faster separations). All fragments of a given length migrate through the capillary gel in a single peak, and the color associated with each peak is detected using a laser beam. The DNA sequence is read by determining the sequence of colors in the peaks as they pass the detector. This information is fed directly to a computer, which determines the sequence. Nucleoside attached to silica support 1 42Repeat steps to until all residues are added CH 2 H O H HH Base 1 O OH H 3H11032 5H11032 DMT Nucleoside protected at 5H11032 hydroxyl R Si O H O H HH Base 1 O H CH 2 H DMT O CH 2 H O H H Base 1 O H R Si H O H OP H H Base 2 O R Si CH 2 H O HH HH Base 1 O CH 2 H O O O H 2 NC (CH 2 ) DMT O H OP H HH Base 2 O R Si CH 2 H O HH HH Base 1 O CH 2 H O O 2 NC (CH 2 ) DMT Oxidation to form triester 4 DMT Protecting group removed 2 2 H Next nucleotide added 3 N H11001 CH 2 CH 2 H NCH(CH 3 ) 2 CH (CH 3 ) H OP H HH Base 2 O Nucleotide activated at 3H11032 position CH 2 H O O (CH 3 ) (CH 3 ) 2 NC (CH 2 ) Cyanoethyl protecting group Diisopropylamine byproduct DMT Diisopropylamino activating group Remove protecting groups from bases Remove cyanoethyl groups from phosphates Cleave chain from silica support7 5 6 5H11032 3H11032 Oligonucleotide chain 8.3 Nucleic Acid Chemistry 299 FIGURE 8–38 Chemical synthesis of DNA. Automated DNA synthe- sis is conceptually similar to the synthesis of polypeptides on a solid support. The oligonucleotide is built up on the solid support (silica), one nucleotide at a time, in a repeated series of chemical reactions with suitably protected nucleotide precursors. 1 The first nucleoside (which will be the 3H11032 end) is attached to the silica support at the 3H11032 hydroxyl (through a linking group, R) and is protected at the 5H11032 hy- droxyl with an acid-labile dimethoxytrityl group (DMT). The reactive groups on all bases are also chemically protected. 2 The protecting DMT group is removed by washing the column with acid (the DMT group is colored, so this reaction can be followed spectrophotomet- rically). 3 The next nucleotide is activated with a diisopropylamino group and reacted with the bound nucleotide to form a 5H11032,3H11032 linkage, which in step 4 is oxidized with iodine to produce a phosphotri- ester linkage. (One of the phosphate oxygens carries a cyanoethyl pro- tecting group.) Reactions 2 through 4 are repeated until all nu- cleotides are added. At each step, excess nucleotide is removed before addition of the next nucleotide. In steps 5 and 6 the remaining protecting groups on the bases and the phosphates are removed, and in 7 the oligonucleotide is separated from the solid support and pu- rified. The chemical synthesis of RNA is somewhat more complicated because of the need to protect the 2H11032 hydroxyl of ribose without ad- versely affecting the reactivity of the 3H11032 hydroxyl. SUMMARY 8.3 Nucleic Acid Chemistry ■ Native DNA undergoes reversible unwinding and separation of strands (melting) on heating or at extremes of pH. DNAs rich in GqC pairs have higher melting points than DNAs rich in AUT pairs. ■ Denatured single-stranded DNAs from two species can form a hybrid duplex, the degree of hybridization depending on the extent of sequence similarity. Hybridization is the basis for important techniques used to study and isolate specific genes and RNAs. ■ DNA is a relatively stable polymer. Spontaneous reactions such as deamination of certain bases, hydrolysis of base-sugar N-glycosyl bonds, radiation-induced formation of pyrimidine dimers, and oxidative damage occur at very low rates, yet are important because of cells’ very low tolerance for changes in genetic material. ■ DNA sequences can be determined and DNA polymers synthesized with simple, automated protocols involving chemical and enzymatic methods. 8.4 Other Functions of Nucleotides In addition to their roles as the subunits of nucleic acids, nucleotides have a variety of other functions in every cell: as energy carriers, components of enzyme cofac- tors, and chemical messengers. Nucleotides Carry Chemical Energy in Cells The phosphate group covalently linked at the 5H11032 hy- droxyl of a ribonucleotide may have one or two addi- tional phosphates attached. The resulting molecules are referred to as nucleoside mono-, di-, and triphosphates (Fig. 8–39). Starting from the ribose, the three phos- phates are generally labeled H9251, H9252, and H9253. Hydrolysis of nucleoside triphosphates provides the chemical energy to drive a wide variety of cellular reactions. Adenosine 5H11032-triphosphate, ATP, is by far the most widely used for this purpose, but UTP, GTP, and CTP are also used in some reactions. Nucleoside triphosphates also serve as the activated precursors of DNA and RNA synthesis, as described in Chapters 25 and 26. The energy released by hydrolysis of ATP and the other nucleoside triphosphates is accounted for by the structure of the triphosphate group. The bond between the ribose and the H9251 phosphate is an ester linkage. The H9251,H9252 and H9252,H9253 linkages are phosphoanhydrides (Fig. 8–40). Hydrolysis of the ester linkage yields about 14 kJ/mol under standard conditions, whereas hydrolysis of each anhydride bond yields about 30 kJ/mol. ATP hy- drolysis often plays an important thermodynamic role in biosynthesis. When coupled to a reaction with a pos- itive free-energy change, ATP hydrolysis shifts the equi- librium of the overall process to favor product forma- Chapter 8 Nucleotides and Nucleic Acids300 Abbreviations of ribonucleoside 5H11032-phosphates Base Mono- Di- Tri- Adenine Guanine Cytosine Thymine AMP GMP CMP UMP ADP GDP CDP UDP ATP GTP CTP UTP Abbreviations of deoxyribonucleoside Base Mono- Di- Tri- Adenine Guanine Cytosine Uracil dAMP dGMP dCMP dTMP dADP dGDP dCDP dTDP dATP dGTP dCTP dTTP 5H11032-phosphates O H11002 OCH 2 H O H11002 P O H11002 O H11002 O P O OOP OO H H H H Base O OH H9251H9252H9253 NMP NDP NTP FIGURE 8–39 Nucleoside phosphates. General structure of the nucleoside 5H11032-mono-, di-, and triphosphates (NMPs, NDPs, and NTPs) and their standard abbreviations. In the deoxyribonucleoside phosphates (dNMPs, dNDPs, and dNTPs), the pentose is 2H11032-deoxy-D-ribose. FIGURE 8–40 The phosphate ester and phosphoanhydride bonds of ATP. Hydrolysis of an anhydride bond yields more energy than hy- drolysis of the ester. A carboxylic acid anhydride and carboxylic acid ester are shown for comparison. tion (recall the relationship between equilibrium con- stant and free-energy change described by Eqn 6–3 on p. 195). Adenine Nucleotides Are Components of Many Enzyme Cofactors A variety of enzyme cofactors serving a wide range of chemical functions include adenosine as part of their structure (Fig. 8–41). They are unrelated structurally except for the presence of adenosine. In none of these cofactors does the adenosine portion participate directly in the primary function, but removal of adenosine gen- erally results in a drastic reduction of cofactor activi- ties. For example, removal of the adenine nucleotide (3H11032-phosphoadenosine diphosphate) from acetoacetyl- CoA, the coenzyme A derivative of acetoacetate, re- duces its reactivity as a substrate for H9252-ketoacyl-CoA transferase (an enzyme of lipid metabolism) by a factor of 10 6 . Although this requirement for adenosine has not been investigated in detail, it must involve the binding energy between enzyme and substrate (or cofactor) that is used both in catalysis and in stabilizing the initial enzyme-substrate complex (Chapter 6). In the case of H9252-ketoacyl-CoA transferase, the nucleotide moiety of coenzyme A appears to be a binding “handle” that helps to pull the substrate (acetoacetyl-CoA) into the active site. Similar roles may be found for the nucleoside por- tion of other nucleotide cofactors. Why is adenosine, rather than some other large mol- ecule, used in these structures? The answer here may involve a form of evolutionary economy. Adenosine is 8.4 Other Functions of Nucleotides 301 H11002 O H O H11002 P O H11002 O O O P H H H OOH CH 2 O OCH 3 C N CH 2 NH 2 N N N NC CC N CH 2 C NCH 2 CH 2 H HH H CH 3 HS O O OH CH 3 5H11032 P O O O H11002 O H11002 3H11032-Phosphoadenosine diphosphate (3H11032-P-ADP) Pantothenic acidb-Mercaptoethylamine Coenzyme A CH 2 CHOH CHOH CHOH CH 2 O O P H11001 O O O P O H11002 CH 2 H O H H N NH 2 N N N H OH 2H11032 4H11032 1H11032 3H11032 CH 2 O H ON O N N CH 3 NH 2 Riboflavin Nicotinamide adenine dinucleotide (NAD H11545 ) H O H H N N N N H HOH CH 2 O NH 2 Flavin adenine dinucleotide (FAD) H O H H N H HOH CH 2 O Nicotinamide OP H11002 O O O O PO H11002 O O FIGURE 8–41 Some coenzymes containing adenosine. The adenosine portion is shaded in light red. Coenzyme A (CoA) functions in acyl group transfer reactions; the acyl group (such as the acetyl or acetoacetyl group) is attached to the CoA through a thioester linkage to the H9252-mercaptoethylamine moiety. NAD H11001 func- tions in hydride transfers, and FAD, the active form of vitamin B 2 (riboflavin), in electron transfers. Another coenzyme incorporating adenosine is 5H11032-deoxyadenosylcobalamin, the active form of vita- min B 12 (see Box 17-2), which participates in intramolecular group transfers between adjacent carbons. certainly not unique in the amount of potential binding energy it can contribute. The importance of adenosine probably lies not so much in some special chemical char- acteristic as in the evolutionary advantage of using one compound for multiple roles. Once ATP became the uni- versal source of chemical energy, systems developed to synthesize ATP in greater abundance than the other nu- cleotides; because it is abundant, it becomes the logical choice for incorporation into a wide variety of struc- tures. The economy extends to protein structure. A sin- gle protein domain that binds adenosine can be used in a wide variety of enzymes. Such a domain, called a nucleotide-binding fold, is found in many enzymes that bind ATP and nucleotide cofactors. Some Nucleotides Are Regulatory Molecules Cells respond to their environment by taking cues from hormones or other external chemical signals. The in- teraction of these extracellular chemical signals (“first messengers”) with receptors on the cell surface often leads to the production of second messengers inside the cell, which in turn leads to adaptive changes in the cell interior (Chapter 12). Often, the second mes- senger is a nucleotide (Fig. 8–42). One of the most common is adenosine 3H11541,5H11541-cyclic monophosphate (cyclic AMP, or cAMP), formed from ATP in a reac- tion catalyzed by adenylyl cyclase, an enzyme associ- ated with the inner face of the plasma membrane. Cyclic AMP serves regulatory functions in virtually every cell outside the plant kingdom. Guanosine 3H11032,5H11032-cyclic mono- phosphate (cGMP) occurs in many cells and also has regulatory functions. Another regulatory nucleotide, ppGpp (Fig. 8–42), is produced in bacteria in response to a slowdown in protein synthesis during amino acid starvation. This nu- cleotide inhibits the synthesis of the rRNA and tRNA molecules (see Fig. 28–24) needed for protein synthe- sis, preventing the unnecessary production of nucleic acids. SUMMARY 8.4 Other Functions of Nucleotides ■ ATP is the central carrier of chemical energy in cells. The presence of an adenosine moiety in a variety of enzyme cofactors may be related to binding-energy requirements. ■ Cyclic AMP, formed from ATP in a reaction catalyzed by adenylyl cyclase, is a common second messenger produced in response to hormones and other chemical signals. Chapter 8 Nucleotides and Nucleic Acids302 FIGURE 8–42 Three regulatory nucleotides. Key Terms gene 273 ribosomal RNA (rRNA) 273 messenger RNA (mRNA) 273 transfer RNA (tRNA) 273 nucleotide 273 nucleoside 273 pyrimidine 273 purine 273 deoxyribonucleotides 274 ribonucleotide 274 phosphodiester linkage 277 Terms in bold are defined in the glossary. 5H11032 end 277 3H11032 end 277 oligonucleotide 278 polynucleotide 278 base pair 279 major groove 282 minor groove 282 B-form DNA 284 A-form DNA 284 Z-form DNA 284 palindrome 285 hairpin 285 cruciform 285 triplex DNA 286 G tetraplex 287 H-DNA 287 monocistronic mRNA 287 polycistronic mRNA 288 mutation 293 second messenger 302 adenosine 3H11541,5H11541-cyclic monophos- phate (cyclic AMP, cAMP) 302 Chapter 8 Problems 303 Further Reading General Chang, K.Y. & Varani, G. (1997) Nucleic acids structure and recognition. Nat. Struct. Biol. 4 (Suppl.), 854–858. Describes the application of NMR to determination of nucleic acid structure. Friedberg, E.C., Walker, G.C., & Siede, W. (1995) DNA Repair and Mutagenesis, W. H. Freeman and Company, New York. A good source for more information on the chemistry of nucleotides and nucleic acids. Hecht, S.M. (ed.) (1996) Bioorganic Chemistry: Nucleic Acids, Oxford University Press, Oxford. A very useful set of articles. Kornberg, A. & Baker, T.A. (1991) DNA Replication, 2nd edn, W. H. Freeman and Company, New York. The best place to start to learn more about DNA structure. Sinden, R.R. (1994) DNA Structure and Function, Academic Press, Inc., San Diego. Good discussion of many topics covered in this chapter. Historical Judson, H.F. (1996) The Eighth Day of Creation: Makers of the Revolution in Biology, expanded edn, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY. Olby, R.C. (1994) The Path to the Double Helix: The Discovery of DNA, Dover Publications, Inc., New York. Sayre, A. (1978) Rosalind Franklin and DNA, W. W. Norton & Co., Inc., New York. Watson, J.D. (1968) The Double Helix: A Personal Account of the Discovery of the Structure of DNA, Atheneum, New York. [Paperback edition, Touchstone Books, 2001.] Variations in DNA Structure Frank-Kamenetskii, M.D. & Mirkin, S.M. (1995) Triplex DNA structures. Annu. Rev. Biochem. 64, 65–95. Herbert, A. & Rich, A. (1996) The biology of left-handed Z-DNA. J. Biol. Chem. 271, 11,595–11,598. Htun, H. & Dahlberg, J.E. (1989) Topology and formation of triple-stranded H-DNA. Science 243, 1571–1576. Keniry, M.A. (2000) Quadruplex structures in nucleic acids. Biopolymers 56, 123–146. Good summary of the structural properties of quadruplexes. Moore, P.B. (1999) Structural motifs in RNA. Annu. Rev. Biochem. 68, 287–300. Shafer, R.H. (1998) Stability and structure of model DNA triplexes and quadruplexes and their interactions with small ligands. Prog. Nucleic Acid Res. Mol. Biol. 59, 55–94. Wells, R.D. (1988) Unusual DNA structures. J. Biol. Chem. 263, 1095–1098. Minireview; a concise summary. Nucleic Acid Chemistry Collins, A.R. (1999) Oxidative DNA damage, antioxidants, and cancer. Bioessays 21, 238–246. Marnett, L.J. & Plastaras, J.P. (2001) Endogenous DNA damage and mutation. Trends Genet. 17, 214–221. ATP As Energy Carrier Jencks, W.P. (1987) Economics of enzyme catalysis. Cold Spring Harb. Symp. Quant. Biol. 52, 65–73. A relatively short article, full of insights. 1. Nucleotide Structure Which positions in a purine ring of a purine nucleotide in DNA have the potential to form hydrogen bonds but are not involved in Watson-Crick base pairing? 2. Base Sequence of Complementary DNA Strands One strand of a double-helical DNA has the sequence (5H11032)GCGCAATATTTCTCAAAATATTGCGC(3H11032). Write the base sequence of the complementary strand. What special type of sequence is contained in this DNA segment? Does the dou- ble-stranded DNA have the potential to form any alternative structures? 3. DNA of the Human Body Calculate the weight in grams of a double-helical DNA molecule stretching from the earth to the moon (~320,000 km). The DNA double helix weighs about 1 H11003 10 H1100218 g per 1,000 nucleotide pairs; each base pair extends 3.4 ?. For an interesting comparison, your body contains about 0.5 g of DNA! 4. DNA Bending Assume that a poly(A) tract five base pairs long produces a 20H11034 bend in a DNA strand. Calculate the total (net) bend produced in a DNA if the center base pairs (the third of five) of two successive (dA) 5 tracts are lo- cated (a) 10 base pairs apart; (b) 15 base pairs apart. As- sume 10 base pairs per turn in the DNA double helix. 5. Distinction between DNA Structure and RNA Structure Hairpins may form at palindromic sequences in single strands of either RNA or DNA. How is the helical struc- ture of a long and fully base-paired (except at the end) hair- pin in RNA different from that of a similar hairpin in DNA? 6. Nucleotide Chemistry The cells of many eukaryotic organisms have highly specialized systems that specifically repair G–T mismatches in DNA. The mismatch is repaired to form a GqC (not AUT) base pair. This G–T mismatch repair mechanism occurs in addition to a more general system that repairs virtually all mismatches. Can you suggest why cells might require a specialized system to repair G–T mismatches? 7. Nucleic Acid Structure Explain why the absorption of UV light by double-stranded DNA increases (hyperchromic effect) when the DNA is denatured. Problems Chapter 8 Nucleotides and Nucleic Acids304 8. Determination of Protein Concentration in a So- lution Containing Proteins and Nucleic Acids The con- centration of protein or nucleic acid in a solution containing both can be estimated by using their different light absorp- tion properties: proteins absorb most strongly at 280 nm and nucleic acids at 260 nm. Their respective concentrations in a mixture can be estimated by measuring the absorbance (A) of the solution at 280 nm and 260 nm and using the table be- low, which gives R 280/260 , the ratio of absorbances at 280 and 260 nm; the percentage of total mass that is nucleic acid; and a factor, F, that corrects the A 280 reading and gives a more accurate protein estimate. The protein concentration (in mg/ml) H11005 F H11003 A 280 (assuming the cuvette is 1 cm wide). Cal- culate the protein concentration in a solution of A 280 H11005 0.69 and A 260 H11005 0.94. 9. Base Pairing in DNA In samples of DNA isolated from two unidentified species of bacteria, X and Y, adenine makes up 32% and 17%, respectively, of the total bases. What rela- tive proportions of adenine, guanine, thymine, and cytosine would you expect to find in the two DNA samples? What as- sumptions have you made? One of these species was isolated from a hot spring (64 H11034C). Suggest which species is the ther- mophilic bacterium. What is the basis for your answer? 10. Solubility of the Components of DNA Draw the fol- lowing structures and rate their relative solubilities in water (most soluble to least soluble): deoxyribose, guanine, phos- phate. How are these solubilities consistent with the three- dimensional structure of double-stranded DNA? 11. DNA Sequencing The following DNA fragment was sequenced by the Sanger method. The red asterisk indicates a fluorescent label. A sample of the DNA was reacted with DNA polymerase and each of the nucleotide mixtures (in an appropriate buffer) listed below. Dideoxynucleotides (ddNTPs) were added in relatively small amounts. 1. dATP, dTTP, dCTP, dGTP, ddTTP 2. dATP, dTTP, dCTP, dGTP, ddGTP 3. dATP, dCTP, dGTP, ddTTP 4. dATP, dTTP, dCTP, dGTP The resulting DNA was separated by electrophoresis on an agarose gel, and the fluorescent bands on the gel were located. The band pattern resulting from nucleotide mixture 1 is shown below. Assuming that all mixtures were run on the same gel, what did the remaining lanes of the gel look like? 12. Snake Venom Phosphodiesterase An exonuclease is an enzyme that sequentially cleaves nucleotides from the end of a polynucleotide strand. Snake venom phosphodiesterase, which hydrolyzes nucleotides from the 3H11032 end of any oligonu- cleotide with a free 3H11032-hydroxyl group, cleaves between the 3H11032 hydroxyl of the ribose or deoxyribose and the phosphoryl group of the next nucleotide. It acts on single-stranded DNA or RNA and has no base specificity. This enzyme was used in sequence determination experiments before the development of modern nucleic acid sequencing techniques. What are the products of partial digestion by snake venom phosphodi- esterase of an oligonucleotide with the following sequence? (5H11032)GCGCCAUUGC(3H11032)–OH 13. Preserving DNA in Bacterial Endospores Bacter- ial endospores form when the environment is no longer con- ducive to active cell metabolism. The soil bacterium Bacillus subtilis, for example, begins the process of sporulation when one or more nutrients are depleted. The end product is a Proportion of R 280/260 nucleic acid (%) F 1.75 0.00 1.116 1.63 0.25 1.081 1.52 0.50 1.054 1.40 0.75 1.023 1.36 1.00 0.994 1.30 1.25 0.970 1.25 1.50 0.944 1.16 2.00 0.899 1.09 2.50 0.852 1.03 3.00 0.814 0.979 3.50 0.776 0.939 4.00 0.743 0.874 5.00 0.682 0.846 5.50 0.656 0.822 6.00 0.632 0.804 6.50 0.607 0.784 7.00 0.585 0.767 7.50 0.565 0.753 8.00 0.545 0.730 9.00 0.508 0.705 10.00 0.478 0.671 12.00 0.422 0.644 14.00 0.377 0.615 17.00 0.322 0.595 20.00 0.278 Chapter 8 Problems 305 small, metabolically dormant structure that can survive al- most indefinitely with no detectable metabolism. Spores have mechanisms to prevent accumulation of potentially lethal mu- tations in their DNA over periods of dormancy that can ex- ceed 1,000 years. B. subtilis spores are much more resistant than the organism’s growing cells to heat, UV radiation, and oxidizing agents, all of which promote mutations. (a) One factor that prevents potential DNA damage in spores is their greatly decreased water content. How would this affect some types of mutations? (b) Endospores have a category of proteins called small acid-soluble proteins (SASPs) that bind to their DNA, pre- venting formation of cyclobutane-type dimers. What causes cyclobutane dimers, and why do bacterial endospores need mechanisms to prevent their formation? Biochemistry on the Internet 14. The Structure of DNA Elucidation of the three- dimensional structure of DNA helped researchers understand how this molecule conveys information that can be faithfully replicated from one generation to the next. To see the second- ary structure of double-stranded DNA, go to the Protein Data Bank website (www.rcsb.org/pdb). Use the PDB identifiers listed below to retrieve the data pages for the two forms of DNA. Open the structures using RasMol or Chime, and use the dif- ferent viewing options to complete the following exercises. (a) Obtain the file for 141D, a highly conserved, re- peated DNA sequence from the end of the HIV-1 (the virus that causes AIDS) genome. Display the molecule as a stick or ball-and-stick structure. Identify the sugar–phosphate backbone for each strand of the DNA duplex. Locate and iden- tify individual bases. Which is the 5H11032 end of this molecule? Locate the major and minor grooves. Is this a right- or left- handed helix? (b) Obtain the file for 145D, a DNA with the Z confor- mation. Display the molecule as a stick or ball-and-stick struc- ture. Identify the sugar–phosphate backbone for each strand of the DNA duplex. Is this a right- or left-handed helix? (c) To fully appreciate the secondary structure of DNA, select “Stereo” in the Options menu in the viewer. You will see two images of the DNA molecule. Sit with your nose ap- proximately 10 inches from the monitor and focus on the tip of your nose. In the background you should see three images of the DNA helix. Shift your focus from the tip of your nose to the middle image, which should appear three-dimensional. (Note that only one of the two authors can make this work.) For additional tips, see the Study Guide or the textbook web- site (www.whfreeman.com/lehninger). chapter W e now turn to a technology that is fundamental to the advance of modern biological sciences, defin- ing present and future biochemical frontiers and illus- trating many important principles of biochemistry. Elucidation of the laws governing enzymatic catalysis, macromolecular structure, cellular metabolism, and in- formation pathways allows research to be directed at in- creasingly complex biochemical processes. Cell division, immunity, embryogenesis, vision, taste, oncogenesis, cognition—all are orchestrated in an elaborate sym- phony of molecular and macromolecular interactions that we are now beginning to understand with increasing clarity. The real implications of the biochemical journey begun in the nineteenth century are found in the ever- increasing power to analyze and alter living systems. To understand a complex biological process, a bio- chemist isolates and studies the individual components in vitro, then pieces together the parts to get a coher- ent picture of the overall process. A major source of mo- lecular insights is the cell’s own information archive, its DNA. The sheer size of chromosomes, however, pres- ents an enormous challenge: how does one find and study a particular gene among the tens of thousands of genes nested in the billions of base pairs of a mammalian genome? Solutions began to emerge in the 1970s. Decades of advances by thousands of scientists working in genetics, biochemistry, cell biology, and phys- ical chemistry came together in the laboratories of Paul Berg, Herbert Boyer, and Stanley Cohen to yield tech- niques for locating, isolating, preparing, and studying small segments of DNA derived from much larger chro- mosomes. Techniques for DNA cloning paved the way to the modern fields of genomics and proteomics, the study of genes and proteins on the scale of whole cells and organisms. These new methods are transforming ba- sic research, agriculture, medicine, ecology, forensics, and many other fields, while occasionally presenting so- ciety with difficult choices and ethical dilemmas. We begin this chapter with an outline of the funda- mental biochemical principles of the now-classic disci- pline of DNA cloning. Next, after laying the groundwork for a discussion of genomics, we illustrate the range of applications and the potential of these technologies, with a broad emphasis on modern advances in genomics and proteomics. 9.1 DNA Cloning: The Basics A clone is an identical copy. This term originally applied to cells of a single type, isolated and allowed to repro- duce to create a population of identical cells. DNA cloning involves separating a specific gene or DNA seg- ment from a larger chromosome, attaching it to a small molecule of carrier DNA, and then replicating this mod- ified DNA thousands or millions of times through both an increase in cell number and the creation of multiple DNA-BASED INFORMATION TECHNOLOGIES 9.1 DNA Cloning: The Basics 306 9.2 From Genes to Genomes 317 9.3 From Genomes to Proteomes 325 9.4 Genome Alterations and New Products of Biotechnology 330 Of all the natural systems, living matter is the one which, in the face of great transformations, preserves inscribed in its organization the largest amount of its own past history. —Emile Zuckerkandl and Linus Pauling, article in Journal of Theoretical Biology, 1965 9 306 8885d_c09_306-342 2/7/04 8:14 AM Page 306 mac76 mac76:385_reb: copies of the cloned DNA in each cell. The result is selective amplification of a par- ticular gene or DNA segment. Cloning of DNA from any organism entails five gen- eral procedures: 1. Cutting DNA at precise locations. Sequence-specific endonucleases (re- striction endonucleases) provide the necessary molecular scissors. 2. Selecting a small molecule of DNA capable of self-replication. These DNAs are called cloning vectors (a vector is a delivery agent). They are typically plasmids or viral DNAs. 3. Joining two DNA fragments covalently. The enzyme DNA ligase links the cloning vector and DNA to be cloned. Composite DNA molecules comprising covalently linked segments from two or more sources are called recombinant DNAs. 4. Moving recombinant DNA from the test tube to a host cell that will provide the enzymatic machin- ery for DNA replication. 5. Selecting or identifying host cells that contain recombinant DNA. The methods used to accomplish these and related tasks are collectively referred to as recombinant DNA tech- nology or, more informally, genetic engineering. Much of our initial discussion will focus on DNA cloning in the bacterium Escherichia coli, the first or- ganism used for recombinant DNA work and still the most common host cell. E. coli has many advantages: its DNA metabolism (like many other of its biochemical processes) is well understood; many naturally occurring cloning vectors associated with E. coli, such as plasmids and bacteriophages (bacterial viruses; also called phages), are well characterized; and techniques are available for moving DNA expeditiously from one bac- terial cell to another. We also address DNA cloning in other organisms, a topic discussed more fully later in the chapter. Restriction Endonucleases and DNA Ligase Yield Recombinant DNA Particularly important to recombinant DNA technology is a set of enzymes (Table 9–1) made available through decades of research on nucleic acid metabolism. Two classes of enzymes lie at the heart of the general ap- proach to generating and propagating a recombinant DNA molecule (Fig. 9–1). First, restriction endonu- cleases (also called restriction enzymes) recognize and cleave DNA at specific DNA sequences (recognition se- quences or restriction sites) to generate a set of smaller fragments. Second, the DNA fragment to be cloned can be joined to a suitable cloning vector by using DNA lig- ases to link the DNA molecules together. The recombi- nant vector is then introduced into a host cell, which amplifies the fragment in the course of many genera- tions of cell division. Restriction endonucleases are found in a wide range of bacterial species. Werner Arber discovered in the early 1960s that their biological function is to recognize and cleave foreign DNA (the DNA of an infecting virus, for example); such DNA is said to be restricted. In the host cell’s DNA, the sequence that would be recognized 9.1 DNA Cloning: The Basics 307 Paul Berg Herbert Boyer Stanley N. Cohen TABLE 9–1 Some Enzymes Used in Recombinant DNA Technology Enzyme(s) Function Type II restriction endonucleases Cleave DNAs at specific base sequences DNA ligase Joins two DNA molecules or fragments DNA polymerase I (E. coli) Fills gaps in duplexes by stepwise addition of nucleotides to 3H11032 ends Reverse transcriptase Makes a DNA copy of an RNA molecule Polynucleotide kinase Adds a phosphate to the 5H11032-OH end of a polynucleotide to label it or permit ligation Terminal transferase Adds homopolymer tails to the 3H11032-OH ends of a linear duplex Exonuclease III Removes nucleotide residues from the 3H11032 ends of a DNA strand Bacteriophage H9261 exonuclease Removes nucleotides from the 5H11032 ends of a duplex to expose single-stranded 3H11032 ends Alkaline phosphatase Removes terminal phosphates from either the 5H11032 or 3H11032 end (or both) 8885d_c09_306-342 2/7/04 8:14 AM Page 307 mac76 mac76:385_reb: by its own restriction endonuclease is protected from digestion by methylation of the DNA, catalyzed by a spe- cific DNA methylase. The restriction endonuclease and the corresponding methylase are sometimes referred to as a restriction-modification system. There are three types of restriction endonucleases, designated I, II, and III. Types I and III are generally large, multisubunit complexes containing both the endonucle- Chapter 9 DNA-Based Information Technologies308 Cloning vector is cleaved with restriction endonuclease. Cloning vector (plasmid) DNA is introduced into the host cell. Recombinant vector Eukaryotic chromosome DNA ligase DNA fragment of interest is obtained by cleaving chromosome with a restriction endonuclease. Fragments are ligated to the prepared cloning vector. 1 2 3 4 Propagation (cloning) produces many copies of recombinant DNA. 5 FIGURE 9–1 Schematic illustration of DNA cloning. A cloning vec- tor and eukaryotic chromosomes are separately cleaved with the same restriction endonuclease. The fragments to be cloned are then ligated to the cloning vector. The resulting recombinant DNA (only one re- combinant vector is shown here) is introduced into a host cell where it can be propagated (cloned). Note that this drawing is not to scale: the size of the E. coli chromosome relative to that of a typical cloning vector (such as a plasmid) is much greater than depicted here. ase and methylase activities. Type I restriction endonu- cleases cleave DNA at random sites that can be more than 1,000 base pairs (bp) from the recognition se- quence. Type III restriction endonucleases cleave the DNA about 25 bp from the recognition sequence. Both types move along the DNA in a reaction that requires the energy of ATP. Type II restriction endonucleases, first isolated by Hamilton Smith in 1970, are simpler, re- quire no ATP, and cleave the DNA within the recogni- tion sequence itself. The extraordinary utility of this group of restriction endonucleases was demonstrated by Daniel Nathans, who first used them to develop novel methods for mapping and analyzing genes and genomes. Thousands of restriction endonucleases have been discovered in different bacterial species, and more than 100 different DNA sequences are recognized by one or more of these enzymes. The recognition sequences are usually 4 to 6 bp long and palindromic (see Fig. 8–20). Table 9–2 lists sequences recognized by a few type II restriction endonucleases. In some cases, the interac- tion between a restriction endonuclease and its target sequence has been elucidated in exquisite molecular de- tail; for example, Figure 9–2 shows the complex of the type II restriction endonuclease EcoRV and its target sequence. Some restriction endonucleases make staggered cuts on the two DNA strands, leaving two to four nu- cleotides of one strand unpaired at each resulting end. These unpaired strands are referred to as sticky ends (Fig. 9–3a), because they can base-pair with each other or with complementary sticky ends of other DNA frag- ments. Other restriction endonucleases cleave both strands of DNA at the opposing phosphodiester bonds, leaving no unpaired bases on the ends, often called blunt ends (Fig. 9–3b). The average size of the DNA fragments produced by cleaving genomic DNA with a restriction endonuclease depends on the frequency with which a particular re- striction site occurs in the DNA molecule; this in turn depends largely on the size of the recognition sequence. In a DNA molecule with a random sequence in which all four nucleotides were equally abundant, a 6 bp sequence recognized by a restriction endonuclease such as BamHI would occur on average once every 4 6 (4,096) bp, as- suming the DNA had a 50% GmC content. Enzymes that recognize a 4 bp sequence would produce smaller DNA fragments from a random-sequence DNA molecule; a recognition sequence of this size would be expected to occur about once every 4 4 (256) bp. In natural DNA mol- ecules, particular recognition sequences tend to occur less frequently than this because nucleotide sequences in DNA are not random and the four nucleotides are not equally abundant. In laboratory experiments, the aver- age size of the fragments produced by restriction en- donuclease cleavage of a large DNA can be increased by simply terminating the reaction before completion; the result is called a partial digest. Fragment size can also 8885d_c09_306-342 2/7/04 8:14 AM Page 308 mac76 mac76:385_reb: be increased by using a special class of endonucleases called homing endonucleases (see Fig. 26–34). These recognize and cleave much longer DNA sequences (14 to 20 bp). Once a DNA molecule has been cleaved into frag- ments, a particular fragment of known size can be en- riched by agarose or acrylamide gel electrophoresis or by HPLC (pp. 92, 90). For a typical mammalian genome, however, cleavage by a restriction endonuclease usually yields too many different DNA fragments to permit iso- lation of a particular fragment by electrophoresis or HPLC. A common intermediate step in the cloning of a specific gene or DNA segment is the construction of a DNA library (as described in Section 9.2). After the target DNA fragment is isolated, DNA lig- ase can be used to join it to a similarly digested cloning vector—that is, a vector digested by the same restric- tion endonuclease; a fragment generated by EcoRI, for example, generally will not link to a fragment generated by BamHI. As described in more detail in Chapter 25 (see Fig. 25–16), DNA ligase catalyzes the formation of new phosphodiester bonds in a reaction that uses ATP 9.1 DNA Cloning: The Basics 309 g * BamHI (5H11032) G G A T C C (3H11032) CCT AGG * h g * CIaI(5H11032) A T C G A T (3H11032) T A G C T A * h g * EcoRI (5H11032) G A A T T C (3H11032) C T T A A G * h g EcoRV (5H11032) G A T A T C (3H11032) C T A T A G h g * HaelII (5H11032) G G C C (3H11032) C C G G *h g HindIII (5H11032) A A G C T T (3H11032) T T C G A A h g Notl(5H11032) G C G G C C G C (3H11032) C G C C G G C G h * g Pstl(5H11032) C T G C A G (3H11032) G A C G T C h * g Pvull (5H11032) C A G C T G (3H11032) G T C G A C h g Tth111l (5H11032) G A C N N N G T C (3H11032) C T G N N N C A G h Recognition Sequences for Some Type II Restriction EndonucleasesTABLE 9–2 Arrows indicate the phosphodiester bonds cleaved by each restriction endonuclease. Asterisks indicate bases that are methylated by the corresponding methylase (where known). N denotes any base. Note that the name of each enzyme consists of a three-letter abbreviation (in italics) of the bacterial species from which it is derived, sometimes followed by a strain designation and Roman numerals to distinguish different restriction endonucleases isolated from the same bacterial species. Thus BamHI is the first (I) restriction endonuclease characterized from Bacillus amyloliquefaciens, strain H. FIGURE 9–2 Interaction of EcoRV restriction endonuclease with its target sequence. (a) The dimeric EcoRV endonuclease (its two subunits in light blue and gray) is bound to the products of DNA cleavage at the sequence recognized by the enzyme. The DNA backbone is shown in two shades of blue to distinguish the segments separated by cleavage (PDB ID 1RVC). (b) In this view, showing just the DNA, the DNA segment has been turned 180H11034. The enzyme creates blunt ends; the cleavage points appear staggered on the two DNA strands because the DNA is kinked. Bound magnesium ions (orange) play a role in catalysis of the cleavage reaction. Restriction Endonucleases (b) (a) 8885d_c09_306-342 2/7/04 8:14 AM Page 309 mac76 mac76:385_reb: Chapter 9 DNA-Based Information Technologies310 G G T G A A T T C A G C T T C G C A T T A G C A G C T G T A G C C C A Cleavage site Cleavage site Recognition sequences Chromosomal DNA G G T G C C A C T T A A EcoRI restriction endonuclease PvuII restriction endonuclease G A A T T C A G C T T C G C A T T A G C A G G A C A T C G C T G T A G C Sticky ends Blunt ends DNA ligase Plasmid cloning vector cleaved with EcoRI and PvuII (a) (b) CTTAAGTCGAA CGGTATCGTCGACATCG TCGAAGCGTAATC GTC A A T T C C T G C A G A A G C T T C C G G A T C C C C G G G G PstI Synthetic polylinker DNA ligase Plasmid cloning vector cleaved with EcoRI (c) HindIII BamHI SmaI GACGTCTTCGAAGGCCTAGGGGCCCTTAA C T T A A G A A T T CG C T T A A G A A T T CG A A T T C G T T A A C G P o ly link e r P s t I H i n d II I Ba m H I S m a I E c o R I FIGURE 9–3 Cleavage of DNA mole- cules by restriction endonucleases. Restriction endonucleases recognize and cleave only specific sequences, leaving either (a) sticky ends (with protruding single strands) or (b) blunt ends. Fragments can be ligated to other DNAs, such as the cleaved cloning vector (a plasmid) shown here. This reaction is facilitated by the annealing of complementary sticky ends. Ligation is less efficient for DNA fragments with blunt ends than for those with complementary sticky ends, and DNA fragments with different (noncomplementary) sticky ends generally are not ligated. (c) A synthetic DNA fragment with recognition sequences for several restriction endonucleases can be inserted into a plasmid that has been cleaved by a restriction endonuclease. The insert is called a linker; an insert with multiple restriction sites is called a polylinker. or a similar cofactor. The base-pairing of complemen- tary sticky ends greatly facilitates the ligation reaction (Fig. 9–3a). Blunt ends can also be ligated, albeit less efficiently. Researchers can create new DNA sequences by inserting synthetic DNA fragments (called linkers) between the ends that are being ligated. Inserted DNA fragments with multiple recognition sequences for re- striction endonucleases (often useful later as points for inserting additional DNA by cleavage and ligation) are called polylinkers (Fig. 9–3c). The effectiveness of sticky ends in selectively join- ing two DNA fragments was apparent in the earliest recombinant DNA experiments. Before restriction endo- nucleases were widely available, some workers found they could generate sticky ends by the combined action of the bacteriophage H9261 exonuclease and terminal trans- ferase (Table 9–1). The fragments to be joined were given complementary homopolymeric tails. Peter Lobban and Dale Kaiser used this method in 1971 in the first ex- periments to join naturally occurring DNA fragments. 8885d_c09_306-342 2/7/04 8:14 AM Page 310 mac76 mac76:385_reb: Similar methods were used soon after in the laboratory of Paul Berg to join DNA segments from simian virus 40 (SV40) to DNA derived from bacteriophage H9261, thereby creating the first recombinant DNA molecule with DNA segments from different species. Cloning Vectors Allow Amplification of Inserted DNA Segments The principles that govern the delivery of recombinant DNA in clonable form to a host cell, and its subsequent amplification in the host, are well illustrated by consid- ering three popular cloning vectors commonly used in experiments with E. coli—plasmids, bacteriophages, and bacterial artificial chromosomes—and a vector used to clone large DNA segments in yeast. Plasmids Plasmids are circular DNA molecules that replicate separately from the host chromosome. Natu- rally occurring bacterial plasmids range in size from 5,000 to 400,000 bp. They can be introduced into bac- terial cells by a process called transformation. The cells (generally E. coli) and plasmid DNA are incubated together at 0 H11034C in a calcium chloride solution, then sub- jected to a shock by rapidly shifting the temperature to 37 to 43 H11034C. For reasons not well understood, some of the cells treated in this way take up the plasmid DNA. Some species of bacteria are naturally competent for DNA uptake and do not require the calcium chloride treatment. In an alternative method, cells incubated with the plasmid DNA are subjected to a high-voltage pulse. This approach, called electroporation, tran- siently renders the bacterial membrane permeable to large molecules. Regardless of the approach, few cells actually take up the plasmid DNA, so a method is needed to select those that do. The usual strategy is to use a plasmid that includes a gene that the host cell requires for growth under specific conditions, such as a gene that confers resistance to an antibiotic. Only cells transformed by the recombinant plasmid can grow in the presence of that antibiotic, making any cell that contains the plasmid “se- lectable” under those growth conditions. Such a gene is called a selectable marker. Investigators have developed many different plas- mid vectors suitable for cloning by modifying naturally occurring plasmids. The E. coli plasmid pBR322 offers a good example of the features useful in a cloning vec- tor (Fig. 9–4): 1. pBR322 has an origin of replication, ori, a sequence where replication is initiated by cellular enzymes (Chapter 25). This sequence is required to propagate the plasmid and maintain it at a level of 10 to 20 copies per cell. 2. The plasmid contains two genes that confer resistance to different antibiotics (tet R , amp R ), allowing the identification of cells that contain the intact plasmid or a recombinant version of the plasmid (Fig. 9–5). 3. Several unique recognition sequences in pBR322 (PstI, EcoRI, BamHI, SalI, PvuII) are targets for different restriction endonucleases, providing sites where the plasmid can later be cut to insert for- eign DNA. 4. The small size of the plasmid (4,361 bp) facilitates its entry into cells and the biochemical manipula- tion of the DNA. Transformation of typical bacterial cells with purified DNA (never a very efficient process) becomes less suc- cessful as plasmid size increases, and it is difficult to clone DNA segments longer than about 15,000 bp when plasmids are used as the vector. Bacteriophages Bacteriophage H9261 has a very efficient mechanism for delivering its 48,502 bp of DNA into a bacterium, and it can be used as a vector to clone some- what larger DNA segments (Fig. 9–6). Two key features contribute to its utility: 1. About one-third of the H9261 genome is nonessential and can be replaced with foreign DNA. 2. DNA is packaged into infectious phage particles only if it is between 40,000 and 53,000 bp long, a constraint that can be used to ensure packaging of recombinant DNA only. 9.1 DNA Cloning: The Basics 311 Ampicillin resistance (amp R ) Tetracycline resistance (tet R ) Origin of replication (ori) PvuII SalI BamHI EcoRI PstI pBR322 (4,361bp) FIGURE 9–4 The constructed E. coli plasmid pBR322. Note the lo- cation of some important restriction sites—for PstI, EcoRI, BamHI, SalI, and PvuII; ampicillin- and tetracycline-resistance genes; and the repli- cation origin (ori). Constructed in 1977, this was one of the early plas- mids designed expressly for cloning in E. coli. 8885d_c09_306-342 2/7/04 8:14 AM Page 311 mac76 mac76:385_reb: Researchers have developed bacteriophage H9261 vec- tors that can be readily cleaved into three pieces, two of which contain essential genes but which together are only about 30,000 bp long. The third piece, “filler” DNA, is discarded when the vector is to be used for cloning, and additional DNA is inserted between the two essen- tial segments to generate ligated DNA molecules long enough to produce viable phage particles. In effect, the packaging mechanism selects for recombinant viral DNAs. Bacteriophage H9261 vectors permit the cloning of DNA fragments of up to 23,000 bp. Once the bacteriophage H9261 fragments are ligated to foreign DNA fragments of suit- able size, the resulting recombinant DNAs can be pack- PstI restriction endonuclease pBR322 plasmids amp R tet R Foreign DNA DNA ligase Agar containing tetracycline (control) Colonies with recombinant plasmids Host DNA transformation of E. coli cells 2 Foreign DNA is ligated to cleaved pBR322. Where ligation is successful, the ampicillin-resistance element is disrupted. The tetracycline-resistance element remains intact. 1 pBR322 is cleaved at the ampicillin- resistance element by PstI. 3 E. coli cells are transformed, then grown on agar plates containing tetracycline to select for those that have taken up plasmid. 4 Individual colonies are transferred to matching positions on additional plates. One plate contains tetracycline, the other tetracycline and ampicillin. 5 Cells that grow on tetracycline but not on tetracycline + ampicillin contain recombinant plasmids with disrupted ampicillin resistance, hence the foreign DNA. Cells with pBR322 without foreign DNA retain ampicillin resistance and grow on both plates. Agar containing tetracycline Agar containing ampicillin + tetracycline All colonies have plasmids selection of transformed cells colonies transferred for testing FIGURE 9–5 Use of pBR322 to clone and identify foreign DNA in E. coli. Plasmid Cloning restriction endonuclease Filler DNA (not needed for packaging) Lack essential DNA and/or are too small to be packagedRecombinant DNAs DNA ligase in vitro packaging λ bacteriophage containing foreign DNA Foreign DNA fragments FIGURE 9–6 Bacteriophage H9261 cloning vectors. Recombinant DNA methods are used to modify the bacteriophage H9261 genome, removing the genes not needed for phage production and replacing them with “filler” DNA to make the phage DNA large enough for packaging into phage particles. As shown here, the filler is replaced with foreign DNA in cloning experiments. Recombinants are packaged into viable phage particles in vitro only if they include an appropriately sized foreign DNA fragment as well as both of the essential H9261 DNA end fragments. 8885d_c09_306-342 2/7/04 8:14 AM Page 312 mac76 mac76:385_reb: 9.1 DNA Cloning: The Basics 313 FIGURE 9–7 (above right) Bacterial artificial chromosomes (BACs) as cloning vectors. The vector is a relatively simple plasmid, with a repli- cation origin (ori) that directs replication. The par genes, derived from a type of plasmid called an F plasmid, assist in the even distribution of plasmids to daughter cells at cell division. This increases the likeli- hood of each daughter cell carrying one copy of the plasmid, even when few copies are present. The low number of copies is useful in cloning large segments of DNA because it limits the opportunities for unwanted recombination reactions that can unpredictably alter large cloned DNAs over time. The BAC includes selectable markers. A lacZ gene (required for the production of the enzyme H9252-galactosidase) is sit- uated in the cloning region such that it is inactivated by cloned DNA inserts. Introduction of recombinant BACs into cells by electroporation is promoted by the use of cells with an altered (more porous) cell wall. Recombinant DNAs are screened for resistance to the antibiotic chlo- ramphenicol (Cm R ). Plates also contain a substrate for H9252-galactosidase that yields a colored product. Colonies with active H9252-galactosidase and hence no DNA insert in the BAC vector turn blue; colonies without H9252-galactosidase activity—and thus with the desired DNA inserts—are white. Cm R F plasmid par genes BAC vector ori Cloning sites (include lacZ) restriction endonuclease DNA ligase Large foreign DNA fragment with appropriate sticky ends Colonies with recombinant BACs are white. Agar containing chloramphenicol and substrate for b-galactosidase electroporation Recombinant BAC selection of chloramphenicol- resistant cells aged into phage particles by adding them to crude bac- terial cell extracts that contain all the proteins needed to assemble a complete phage. This is called in vitro packaging (Fig. 9–6). All viable phage particles will contain a foreign DNA fragment. The subsequent trans- mission of the recombinant DNA into E. coli cells is highly efficient. Bacterial Artificial Chromosomes (BACs) Bacterial artificial chromosomes are simply plasmids designed for the cloning of very long segments (typically 100,000 to 300,000 bp) of DNA (Fig. 9–7). They generally include selectable markers such as resistance to the antibiotic chloramphenicol (Cm R ), as well as a very stable origin of replication (ori) that maintains the plasmid at one or two copies per cell. DNA fragments of several hundred thousand base pairs are cloned into the BAC vector. The large circular DNAs are then introduced into host bac- teria by electroporation. These procedures use host bacteria with mutations that compromise the structure of their cell wall, permitting the uptake of the large DNA molecules. Yeast Artificial Chromosomes (YACs) E. coli cells are by no means the only hosts for genetic engineering. Yeasts are particularly convenient eukaryotic organisms for this work. As with E. coli, yeast genetics is a well-developed discipline. The genome of the most commonly used yeast, Saccharomyces cerevisiae, contains only 14 H11003 10 6 bp (a simple genome by eukaryotic standards, less than four times the size of the E. coli chromosome), and its entire sequence is known. Yeast is also very easy to maintain and grow on a large scale in the laboratory. Plasmid vectors have been constructed for yeast, em- ploying the same principles that govern the use of E. coli vectors described above. Convenient methods are now available for moving DNA into and out of yeast cells, facilitating the study of many aspects of eukaryotic cell biochemistry. Some recombinant plasmids incorporate multiple replication origins and other elements that al- low them to be used in more than one species (for ex- ample, yeast or E. coli). Plasmids that can be propa- gated in cells of two or more different species are called shuttle vectors. 8885d_c09_306-342 2/7/04 8:14 AM Page 313 mac76 mac76:385_reb: Research work with large genomes and the associ- ated need for high-capacity cloning vectors led to the development of yeast artificial chromosomes (YACS; Fig. 9–8). YAC vectors contain all the elements needed to maintain a eukaryotic chromosome in the yeast nucleus: a yeast origin of replication, two selec- table markers, and specialized sequences (derived from the telomeres and centromere, regions of the chromo- some discussed in Chapter 24) needed for stability and proper segregation of the chromosomes at cell division. Before being used in cloning, the vector is propagated as a circular bacterial plasmid. Cleavage with a restric- tion endonuclease (BamH1 in Fig. 9–8) removes a length of DNA between two telomere sequences (TEL), leaving the telomeres at the ends of the linearized DNA. Cleavage at another internal site (EcoRI in Fig. 9–8) di- vides the vector into two DNA segments, referred to as vector arms, each with a different selectable marker. The genomic DNA is prepared by partial digestion with restriction endonucleases (EcoRI in Fig. 9–8) to obtain a suitable fragment size. Genomic fragments are then separated by pulsed field gel electrophoresis, a variation of gel electrophoresis (see Fig. 3–19) that allows the separation of very large DNA segments. The DNA fragments of appropriate size (up to about 2 H11003 10 6 bp) are mixed with the prepared vector arms and ligated. The ligation mixture is then used to trans- form treated yeast cells with very large DNA molecules. Culture on a medium that requires the presence of both selectable marker genes ensures the growth of only those yeast cells that contain an artificial chromosome with a large insert sandwiched between the two vector arms (Fig. 9–8). The stability of YAC clones increases with size (up to a point). Those with inserts of more than 150,000 bp are nearly as stable as normal cellular chromosomes, whereas those with inserts of less than 100,000 bp are gradually lost during mitosis (so gener- ally there are no yeast cell clones carrying only the two vector ends ligated together or with only short inserts). YACs that lack a telomere at either end are rapidly degraded. Specific DNA Sequences Are Detectable by Hybridization DNA hybridization, a process outlined in Chapter 8 (see Fig. 8–32), is the most common sequence-based process for detecting a particular gene or segment of nucleic acid. There are many variations of the basic method, most making use of a labeled (such as radioactive) DNA or RNA fragment, known as a probe, complementary to the DNA being sought. In one classic approach to de- tect a particular DNA sequence within a DNA library (a collection of DNA clones), nitrocellulose paper is pressed onto an agar plate containing many individual bacterial colonies from the library, each colony with a different recombinant DNA. Some cells from each colony adhere to the paper, forming a replica of the plate. The paper is treated with alkali to disrupt the cells and denature the DNA within, which remains bound to the region of the paper around the colony from which it came. Added radioactive DNA probe anneals only to its complementary DNA. After any unannealed probe DNA is washed away, the hybridized DNA can be de- tected by autoradiography (Fig. 9–9). Chapter 9 DNA-Based Information Technologies314 TEL Xori EcoRI digestion creates two arms BamHI digestion creates linear chromosome with telomeric ends EcoRI EcoRI BamHIBamHI CEN ori CEN CEN ori Selectable marker X Selectable marker Y TEL TEL Y TEL TEL XY Right arm has selectable marker Y YAC Left arm has selectable marker X TEL Fragments of genomic DNA generated by light digestion with EcoRI Ligate Transform Enzymatic digestion of cell wall Yeast cell Yeast spheroplast Yeast with YAC clone Select for X and Y FIGURE 9–8 Construction of a yeast artificial chromosome (YAC). A YAC vector includes an origin of replication (ori), a centromere (CEN), two telomeres (TEL), and selectable markers (X and Y). Digestion with BamH1 and EcoRI generates two separate DNA arms, each with a telomeric end and one selectable marker. A large segment of DNA (e.g., up to 2 H11003 10 6 bp from the human genome) is ligated to the two arms to create a yeast artificial chromosome. The YAC transforms yeast cells (prepared by removal of the cell wall to form spheroplasts), and the cells are selected for X and Y; the surviving cells propagate the DNA insert. 8885d_c09_306-342 2/7/04 8:14 AM Page 314 mac76 mac76:385_reb: A common limiting step in detecting and cloning a gene is the generation of a complementary strand of nucleic acid to use as a probe. The origin of a probe de- pends on what is known about the gene under investi- gation. Sometimes a homologous gene cloned from another species makes a suitable probe. Or, if the pro- tein product of a gene has been purified, probes can be designed and synthesized by working backward from the amino acid sequence, deducing the DNA sequence that would code for it (Fig. 9–10). Now, researchers typically obtain the necessary DNA sequence information from sequence databases that detail the structure of millions of genes from a wide range of organisms. Expression of Cloned Genes Produces Large Quantities of Protein Frequently it is the product of the cloned gene, rather than the gene itself, that is of primary interest—partic- ularly when the protein has commercial, therapeutic, or research value. With an increased understanding of the fundamentals of DNA, RNA, and protein metabolism and their regulation in E. coli, investigators can now ma- nipulate cells to express cloned genes in order to study their protein products. Most eukaryotic genes lack the DNA sequence ele- ments—such as promoters, sequences that instruct RNA polymerase where to bind—required for their expression in E. coli cells, so bacterial regulatory sequences for transcription and translation must be inserted at ap- propriate positions relative to the eukaryotic gene in the vector DNA. (Promoters, regulatory sequences, and other aspects of the regulation of gene expression are discussed in Chapter 28.) In some cases cloned genes are so efficiently expressed that their protein product represents 10% or more of the cellular protein; they are said to be overexpressed. At these concentrations some foreign proteins can kill an E. coli cell, so gene expres- sion must be limited to the few hours before the planned harvest of the cells. Cloning vectors with the transcription and transla- tion signals needed for the regulated expression of a cloned gene are often called expression vectors. The rate of expression of the cloned gene is controlled by replacing the gene’s own promoter and regulatory se- quences with more efficient and convenient versions supplied by the vector. Generally, a well-characterized promoter and its regulatory elements are positioned near several unique restriction sites for cloning, so that genes inserted at the restriction sites will be expressed from the regulated promoter element (Fig. 9–11). Some of these vectors incorporate other features, such as a bacterial ribosome binding site to enhance translation of the mRNA derived from the gene, or a transcription termination sequence. Genes can similarly be cloned and expressed in eu- karyotic cells, with various species of yeast as the usual hosts. A eukaryotic host can sometimes promote post- translational modifications (changes in protein structure made after synthesis on the ribosomes) that might be required for the function of a cloned eukaryotic protein. 9.1 DNA Cloning: The Basics 315 Press nitrocellulose paper onto the agar plate. Some cells from each colony stick to the paper. Agar plate with transformed bacterial colonies Nitrocellulose paper DNA bound to paper Probe annealed to colonies of interest Treat with alkali to disrupt cells and expose denatured DNA. Incubate the paper with the radiolabeled probe, then wash. Radiolabeled DNA probe Expose x-ray film to paper. FIGURE 9–9 Use of hybridization to identify a clone with a par- ticular DNA segment. The radioactive DNA probe hybridizes to complementary DNA and is revealed by autoradiography. Once the labeled colonies have been identified, the corresponding colonies on the original agar plate can be used as a source of cloned DNA for further study. 8885d_c09_306-342 2/7/04 8:14 AM Page 315 mac76 mac76:385_reb: Alterations in Cloned Genes Produce Modified Proteins Cloning techniques can be used not only to overproduce proteins but to produce protein products subtly altered from their native forms. Specific amino acids may be re- placed individually by site-directed mutagenesis. This powerful approach to studying protein structure and function changes the amino acid sequence of a pro- tein by altering the DNA sequence of the cloned gene. If appropriate restriction sites flank the sequence to be altered, researchers can simply remove a DNA segment and replace it with a synthetic one that is identical to the original except for the desired change (Fig. 9–12a). When suitably located restriction sites are not present, an approach called oligonucleotide-directed muta- genesis (Fig. 9–12b) can create a specific DNA se- quence change. A short synthetic DNA strand with a specific base change is annealed to a single-stranded copy of the cloned gene within a suitable vector. The mismatch of a single base pair in 15 to 20 bp does not prevent annealing if it is done at an appropriate tem- perature. The annealed strand serves as a primer for the synthesis of a strand complementary to the plasmid vector. This slightly mismatched duplex recombinant plasmid is then used to transform bacteria, where the mismatch is repaired by cellular DNA repair enzymes (Chapter 25). About half of the repair events will re- move and replace the altered base and restore the gene to its original sequence; the other half will remove and Chapter 9 DNA-Based Information Technologies316 H 3 N H11001 Gly Leu Pro Trp Glu Asp Met Trp Phe Val Arg COO H11002 Known amino acid sequence (5H11032) G G APossible codons G G C G G U G G G U U G C C U U A C C C C C C C U C C G C C A U G G G A G G A A G A U G A C A U G U G G U U U U U C G U C G U U G U G G U A A G G C G A C G C A G A (3H11032) C G U C G GRegion of minimal degeneracy Synthetic probes 20 nucleotides long, 8 possible sequences U G G G A G A A U G U G G U U G U G A C U C U AU U U U C U G FIGURE 9–10 Probe to detect the gene for a protein of known amino acid sequence. Because more than one DNA sequence can code for any given amino acid sequence, the genetic code is said to be “de- generate.” (As described in Chapter 27, an amino acid is coded for by a set of three nucleotides called a codon. Most amino acids have two or more codons; see Fig. 27–7.) Thus the correct DNA sequence for a known amino acid sequence cannot be known in advance. The probe is designed to be complementary to a region of the gene with minimal degeneracy, that is, a region with the fewest possible codons for the amino acids—two codons at most in the example shown here. Oligonucleotides are synthesized with selectively randomized se- quences, so that they contain either of the two possible nucleotides at each position of potential degeneracy (shaded in pink). The oligonu- cleotide shown here represents a mixture of eight different sequences: one of the eight will complement the gene perfectly, and all eight will match at least 17 of the 20 positions. Bacterial promoter (P) and operator (O) sequences Ribosome binding site ori P O Gene encoding repressor that binds O and regulates P Polylinker with unique sites for several restriction endonucleases (i.e., cloning sites) Transcription termination sequence Selectable genetic marker (e.g., antibiotic resistance) FIGURE 9–11 DNA sequences in a typical E. coli expression vector. The gene to be expressed is inserted into one of the restriction sites in the polylinker, near the promoter (P), with the end encoding the amino terminus proximal to the promoter. The promoter allows effi- cient transcription of the inserted gene, and the transcription termi- nation sequence sometimes improves the amount and stability of the mRNA produced. The operator (O) permits regulation by means of a repressor that binds to it (Chapter 28). The ribosome binding site pro- vides sequence signals needed for efficient translation of the mRNA derived from the gene. The selectable marker allows the selection of cells containing the recombinant DNA. 8885d_c09_306-342 2/7/04 8:14 AM Page 316 mac76 mac76:385_reb: replace the normal base, retaining the desired muta- tion. Transformants are screened (often by sequencing their plasmid DNA) until a bacterial colony containing a plasmid with the altered sequence is found. Changes can also be introduced that involve more than one base pair. Large parts of a gene can be deleted by cutting out a segment with restriction endonucleases and ligating the remaining portions to form a smaller gene. Parts of two different genes can be ligated to cre- ate new combinations. The product of such a fused gene is called a fusion protein. Researchers now have ingenious methods to bring about virtually any genetic alteration in vitro. Reintro- duction of the altered DNA into the cell permits inves- tigation of the consequences of the alteration. Site- directed mutagenesis has greatly facilitated research on proteins by allowing investigators to make specific changes in the primary structure of a protein and to ex- amine the effects of these changes on the folding, three- dimensional structure, and activity of the protein. SUMMARY 9.1 DNA Cloning: The Basics ■ DNA cloning and genetic engineering involve the cleavage of DNA and assembly of DNA segments in new combinations—recombinant DNA. ■ Cloning entails cutting DNA into fragments with enzymes; selecting and possibly modifying a fragment of interest; inserting the DNA fragment into a suitable cloning vector; transferring the vector with the DNA insert into a host cell for replication; and identifying and selecting cells that contain the DNA fragment. ■ Key enzymes in gene cloning include restriction endonucleases (especially the type II enzymes) and DNA ligase. ■ Cloning vectors include plasmids, bacteriophages, and, for the longest DNA inserts, bacterial artificial chromosomes (BACs) and yeast artificial chromosomes (YACs). ■ Cells containing particular DNA sequences can be identified by DNA hybridization methods. ■ Genetic engineering techniques manipulate cells to express and/or alter cloned genes. 9.2 From Genes to Genomes The modern science of genomics now permits the study of DNA on a cellular scale, from individual genes to the entire genetic complement of an organism—its genome. Genomic databases are growing rapidly, as one se- quencing milestone is superseded by the next. Biology in the twenty-first century will move forward with the aid of informational resources undreamed of only a few years ago. We now turn to a consideration of some of the technologies fueling these advances. 9.2 From Genes to Genomes 317 Recombinant plasmid DNA Gene Synthetic DNA fragment with specific base- pair change restriction endonucleases DNA ligase Plasmid contains gene with desired base-pair change. (a) Single strand of recombinant plasmid DNA Gene DNA polymerase, dNTPs, DNA ligase In E. coli cells, about half the plasmids will have gene with desired base-pair change. (b) Oligonucleotide with sequence change transformation of E. coli cells; repair of DNA A G C A G C G C C G C C A G C T C G G G C C C G A G CG C C FIGURE 9–12 Two approaches to site-directed mutagenesis. (a) A synthetic DNA segment replaces a DNA fragment that has been re- moved by cleavage with a restriction endonuclease. (b) A synthetic oligonucleotide with a desired sequence change at one position is hy- bridized to a single-stranded copy of the gene to be altered. This acts as primer for synthesis of a duplex DNA (with one mismatch), which is then used to transform cells. Cellular DNA repair systems will con- vert about 50% of the mismatches to reflect the desired sequence change. 8885d_c09_306-342 2/7/04 8:14 AM Page 317 mac76 mac76:385_reb: DNA Libraries Provide Specialized Catalogs of Genetic Information A DNA library is a collection of DNA clones, gathered together as a source of DNA for sequencing, gene dis- covery, or gene function studies. The library can take a variety of forms, depending on the source of the DNA. Among the largest types of DNA library is a genomic library, produced when the complete genome of a par- ticular organism is cleaved into thousands of fragments, and all the fragments are cloned by insertion into a cloning vector. The first step in preparing a genomic library is par- tial digestion of the DNA by restriction endonucleases, such that any given sequence will appear in fragments of a range of sizes—a range that is compatible with the cloning vector and ensures that virtually all sequences are represented among the clones in the library. Frag- ments that are too large or too small for cloning are re- moved by centrifugation or electrophoresis. The cloning vector, such as a BAC or YAC plasmid, is cleaved with the same restriction endonuclease and ligated to the ge- nomic DNA fragments. The ligated DNA mixture is then used to transform bacterial or yeast cells to produce a library of cell types, each type harboring a different re- combinant DNA molecule. Ideally, all the DNA in the genome under study will be represented in the library. Each transformed bacterium or yeast cell grows into a colony, or “clone,” of identical cells, each cell bearing the same recombinant plasmid. Using hybridization methods, researchers can order individual clones in a library by identifying clones with overlapping sequences. A set of overlapping clones rep- resents a catalog for a long contiguous segment of a genome, often referred to as a contig (Fig. 9–13). Pre- viously studied sequences or entire genes can be located within the library using hybridization methods to de- termine which library clones harbor the known se- quence. If the sequence has already been mapped on a chromosome, investigators can determine the location (in the genome) of the cloned DNA and any contig of which it is a part. A well-characterized library may con- tain thousands of long contigs, all assigned to and or- dered on particular chromosomes to form a detailed physical map. The known sequences within the library (each called a sequence-tagged site, or STS) can pro- vide landmarks for genomic sequencing projects. As more and more genome sequences become avail- able, the utility of genomic libraries is diminishing and investigators are constructing more specialized libraries designed to study gene function. An example is a library that includes only those genes that are expressed—that is, are transcribed into RNA—in a given organism or even in certain cells or tissues. Such a library lacks the noncoding DNA that makes up a large portion of many eukaryotic genomes. The researcher first extracts mRNA from an organism or from specific cells of an or- ganism and then prepares complementary DNAs (cDNAs) from the RNA in a multistep reaction cat- alyzed by the enzyme reverse transcriptase (Fig. 9–14). The resulting double-stranded DNA fragments are then inserted into a suitable vector and cloned, creating a population of clones called a cDNA library. The search for a particular gene is made easier by focusing on a cDNA library generated from the mRNAs of a cell known to express that gene. For example, if we wished to clone globin genes, we could first generate a cDNA library from erythrocyte precursor cells, in which about half the mRNAs code for globins. To aid in the mapping of large genomes, cDNAs in a library can be partially sequenced at random to produce a useful type of STS called an ex- pressed sequence tag (EST). ESTs, ranging in size from a few dozen to several hundred base pairs, can be positioned within the larger genome map, providing markers for expressed genes. Hundreds of thousands of ESTs were included in the detailed physical maps used as a guide to sequencing the human genome. A cDNA library can be made even more specialized by cloning a cDNA or cDNA fragment into a vector that fuses the cDNA sequence with the sequence for a marker, or reporter gene; the fused genes form a “re- porter construct.” Two useful markers are the genes for green fluorescent protein and epitope tags. A target Chapter 9 DNA-Based Information Technologies318 A Segment of chromosome from organism X BAC clones 18 29 3 4 5 6 7 BC D E F G H I J K L M N O P Q – – –– – – FIGURE 9–13 Ordering of the clones in a DNA library. Shown here is a segment of a chromosome from a hypothetical organism X, with markers A through Q representing sequence-tagged sites (STSs—DNA segments of known sequence, including known genes). Below the chromosome is an array of ordered BAC clones, numbered 1 to 9. Or- dering the clones on the genetic map is a many-stage process. The presence or absence of an STS on an individual clone can be deter- mined by hybridization—for example, by probing each clone with PCR-amplified DNA from the STS. Once the STSs on each BAC clone are identified, the clones (and the STSs themselves, if their location is not yet known) can be ordered on the map. For example, compare clones 3, 4, and 5. Marker E (blue) is found on all three clones; F (red) on clones 4 and 5, but not on 3; and G (green) only on clone 5. This indicates that the order of the sites is E, F, G. The clones partially over- lap and their order must be 3, 4, 5. The resulting ordered series of clones is called a contig. 8885d_c09_306-342 2/7/04 8:14 AM Page 318 mac76 mac76:385_reb: gene fused with a gene for green fluorescent protein (GFP) generates a fusion protein that is highly fluo- rescent—it literally lights up (Fig. 9–15a). Just a few molecules of this protein can be observed microscopi- cally, allowing the study of its location and movements in a cell. An epitope tag is a short protein sequence that is bound tightly by a well-characterized monoclonal antibody (Chapter 5). The tagged protein can be specif- ically precipitated from a crude protein extract by in- teraction with the antibody (Fig. 9–15b). If any other proteins bind to the tagged protein, those will precipi- tate as well, providing information about protein-protein interactions in a cell. The diversity and utility of spe- cialized DNA libraries are growing every year. The Polymerase Chain Reaction Amplifies Specific DNA Sequences The Human Genome Project, along with the many as- sociated efforts to sequence the genomes of organisms of every type, is providing unprecedented access to gene sequence information. This in turn is simplifying the process of cloning individual genes for more detailed biochemical analysis. If we know the sequence of at least the flanking parts of a DNA segment to be cloned, we can hugely amplify the number of copies of that DNA segment, using the polymerase chain reaction (PCR), a process conceived by Kary Mullis in 1983. The amplified DNA can be cloned directly or used in a vari- ety of analytical procedures. The PCR procedure has an elegant simplicity. Two synthetic oligonucleotides are prepared, complementary to sequences on opposite strands of the target DNA at positions just beyond the ends of the segment to be am- plified. The oligonucleotides serve as replication primers that can be extended by DNA polymerase. The 3H11032 ends of the hybridized probes are oriented toward each other and positioned to prime DNA synthesis across the desired DNA segment (Fig. 9–16). (DNA polymerases 9.2 From Genes to Genomes 319 5H11032 A A A A A A A A mRNA 5H11032 5H11032 mRNA-DNA hybrid 3H11032 A A A A A A A A 3H11032 T T T T T T T T A A A A A A A A 3H11032 5H11032 Duplex DNA 3H11032 Reverse transcriptase and dNTPs yield a complementary DNA strand. mRNA is degraded with alkali. DNA polymerase I and dNTPs yield double-stranded DNA. mRNA template is annealed to synthetic oligonucleotide (oligo dT) primer. T T T T T T T T T T T T T T T T A A A A A A A A T T T T T T T T FIGURE 9–14 Construction of a cDNA library from mRNA. A cell’s mRNA includes transcripts from thousands of genes, and the cDNAs generated are correspondingly heterogeneous. The duplex DNA pro- duced by this method is inserted into an appropriate cloning vector. Reverse transcriptase can synthesize DNA on an RNA or a DNA tem- plate (see Fig. 26–29). FIGURE 9–15 Specialized DNA libraries. (a) Cloning of cDNA next to a gene for green fluorescent protein (GFP) creates a reporter con- struct. RNA transcription proceeds through the gene of interest (insert DNA) and the reporter gene, and the mRNA transcript is then ex- pressed as a fusion protein. The GFP part of the protein is visible in the fluorescence microscope. The photograph shows a nematode worm containing a GFP fusion protein expressed only in the four “touch” neurons that run the length of its body. Reporter Con- structs (b) If the cDNA is cloned next to a gene for an epitope tag, the resulting fusion protein can be precipitated by antibodies to the epitope. Any other proteins that interact with the tagged protein also precipitate, helping to elucidate protein-protein interactions. Transcription Insert cDNA Insert cDNA Size markers Pure tagged protein Precipitate tagged protein with specific antibody. Make cell extract. Express tagged protein in a cell. Precipitate Precipitate Separate precipitated proteins. Identify new proteins in precipitate (e.g., with mass spectrometry). GFP (a) (b) Epitope tag 8885d_c09_306-342 2/7/04 8:14 AM Page 319 mac76 mac76:385_reb: Chapter 9 DNA-Based Information Technologies320 (5H11032)GAATTC CTTAAG(5H11032) Heat to separate strands. Replication EcoRI endonuclease Anneal primers containing noncomplementary regions with cleavage site for restriction endonuclease. PCR (5H11032)GAATTC GAATTC(3H11032) CTTAAGCTTAAG AATTC G Clone by insertion at an EcoRI site in a cloning vector. CTTAAG (b) 2 1 CTTAAG(5H11032) (5H11032)GAATTC 1 Heat to separate strands. 2 Add synthetic oligo- nucleotide primers; cool. 3H11032 5H11032 Region of target DNA to be amplified 3 Add thermostable DNA polymerase to catalyze 5H11032 → 3H11032 DNA synthesis. 3H11032 3H11032 5H11032 5H11032 5H11032 5H11032 Repeat steps 1 and 2 . 3H11032 5H11032 DNA synthesis (step 3 ) is catalyzed by the thermostable DNA polymerase (still present). 3H11032 Repeat steps 1 through 3 . 5H11032 3H11032 5H11032 After 25 cycles, the target sequence has been amplified about 10 6 -fold. (a) 5H11032 5H11032 FIGURE 9–16 Amplification of a DNA segment by the polymerase chain reaction. (a) The PCR procedure has three steps. DNA strands are H220711 separated by heating, then H220712 annealed to an excess of short synthetic DNA primers (blue) that flank the region to be amplified; H220713 new DNA is synthesized by polymerization. The three steps are repeated for 25 or 30 cycles. The thermostable DNA polymerase TaqI (from Thermus aquaticus, a bacterial species that grows in hot springs) is not denatured by the heating steps. (b) DNA amplified by PCR can be cloned. The primers can include noncomplementary ends that have a site for cleavage by a restriction endonuclease. Although these parts of the primers do not anneal to the target DNA, the PCR process incorporates them into the DNA that is amplified. Cleavage of the amplified fragments at these sites creates sticky ends, used in ligation of the amplified DNA to a cloning vector. Polymerase Chain Reaction 8885d_c09_306-342 2/10/04 1:52 PM Page 320 mac34 mac34: kec_420: synthesize DNA strands from deoxyribonucleotides, using a DNA template, as described in Chapter 25.) Isolated DNA containing the segment to be amplified is heated briefly to denature it, and then cooled in the presence of a large excess of the synthetic oligonucleo- tide primers. The four deoxynucleoside triphosphates are then added, and the primed DNA segment is repli- cated selectively. The cycle of heating, cooling, and replication is repeated 25 or 30 times over a few hours in an automated process, amplifying the DNA segment flanked by the primers until it can be readily analyzed or cloned. PCR uses a heat-stable DNA polymerase, such as the Taq polymerase (derived from a bacterium that lives at 90 H11034C), which remains active after every heating step and does not have to be replenished. Careful de- sign of the primers used for PCR, such as including re- striction endonuclease cleavage sites, can facilitate the subsequent cloning of the amplified DNA (Fig. 9–16b). This technology is highly sensitive: PCR can detect and amplify as little as one DNA molecule in almost any type of sample. Although DNA degrades over time (p. 293), PCR has allowed successful cloning of DNA from samples more than 40,000 years old. Investigators have used the technique to clone DNA fragments from the mummified remains of humans and extinct animals such as the woolly mammoth, creating the new fields of mo- lecular archaeology and molecular paleontology. DNA from burial sites has been amplified by PCR and used to trace ancient human migrations. Epidemiologists can use PCR-enhanced DNA samples from human remains to trace the evolution of human pathogenic viruses. Thus, in addition to its usefulness for cloning DNA, PCR is a potent tool in forensic medicine (Box 9–1). It is also being used for detection of viral infections before they cause symptoms and for prenatal diagnosis of a wide ar- ray of genetic diseases. The PCR method is also important in advancing the goal of whole genome sequencing. For example, the mapping of expressed sequence tags to particular chro- mosomes often involves amplification of the EST by PCR, followed by hybridization of the amplified DNA to clones in an ordered library. Investigators found many other applications of PCR in the Human Genome Pro- ject, to which we now turn. Genome Sequences Provide the Ultimate Genetic Libraries The genome is the ultimate source of information about an organism, and there is no genome we are more in- terested in than our own. Less than 10 years after the development of practical DNA sequencing methods, se- rious discussions began about the prospects for se- quencing the entire 3 billion base pairs of the human genome. The international Human Genome Project got underway with substantial funding in the late 1980s. The effort eventually included significant contributions from 20 sequencing centers distributed among six nations: the United States, Great Britain, Japan, France, China, and Germany. General coordination was provided by the Office of Genome Research at the National Institutes of Health, led first by James Watson and after 1992 by Francis Collins. At the outset, the task of sequencing a 3 H11003 10 9 bp genome seemed to be a titanic job, but it gradually yielded to advances in technology. The com- pleted sequence of the human genome was published in April 2003, several years ahead of schedule. This advance was the product of a carefully planned international effort spanning 14 years. Research teams first generated a detailed physical map of the human genome, with clones derived from each chromosome or- ganized into a series of long contigs (Fig. 9–17). Each contig contained landmarks in the form of STSs at a dis- tance of every 100,000 bp or less. The genome thus mapped could be divided up between the international sequencing centers, each center sequencing the mapped BAC or YAC clones corresponding to its par- ticular segments of the genome. Because many of the 9.2 From Genes to Genomes 321 DNA is digested into fragments; fragments inserted into BACs. Genomic DNA Contigs are identified and mapped. BAC to be sequenced is fragmented; fragments sequenced at random. Sequence overlaps reveal final sequence. G G G C T A C A T G A T G G G C T A C A T G A T G G T C C A T G A T G G T C FIGURE 9–17 The Human Genome Project strategy. Clones isolated from a genomic library were ordered into a detailed physical map, then individual clones were sequenced by shotgun sequencing pro- tocols. The strategy used by the commercial sequencing effort elimi- nated the step of creating the physical map and sequenced the entire genome by shotgun cloning. 8885d_c09_306-342 2/7/04 8:14 AM Page 321 mac76 mac76:385_reb: clones were more than 100,000 bp long, and modern se- quencing techniques can resolve only 600 to 750 bp of sequence at a time, each clone had to be sequenced in pieces. The sequencing strategy used a shotgun ap- proach, in which researchers used powerful new auto- mated sequencers to sequence random segments of a given clone, then assembled the sequence of the entire clone by computerized identification of overlaps. The number of clone pieces sequenced was determined sta- tistically so that the entire length of the clone was se- quenced four to six times on average. The sequenced DNA was then made available in a database covering the entire genome. Construction of the physical map was a time-consuming task, and its progress was followed in annual reports in major journals throughout the 1990s— by the end of which the map was largely in place. Completion of the entire sequencing project was initially projected for the year 2005, but circumstances and tech- nology intervened to accelerate the process. A competing commercial effort to sequence the human genome was initiated by the newly established Celera Corporation in 1997. Led by J. Craig Venter, the Celera group made use of a different strategy called “whole genome shotgun sequencing,” which eliminates the step of assembling a physical map of the genome. Instead, teams sequenced DNA segments from through- Chapter 9 DNA-Based Information Technologies322 BOX 9–1 WORKING IN BIOCHEMISTRY A Potent Weapon in Forensic Medicine Traditionally, one of the most accurate methods for placing an individual at the scene of a crime has been a fingerprint. With the advent of recombinant DNA technology, a more powerful tool is now available: DNA fingerprinting (also called DNA typing or DNA profiling). DNA fingerprinting is based on sequence poly- morphisms, slight sequence differences (usually sin- gle base-pair changes) between individuals, 1 bp in every 1,000 bp, on average. Each difference from the prototype human genome sequence (the first one ob- tained) occurs in some fraction of the human popula- tion; every individual has some differences. Some of the sequence changes affect recognition sites for re- striction enzymes, resulting in variation in the size of DNA fragments produced by digestion with a partic- ular restriction enzyme. These variations are restric- tion fragment length polymorphisms (RFLPs). The detection of RFLPs relies on a specialized hybridization procedure called Southern blotting (Fig. 1). DNA fragments from digestion of genomic DNA by restriction endonucleases are separated by size electrophoretically, denatured by soaking the agarose gel in alkali, and then blotted onto a nylon membrane to reproduce the distribution of fragments in the gel. The membrane is immersed in a solution containing a radioactively labeled DNA probe. A probe for a sequence that is repeated several times in the human genome generally identifies a few of the thou- sands of DNA fragments generated when the human genome is digested with a restriction endonuclease. Autoradiography reveals the fragments to which the probe hybridizes, as in Figure 9–9. The genomic DNA sequences used in these tests are generally regions containing repetitive DNA (short sequences repeated thousands of times in tan- dem), which are common in the genomes of higher eukaryotes (see Fig. 24–8). The number of repeated units in these DNA regions varies among individuals (except between identical twins). With a suitable probe, the pattern of bands produced by DNA finger- printing is distinctive for each individual. Combining the use of several probes makes the test so selective that it can positively identify a single individual in the entire human population. However, the Southern blot procedure requires relatively fresh DNA samples and larger amounts of DNA than are generally present at a crime scene. RFLP analysis sensitivity is augmented by using PCR (see Fig. 9–16a) to amplify vanishingly small amounts of DNA. This allows investigators to obtain DNA fingerprints from a single hair follicle, a drop of blood, a small semen sample from a rape vic- tim, or samples that might be months or even many years old. These methods are proving decisive in court cases worldwide. In the example in Figure 1, the DNA from a semen sample obtained from a rape and murder vic- tim was compared with DNA samples from the victim and two suspects. Each sample was cleaved into frag- ments and separated by gel electrophoresis. Radioac- tive DNA probes were used to identify a small subset of fragments that contained sequences complemen- tary to the probe. The sizes of the identified fragments varied from one individual to the next, as seen here in the different patterns for the three individuals (vic- tim and two suspects) tested. One suspect’s DNA ex- hibits a banding pattern identical to that of a semen sample taken from the victim. This test used a single probe, but three or four different probes would be used (in separate experiments) to achieve an unam- biguous positive identification. 8885d_c09_306-342 2/7/04 8:14 AM Page 322 mac76 mac76:385_reb: out the genome at random. The sequenced segments were ordered by the computerized identification of sequence overlaps (with some reference to the public project’s detailed physical map). At the outset of the Human Genome Project, shotgun sequencing on this scale had been deemed impractical. However, advances in computer software and sequencing automation had made the approach feasible by 1997. The ensuing race between the private and public sequencing efforts substantially advanced the timeline for completion of the project. Publication of the draft human genome se- quence in 2001 was followed by two years of follow-up work to eliminate nearly a thousand discontinuities and 9.2 From Genes to Genomes 323 Such results have been used to both convict and acquit suspects and, in other cases, to establish pa- ternity with an extraordinary degree of certainty. The impact of these procedures on court cases will con- tinue to grow as societies agree on the standards and as formal methods become widely established in foren- sic laboratories. Even decades-old murder mysteries can be solved: in 1996, DNA fingerprinting helped to confirm the identification of the bones of the last Russ- ian czar and his family, who were assassinated in 1918. Expose x-ray film to membrane. Denature DNA, and transfer to nylon membrane. Separate fragments by agarose gel electrophoresis (unlabeled). Chromosomal DNA (e.g., Suspect 1) Cleave with restriction endonucleases. DNA fragments Radiolabeled DNA probe DNA markersDNA markers DNA markersDNA markers Incubate with probe, then wash. EvidenceVictimSuspect 2Suspect 1 EvidenceVictim Suspect 2Suspect 1 FIGURE 1 The Southern blot procedure, as applied to DNA fingerprinting. This procedure was named after Jeremy Southern, who developed the technique. Francis S. Collins J. Craig Venter 8885d_c09_306-342 2/7/04 8:14 AM Page 323 mac76 mac76:385_reb: to provide high-quality sequence data that are contigu- ous throughout the genome. The Human Genome Project marks the culmination of twentieth-century biology and promises a vastly changed scientific landscape for the new century. The human genome is only part of the story, as the genomes of many other species are also being (or have been) se- quenced, including the yeasts Saccharomyces cere- visiae (completed in 1996) and Schizosaccharomyces pombe (2002), the nematode Caenorhabditis elegans (1998), the fruit fly Drosophila melanogaster (2000), the plant Arabidopsis thaliana (2000), the mouse Mus musculus (2002), zebrafish, and dozens of bacterial and archaebacterial species (Fig. 9–18). Most of the early efforts have been focused on species commonly used in laboratories. However, genome sequencing is destined to branch out to many other species as experience grows and technology improves. Broad efforts to map genes, attempts to identify new proteins and disease genes, and many other initiatives are currently under way. The result is a database with the potential not only to fuel rapid advances in biology but to change the way that humans think about themselves. Early insights pro- vided by the human genome sequence range from the intriguing to the profound. We are not as complicated as we thought. Decades-old estimates that humans pos- sessed about 100,000 genes within the approximately 3.2 H11003 10 9 bp in the human genome have been sup- planted by the discovery that we have only 30,000 to 35,000 genes. This is perhaps three times more genes than a fruit fly (with 13,000) and twice as many as a nematode worm (18,000). Although humans evolved relatively recently, the human genome is very old. Of 1,278 protein families identified in one early screen, only 94 were unique to vertebrates. However, while we share many protein domain types with plants, worms, and flies, we use these domains in more complex arrange- ments. Alternative modes of gene expression (Chapter 26) allow the production of more than one protein from a single gene—a process that humans and other verte- brates engage in more than do bacteria, worms, or any other forms of life. This allows for greater complexity in the proteins generated from our gene complement. We now know that only 1.1% to 1.4% of our DNA actually encodes proteins (Fig. 9–19). More than 50% of our genome consists of short, repeated sequences, the vast majority of which—about 45% of our genome in all—come from transposons, short movable DNA se- quences that are molecular parasites (Chapter 25). Many of the transposons have been there a long time, now altered so that they can no longer move to new genomic locations. Others are still actively moving at low frequencies, helping to make the genome an ever- dynamic and evolving entity. At least a few transposons have been co-opted by their host and appear to serve useful cellular functions. What does all this information tell us about how much one human differs from another? Within the hu- man population are millions of single-base differences, called single nucleotide polymorphisms, or SNPs (pronounced “snips”). Each human differs from the next Chapter 9 DNA-Based Information Technologies324 1990 1991 1992 1993 1994 1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 Genome sequencing begins H. influenzae S. cerevisiae E. coli C. elegans D. melanogaster A. thaliana H. sapiens (draft) H. sapiens (completed) S. pombe M. musculus FIGURE 9–18 Genomic sequencing timeline. Discussions in the mid- 1980s led to initiation of the project in 1989. Preparatory work, in- cluding extensive mapping to provide genome landmarks, occupied much of the 1990s. Separate projects were launched to sequence the genomes of other organisms important to research. The first sequenc- ing efforts to be completed included many bacterial species (such as Haemophilus influenzae), yeast (S. cerevisiae), a nematode worm (C. elegans), the fruit fly (D. melanogaster), and a plant (A. thaliana). Completed sequences for mammalian genomes, including the human genome, began to emerge in 2000. Each genome project has a web- site that serves as a central repository for the latest data. FIGURE 9–19 Snapshot of the human genome. The chart shows the proportions of our genome made up of various types of sequences. Translated into protein 1.1%–1.4% Transposons 45% Other intergenic DNA 20.7% Large duplications 5% Transcribed into RNA as end product 25% Simple repeats (microsatellites) 3% 8885d_c09_306-342 2/7/04 8:14 AM Page 324 mac76 mac76:385_reb: by about 1 bp in every 1,000 bp. From these small ge- netic differences arises the human variety we are all aware of—differences in hair color, eyesight, allergies to medication, foot size, and even (to some unknown degree) behavior. Some of the SNPs are linked to par- ticular human populations and can provide important information about human migrations that occurred thousands of years ago and about our more distant evo- lutionary past. As spectacular as this advance is, the sequencing of the human genome is easy compared with what comes next—the effort to understand all the information in each genome. The genome sequences being added monthly to international databases are roadmaps, parts of which are written in a language we do not yet un- derstand. However, they have great utility in catalyzing the discovery of new proteins and processes affecting every aspect of biochemistry, as will become apparent in chapters to come. SUMMARY 9.2 From Genes to Genomes ■ The science of genomics broadly encompasses the study of genomes and their gene content. ■ Genomic DNA segments can be organized in libraries—such as genomic libraries and cDNA libraries—with a wide range of designs and purposes. ■ The polymerase chain reaction (PCR) can be used to amplify selected DNA segments from a DNA library or an entire genome. ■ In an international cooperative research effort, the genomes of many organisms, including that of humans, have been sequenced in their entirety and are now available in public databases. 9.3 From Genomes to Proteomes A gene is not simply a DNA sequence; it is information that is converted to a useful product—a protein or func- tional RNA molecule—when and if needed by the cell. The first and most obvious step in exploring a large se- quenced genome is to catalog the products of the genes within that genome. Genes that encode RNA as their fi- nal product are somewhat harder to identify than are protein-encoding genes, and even the latter can be very difficult to spot in a vertebrate genome. The explosion of DNA sequence information has also revealed a sober- ing truth. Despite many years of biochemical advances, there are still thousands of proteins in every eukaryotic cell (and quite a few in bacteria) that we know nothing about. These proteins may have functions in processes not yet discovered, or may contribute in unexpected ways to processes we think we understand. In addition, the genomic sequences tell us nothing about the three- dimensional structure of proteins or how proteins are modified after they are synthesized. The proteins, with their myriad critical functions in every cell, are now be- coming the focus of new strategies for whole cell bio- chemistry. The complement of proteins expressed by a genome is called its proteome, a term that first appeared in the research literature in 1995. This concept rapidly evolved into a separate field of investigation, called proteomics. The problem addressed by proteomics research is straightforward, although the solution is not. Each genome presents us with thousands of genes encoding proteins, and ideally we want to know the structure and function of all those proteins. Given that many proteins offer surprises even after years of study, the investiga- tion of an entire proteome is a daunting enterprise. Simply discovering the function of new proteins requires intensive work. Biochemists can now apply shortcuts in the form of a broad array of new and updated tech- nologies. Protein function can be described on three levels. Phenotypic function describes the effects of a protein on the entire organism. For example, the loss of the pro- tein may lead to slower growth of the organism, an al- tered development pattern, or even death. Cellular function is a description of the network of interactions engaged in by a protein at the cellular level. Interactions with other proteins in the cell can help define the kinds of metabolic processes in which the protein participates. Finally, molecular function refers to the precise bio- chemical activity of a protein, including details such as the reactions an enzyme catalyzes or the ligands a re- ceptor binds. For several genomes, such as those of the yeast Sac- charomyces cerevisiae and the plant Arabidopsis, a massive effort is underway to inactivate each gene by genetic engineering and to investigate the effect on the organism. If the growth patterns or other properties of the organism change (or if it does not grow at all), this provides information on the phenotypic function of the protein product of the gene. There are three other main paths to investigating protein function: (1) sequence and structural compar- isons with genes and proteins of known function, (2) determination of when and where a gene is expressed, and (3) investigation of the interactions of the protein with other proteins. We discuss each of these ap- proaches in turn. Sequence or Structural Relationships Provide Information on Protein Function One of the important reasons to sequence many genomes is to provide a database that can be used to assign gene functions by genome comparisons, an enterprise referred 9.3 From Genomes to Proteomes 325 8885d_c09_306-342 2/7/04 8:14 AM Page 325 mac76 mac76:385_reb: to as comparative genomics. Sometimes a newly dis- covered gene is related by sequence homologies to a gene previously studied in another or the same species, and its function can be entirely or partly defined by that relationship. Such genes—of different species but pos- sessing a clear sequence and functional relationship to each other—are called orthologs. Genes similarly re- lated to each other within a single species are called paralogs (see Fig. 1–37). If the function of a gene has been characterized for one species, this information can be used to assign gene function to the ortholog found in the second species. The identity is easiest to make when comparing genomes from relatively closely related species, such as mouse and human, although many clearly orthologous genes have been identified in species as distant as bacteria and humans. Sometimes even the order of genes on a chromosome is conserved over large segments of the genomes of closely related species (Fig. 9–20). Conserved gene order, called syn- teny, provides additional evidence for an orthologous relationship between genes at identical locations within the related segments. Alternatively, certain sequences associated with particular structural motifs (Chapter 4) may be identi- fied within a protein. The presence of a structural mo- tif may suggest that it, say, catalyzes ATP hydrolysis, binds to DNA, or forms a complex with zinc ions, help- ing to define molecular function. These relationships are determined with the aid of increasingly sophisticated computer programs, limited only by the current infor- mation on gene and protein structure and our capacity to associate sequences with particular structural motifs. To further the assignment of function based on structural relationships, a large-scale structural pro- teomics project has been initiated. The goal is to crys- tallize and determine the structure of as many proteins and protein domains as possible, in many cases with lit- tle or no existing information about protein function. The project has been assisted by the automation of some of the tedious steps of protein crystallization (see Box 4–4). As these structures are revealed, they will be made available in the structural databases described in Chap- ter 4. The effort should help define the extent of varia- tion in structural motifs. When a newly discovered pro- tein is found to have structural folds that are clearly related to motifs with known functions in the databases, this information can suggest a molecular function for the protein. Cellular Expression Patterns Can Reveal the Cellular Function of a Gene In every newly sequenced genome, researchers find genes that encode proteins with no evident structural relationships to known genes or proteins. In these cases, other approaches must be used to generate information about gene function. Determining which tissues a gene is expressed in, or what circumstances trigger the ap- pearance of the gene product, can provide valuable clues. Many different approaches have been developed to study these patterns. Two-Dimensional Gel Electrophoresis As shown in Figure 3–22, two-dimensional gel electrophoresis allows the separation and display of up to 1,000 different proteins on a single gel. Mass spectrometry (see Box 3–2) can then be used to partially sequence individual protein spots and assign each to a gene. The appearance and nonappearance (or disappearance) of particular protein spots in samples from different tissues, from similar tis- sues at different stages of development, or from tissues treated in ways that simulate a variety of biological con- ditions can help define cellular function. DNA Microarrays Major refinements of the technology underlying DNA libraries, PCR, and hybridization have come together in the development of DNA microar- rays (sometimes called DNA chips), which allow the rapid and simultaneous screening of many thousands of genes. DNA segments from known genes, a few dozen to hundreds of nucleotides long, are amplified by PCR and placed on a solid surface, using robotic devices that accurately deposit nanoliter quantities of DNA solution. Many thousands of such spots are deposited in a pre- designed array on a surface area of just a few square centimeters. An alternative strategy is to synthesize DNA directly on the solid surface, using photolithogra- phy (Fig. 9–21). Once the chip is constructed, it can be probed with mRNAs or cDNAs from a particular cell type Chapter 9 DNA-Based Information Technologies326 Human 9 Mouse 2 EPB72 PSMB7 DNM1 LMX1B CDK9 STXBP1 AK1 LCN2 Epb7.2 Psmb7 Dnm Lmx1b Cdk9 Stxbp1 Ak1 Lcn2 FIGURE 9–20 Synteny in the mouse and human genomes. Large seg- ments of the mouse and human genomes have closely related genes aligned in the same order on chromosomes, a relationship called syn- teny. This diagram shows segments of human chromosome 9 and mouse chromosome 2. The genes in these segments exhibit a very high degree of homology as well as the same gene order. The differ- ent lettering schemes for the gene names reflect different naming con- ventions in the two organisms. 8885d_c09_306-342 2/7/04 8:14 AM Page 326 mac76 mac76:385_reb: or cell culture to identify the genes being expressed in those cells. A microarray can answer such questions as which genes are expressed at a given stage in the development of an organism. The total complement of mRNA is iso- lated from cells at two different stages of development and converted to cDNA, using reverse transcriptase and fluorescently labeled deoxynucleotides. The fluorescent cDNAs are then mixed and used as probes, each hy- bridizing to complementary sequences on the microar- ray. In Figure 9–22, for example, the labeled nucleotides used to make the cDNA for each sample fluoresce in two different colors. The cDNA from the two samples is mixed and used to probe the microarray. Spots that fluoresce green represent mRNAs more abundant at the single-cell stage; those that fluoresce red represent sequences more abundant later in development. The mRNAs that are equally abundant at both stages of development fluoresce yellow. By using a mixture of two samples to measure relative rather than absolute abun- dance of sequences, the method corrects for variations in the amounts of DNA originally deposited in each spot on the grid and other possible inconsistencies among spots in the microarray. The spots that fluoresce pro- vide a snapshot of all the genes being expressed in the cells at the moment they were harvested—gene ex- pression examined on a genome-wide scale. For a gene of unknown function, the time and circumstances of its expression can provide important clues about its role in the cell. An example of this technique is illustrated in Fig- ure 9–23, showing the dramatic results this technique can produce. Segments from each of the more than 6,000 genes in the completely sequenced yeast genome were separately amplified by PCR, and each segment was deposited in a defined pattern to create the illus- trated microarray. In a sense, this array provides a snap- shot of the entire yeast genome. Protein Chips Proteins, too, can be immobilized on a solid surface and used to help define the presence or absence of other proteins in a sample. For example, re- searchers prepare an array of antibodies to particular proteins by immobilizing them as individual spots on a solid surface. A sample of proteins is added, and if the protein that binds any of the antibodies is present in the sample, it can be detected by a solid-state form of the ELISA assay (see Fig. 5–28). Many other types and ap- plications of protein chips are being developed. Detection of Protein-Protein Interactions Helps to Define Cellular and Molecular Function A key to defining the function of any protein is to de- termine what it binds to. In the case of protein-protein interactions, the association of a protein of unknown function with one whose function is known can provide a useful and compelling “guilt by association.” The tech- niques used in this effort are quite varied. Comparisons of Genome Composition Although not evi- dence of direct association, the mere presence of com- binations of genes in particular genomes can hint at 9.3 From Genomes to Proteomes 327 Light Light Solid surface Desired sequences Solution containing activated A (A*) G* solution C* solution Light Opaque screen over spots 2 and 4 C G T A C C T G G G T A G C C G 12 34 G * G * G * G * G * G * G * A A G G G A A C* C* C* C* C* C* C* A G A C G A * A * A * A * A * A * A * Opaque screen over spots 1, 2, and 3 Opaque screen over spots 1 and 3 Many more cycles FIGURE 9–21 Photolithography. This technique for preparing a DNA microarray makes use of nucleotide precursors that are activated by light, joining one nucleotide to the next in a photoreaction (as op- posed to the chemical process illustrated in Fig. 8–38). A computer is programmed with the oligonucleotide sequences to be synthesized at each point on a solid surface. The surface is washed successively with solutions containing one type of activated nucleotide (A*, G*, etc.). As in the chemical synthesis of DNA, the activated nucleotides are blocked so that only one can be added to a chain in each cycle. A screen covering the surface is opened over the areas programmed to receive a particular nucleotide, and a flash of light joins the nucleotide to the polymers in the uncovered areas. This continues until the re- quired sequences are built up on each spot on the surface. Many poly- mers with the same sequence are generated on each spot, not just the single polymer shown. Also, the surfaces have thousands of spots with different sequences (see Fig. 9–22); this array shows just four spots, to illustrate the strategy. 8885d_c09_306-342 2/7/04 8:14 AM Page 327 mac76 mac76:385_reb: protein function. We can simply search the genomic databases for particular genes, then determine what other genes are present in the same genomes (Fig. 9–24). When two genes always appear together in a genome, it suggests that the proteins they encode may be functionally related. Such correlations are most use- ful if the function of at least one of the proteins is known. Purification of Protein Complexes With the construction of cDNA libraries in which each gene is contiguous with (fused to) an epitope tag, workers can immunoprecipi- tate the protein product of a gene by using the antibody that binds to the epitope (Fig. 9–15b). If the tagged pro- tein is expressed in cells, other proteins that bind to it may also be precipitated with it. Identification of the associated proteins reveals some of the protein-protein interactions of the tagged protein. There are many vari- ations of this process. For example, a crude extract of cells that express a similarly tagged protein is added to a column containing immobilized antibody. The tagged protein binds to the antibody, and proteins that inter- act with the tagged protein are sometimes also retained Chapter 9 DNA-Based Information Technologies328 mRNA 1 Isolate mRNAs from cells at two stages of development; each mRNA sample represents all the genes expressed in the cells at that stage. cDNA DNA microarray reverse transcriptase 2 Convert mRNAs to cDNAs by reverse transcriptase, using fluorescently labeled deoxyribonucleotide triphosphates. 3 Add the cDNAs to a microarray; fluorescent cDNAs anneal to complementary sequences on the microarray. 4 Each fluorescent spot represents a gene expressed in the cells. Removal of unhybridized probe FIGURE 9–22 DNA microarray. A microarray can be prepared from any known DNA sequence, from any source, generated by chemical synthesis or by PCR. The DNA is positioned on a solid surface (usu- ally specially treated glass slides) with the aid of a robotic device ca- pable of depositing very small (nanoliter) drops in precise patterns. UV light cross-links the DNA to the glass slides. Once the DNA is at- tached to the surface, the microarray can be probed with other fluo- rescently labeled nucleic acids. Here, mRNA samples are collected from cells at two different stages in the development of a frog. The cDNA probes are made with nucleotides that fluoresce in different colors for each sample; a mixture of the cDNAs is used to probe the microarray. Green spots represent mRNAs more abundant at the single-cell stage; red spots, sequences more abundant later in devel- opment. The yellow spots indicate approximately equal abundance at both stages. Synthesizing an Oligonucleotide Array FIGURE 9–23 Enlarged image of a DNA microarray. Each glowing spot in this microarray contains DNA from one of the 6,200 genes of the yeast (S. cerevisiae) genome, with every gene represented in the array. The microarray has been probed with fluorescently labeled nu- cleic acid derived from the mRNAs obtained (1) when the cells were growing normally in culture and (2) five hours after the cells began to form spores. The green spots represent genes expressed at higher lev- els during normal growth; the red spots, genes expressed at higher levels during sporulation. The yellow spots represent genes that do not change their levels of expression during sporulation. This image is enlarged; the microarray actually measures only 1.8 H11003 1.8 cm. Screening Oligonucleotide Array for Patterns of Gene Expression 8885d_c09_306-342 2/7/04 8:14 AM Page 328 mac76 mac76:385_reb: on the column. The connection between the protein and the tag is cleaved with a specific protease, and the protein complexes are eluted from the column and analyzed. Researchers can use these methods to define complex networks of interactions within a cell. A variety of useful protein tags are available. A com- mon one is a histidine tag, often just a string of six His residues. A poly-His sequence binds quite tightly to metals such as nickel. If a protein is cloned so that its sequence is contiguous with a His tag, it will have the extra His residues at its carboxyl terminus. The protein can then be purified by chromatography on columns with immobilized nickel. These procedures are conven- ient but require caution, because the additional amino acid residues in an epitope or His tag can affect protein activity. Yeast Two-Hybrid Analysis A sophisticated genetic ap- proach to defining protein-protein interactions is based on the properties of the Gal4 protein (Gal4p), which ac- tivates transcription of certain genes in yeast (see Fig. 28–28). Gal4p has two domains, one that binds to a spe- cific DNA sequence and another that activates the RNA polymerase that synthesizes mRNA from an adjacent reporter gene. The domains are stable when separated, but activation of the RNA polymerase requires interac- tion with the activation domain, which in turn requires positioning by the DNA-binding domain. Hence, the do- mains must be brought together to function correctly (Fig. 9–25a). 9.3 From Genomes to Proteomes 329 Species Protein 1 2 3 4 P1 P2 P3 P4 P5 P6 P7 H11001 H11001 H11001 H11001H11001H11001 H11001 H11001H11001 H11001 H11001 H11001 H11001H11002 H11002H11002 H11002 H11001H11001 H11001H11002 H11002 H11002H11002 H11002H11002H11002 H11002 FIGURE 9–24 Use of comparative genomics to identify functionally related genes. One use of comparative genomics is to prepare phylo- genetic profiles in order to identify genes that always appear together in a genome. This example shows a comparison of genes from four organisms, but in practice, computer searches can look at dozens of species. The designations P1, P2, and so forth refer to proteins en- coded by each species. This technique does not require homologous proteins. In this example, because proteins P3 and P6 always appear together in a genome they may be functionally related. Further test- ing would be needed to confirm this inference. Reporter gene Gal4p DNA- binding domain Gal4p binding site Gal4p activation domain RNA polymerase Increased transcription Reporter gene (a) X Y Yeast strain 1 with Gal4p–binding domain fusions Yeast strain 2 with Gal4p–activation domain fusions Mate to produce diploid cells. Plate on medium requiring interaction of the binding and activation domains for cell survival. Survivors form colonies. Sequence fusion proteins to identify which proteins are interacting. (b) FIGURE 9–25 The yeast two-hybrid system. (a) In this system for de- tecting protein-protein interactions, the aim is to bring together the DNA-binding domain and the activation domain of the yeast Gal4 protein through the interaction of two proteins, X and Y, to which each domain is fused. This interaction is accompanied by the expression of a reporter gene. (b) The two fusions are created in separate yeast strains, which are then mated. The mated mixture is plated on a medium on which the yeast cannot survive unless the reporter gene is expressed. Thus, all surviving colonies have interacting protein fu- sion pairs. Sequencing of the fusion proteins in the survivors reveals which proteins are interacting. Yeast Two-Hybrid Systems 8885d_c09_306-342 2/7/04 8:14 AM Page 329 mac76 mac76:385_reb: In this method, the protein-coding regions of genes to be analyzed are fused to the coding sequences of ei- ther the DNA-binding domain or the activation domain of Gal4p, and the resulting genes express a series of fusion proteins. If a protein fused to the DNA-binding domain interacts with a protein fused to the activation domain, transcription is activated. The reporter gene transcribed by this activation is generally one that yields a protein required for growth, or is an enzyme that cat- alyzes a reaction with a colored product. Thus, when grown on the proper medium, cells that contain such a pair of interacting proteins are easily distinguished from those that do not. Typically, many genes are fused to the Gal4p DNA-binding domain gene in one yeast strain, and many other genes are fused to the Gal4p activation domain gene in another yeast strain, then the yeast strains are mated and individual diploid cells grown into colonies (Fig. 9–25b). This allows for large-scale screen- ing for proteins that interact in the cell. All these techniques provide important clues to pro- tein function. However, they do not replace classical biochemistry. They simply provide researchers with an expedited entrée into important new biological prob- lems. In the end, a detailed functional understanding of any new protein requires traditional biochemical analyses—such as were used for the many well-studied proteins described in this text. When paired with the si- multaneously evolving tools of biochemistry and molec- ular biology, genomics and proteomics are speeding the discovery not only of new proteins but of new biologi- cal processes and mechanisms. SUMMARY 9.3 From Genomes to Proteomes ■ A proteome is the complement of proteins produced by a cell’s genome. The new field of proteomics encompasses an effort to catalog and determine the functions of all the proteins in a cell. ■ One of the most effective ways to determine the function of a new gene is by comparative genomics, the search of databases for genes with similar sequences. Paralogs and orthologs are proteins (and their genes) with clear functional and sequence relationships in the same or in different species. In some cases, the presence of a gene in combination with certain other genes, observed as a pattern in several genomes, can point toward a possible function. ■ Cellular proteomes can be displayed by two- dimensional gel electrophoresis and explored with the aid of mass spectrometry. ■ The cellular function of a protein can sometimes be inferred by determining when and where its gene is expressed. Researchers use DNA microarrays (chips) and protein chips to explore gene expression at the cellular level. ■ Several new techniques, including comparative genomics, immunoprecipitation, and yeast two- hybrid analysis, can identify protein-protein interactions. These interactions provide important clues to protein function. 9.4 Genome Alterations and New Products of Biotechnology We don’t need to look far to find practical applications for the new biotechnologies or to find new opportunities for breakthroughs in basic research. Herein lie both the promise and the challenge of genomics. As our knowl- edge of the genome increases, we will improve our un- derstanding of every aspect of biological function. We will enhance our capacity to engineer organisms and pro- duce new pharmaceutical agents and, as a consequence, will improve human nutrition and health. This promise can be realized only if practical safeguards are in place to ensure responsible application of these techniques. A Bacterial Plant Parasite Aids Cloning in Plants We not only can understand genomes, we can change them. This is perhaps the ultimate manifestation of the new technologies. The introduction of recombinant DNA into plants has enormous implications for agricul- ture, making possible the alteration of the nutritional profile or yield of crops or their resistance to environ- mental stresses, such as insect pests, diseases, cold, salinity, and drought. Fertile plants of some species may be generated from a single transformed cell, so that an introduced gene passes to progeny through the seeds. As yet, researchers have not found any naturally oc- curring plant cell plasmids to facilitate cloning in plants, so the biggest technical challenge is getting DNA into plant cells. An important and adaptable ally in this ef- fort is the soil bacterium Agrobacterium tumefaciens. This bacterium can invade plants at the site of a wound, transform nearby cells, and induce them to form a tu- mor called a crown gall. Agrobacterium contains the large (200,000 bp) Ti plasmid (Fig. 9–26a). When the bacterium is in contact with a damaged plant cell, a 23,000 bp segment of the Ti plasmid called T DNA is transferred from the plasmid and integrated at a ran- dom position in one of the plant cell chromosomes (Fig. 9–26b). The transfer of T DNA from Agrobacterium to the plant cell chromosome depends on two 25 bp re- peats that flank the T DNA and on the products of the virulence (vir) genes on the Ti plasmid (Fig. 9–26a). The T DNA encodes enzymes that convert plant metabolites to two classes of compounds that benefit Chapter 9 DNA-Based Information Technologies330 8885d_c09_306-342 2/7/04 8:14 AM Page 330 mac76 mac76:385_reb: the bacterium (Fig. 9–27). The first group consists of plant growth hormones (auxins and cytokinins) that stimulate growth of the transformed plant cells to form the crown gall tumor. The second constitutes a series of unusual amino acids called opines, which serve as a food source for the bacterium. The opines are produced in high concentrations in the tumor cells and secreted to the surroundings, where they can be metabolized only by Agrobacterium, using enzymes encoded elsewhere on the Ti plasmid. The bacterium thereby diverts plant resources by converting them to a form that benefits only itself. 9.4 Genome Alterations and New Products of Biotechnology 331 vir genes T DNA 25 bp repeats (a) Ti plasmid (b) Wounded plant cell produces acetosyringone. Acetosyringone activates vir genes. Copy of T DNA is transferred and integrated into a plant chromosome. Agrobacterium cell Plant cell nucleus Plant cell synthesizes auxins, cytokinins, opines; tumor forms. FIGURE 9–26 Transfer of DNA to plant cells by a bacterial parasite. (a) The Ti (tumor-inducing) plasmid of Agrobacterium tumefaciens. (b) Wounded plant cells produce and release the phenolic compound acetosyringone. When Agrobacterium detects this compound, the virulence (vir) genes on the Ti plasmid are expressed. The vir genes encode enzymes needed to introduce the T DNA segment of the Ti plasmid into the genome of nearby plant cells. A single-stranded copy of the T DNA is synthesized and transferred to the plant cell, where it is converted to duplex DNA and integrated into a plant cell chro- mosome. The T DNA encodes enzymes that synthesize both plant growth hormones and opines (see Fig. 9–27); the latter compounds are metabolized (as a nutrient source) only by Agrobacterium. Ex- pression of the T DNA genes by transformed plant cells thus leads to both aberrant plant cell growth (tumor formation) and the diversion of plant cell nutrients to the invading bacteria. H CH 2 Opines C Auxins O G D P H G N N N Isopentenyl adenine (i 6 Ade) N H N HHOCH 2 CH 2 C O G D PC D H G CH 3 N N N N H N Indoleacetate H N C E CH 3 CH 2 CH 3 Zeatin Cytokinins COO H11002 A H 2 ) 2 Mannopine O O CHO COO H11002 HN H 2 CHO (C OCH 2 A C O H 2 N OOO B (CHOH) 4 D G N D H 2 H 2 N H11001 OO H11002 CH 3 O A H 2 ) 3 Octopine (CO CH C O HN H C A C OC O OO H11002 OO H11002 O C H11002 OOC C A C NH OC O OO H11002 O H 2 ) 3 (COONHOO H A NH (C CHH 2 ) 2 Nopaline G N D H 2 H 2 N H11001 FIGURE 9–27 Metabolites produced in Agrobacterium-infected plant cells. Auxins and cytokinins are plant growth hormones. The most common auxin, indoleacetate, is derived from tryptophan. Cytokinins are adenine derivatives. Opines generally are derived from amino acid precursors; at least 14 different opines are produced by enzymes en- coded by the Ti plasmids of different Agrobacterium species. 8885d_c09_306-342 2/7/04 8:14 AM Page 331 mac76 mac76:385_reb: This rare example of DNA transfer from a prokary- ote to a eukaryotic cell is a natural genetic engineering process—one that researchers can harness to transfer recombinant DNA (instead of T DNA) to the plant genome. A common cloning strategy employs an Agrobacterium with two different recombinant plas- mids. The first is a Ti plasmid from which the T DNA segment has been removed in the laboratory (Fig. 9–28a). The second is an Agrobacterium–E. coli shut- tle vector in which the 25 bp repeats of the T DNA flank a foreign gene that the researcher wants to introduce into the plant cell, along with a selectable marker such as resistance to the antibiotic kanamycin (Fig. 9–28b). The engineered Agrobacterium is used to infect a leaf, but crown galls are not formed because the T DNA genes for the auxin, cytokinin, and opine biosynthetic enzymes are absent from both plasmids. Instead, the vir gene products from the altered Ti plasmid direct the trans- formation of the plant cells by the foreign gene—the gene flanked by the T DNA 25 bp repeats in the second plasmid. The transformed plant cells can be selected by growth on agar plates that contain kanamycin, and ad- dition of growth hormones induces the formation of new plants that contain the foreign gene in every cell. The successful transfer of recombinant DNA into plants was vividly illustrated by an experiment in which the luciferase gene from fireflies was introduced into the cells of a tobacco plant (Fig. 9–29)—a favorite plant for transformation experiments because its cells are par- ticularly easy to transform with Agrobacterium. The potential of this technology is not limited to the pro- duction of glow-in-the-dark plants, of course. The same approach has been used to produce crop plants that are resistant to herbicides, plant viruses, and insect pests (Fig. 9–30). Potential benefits include increased yields and less need for environmentally harmful agricultural chemicals. Biotechnology can introduce new traits into a plant much faster than traditional methods of plant breeding. A prominent example is the development of soybeans that are resistant to the general herbicide glyphosate (the active ingredient in the product RoundUp). Glyphosate breaks down rapidly in the environment (glyphosate- sensitive plants can be planted in a treated area after as little as 48 hours), and its use does not generally lead to contamination of groundwater or carryover from one year to the next. A field of glyphosate-resistant soybeans can be treated once with glyphosate during a summer grow- ing season to eliminate essentially all weeds in the field, while leaving the soybeans unaffected (Fig. 9–31). Po- tential pitfalls of the technology, such as the evolution of glyphosate-resistant weeds or the escape of difficult- to-control recombinant plants, remain a concern of re- searchers and the public. Chapter 9 DNA-Based Information Technologies332 vir Agrobacterium cell Ti plasmid without T DNA (b) (a) Recombinant plasmid with foreign gene and kanamycin-resistance gene between T DNA 25 bp repeats 25 bp repeats Foreign gene Kanamycin resistance Bacteria invade at wound sites (where leaf is cut). Leaf segments are transferred to agar dish. Plants are regenerated from leaf segments. Agar plate with growth hormones and kanamycin These kanamycin- resistant plants contain the foreign gene. FIGURE 9–28 A two-plasmid strategy to create a recombinant plant. (a) One plasmid is a modified Ti plasmid that contains the vir genes but lacks T DNA. (b) The other plasmid contains a segment of DNA that bears both a foreign gene (the gene of interest, e.g., the gene for the insecticidal protein described in Fig. 9–30) and an antibiotic- resistance element (here, kanamycin resistance), flanked by the two 25 bp repeats of T DNA that are required for transfer of the plasmid genes to the plant chromosome. This plasmid also contains the repli- cation origin needed for propagation in Agrobacterium. When bacteria invade at the site of a wound (the edge of the cut leaf), the vir genes on the first plasmid mediate transfer into the plant genome of the segment of the second plasmid that is flanked by the 25 bp repeats. Leaf segments are placed on an agar dish that contains both kanamycin and appropriate levels of plant growth hormones, and new plants are generated from segments with the transformed cells. Nontransformed cells are killed by the kanamycin. The foreign gene and the antibiotic-resistance element are normally transferred together, so plant cells that grow in this medium generally contain the foreign gene. 8885d_c09_306-342 2/7/04 8:14 AM Page 332 mac76 mac76:385_reb: 9.4 Genome Alterations and New Products of Biotechnology 333 FIGURE 9–29 A tobacco plant expressing the gene for firefly luciferase. Light was produced after the plant was watered with a solution con- taining luciferin, the substrate for the light-producing luciferase enzyme (see Box 13–2). Don’t expect glow-in-the-dark ornamental plants at your local plant nursery anytime soon. The light is actually quite weak; this photograph required a 24-hour exposure. The real point—that this tech- nology allows the introduction of new traits into plants—is nevertheless elegantly made. FIGURE 9–30 Tomato plants engineered to be resistant to insect lar- vae. Two tomato plants were exposed to equal numbers of moth lar- vae. The plant on the left has not been genetically altered. The plant on the right expresses a gene for a protein toxin derived from the bac- terium Bacillus thuringiensis. This protein, introduced by a protocol similar to that depicted in Figure 9–28, is toxic to the larvae of some moth species while being harmless to humans and other organisms. Insect resistance has also been genetically engineered in cotton and other plants. FIGURE 9–31 Glyphosate-resistant soybean plants. The photographs show two areas of a soybean field in Wisconsin. (a) Without glyphosate treatment, this part of the field is overrun with weeds. (b) Glyphosate- resistant soybean plants thrive in the glyphosate-treated section of the field. Glyphosate breaks down rapidly in the environment. Agricul- tural use of engineered plants such as these proceeds only after con- siderable deliberation, balancing the extraordinary promise of the technology with the need to select new traits with care. Both science and society as a whole have a stake in ensuring that the use of the re- sultant plants has no adverse impact on the environment or on hu- man health. (a) (b) PNH H11002 O Glyphosate CH 2 CH 2 O O H11002 COO H11002 Manipulation of Animal Cell Genomes Provides Information on Chromosome Structure and Gene Expression The transformation of animal cells by foreign genetic material offers an important mechanism for expanding our knowledge of the structure and function of animal genomes, as well as for the generation of animals with new traits. This potential has stimulated intensive re- search into more-sophisticated means of cloning animals. Most work of this kind requires a source of cells into which DNA can be introduced. Although intact tissues are often difficult to maintain and manipulate in vitro, many types of animal cells can be isolated and grown in the laboratory if their growth requirements are carefully met. Cells derived from a particular animal tissue and 8885d_c09_306-342 2/7/04 8:14 AM Page 333 mac76 mac76:385_reb: grown under appropriate tissue culture conditions can maintain their differentiated properties (for example, a hepatocyte (liver cell) remains a hepatocyte) for weeks or even months. No suitable plasmidlike vector is available for intro- ducing DNA into an animal cell, so transformation usu- ally requires the integration of the DNA into a host-cell chromosome. The efficient delivery of DNA to a cell nu- cleus and integration of this DNA into a chromosome without disrupting any critical genes remain the major technical problems in the genetic engineering of animal cells. Available methods for carrying DNA into an animal cell vary in efficiency and convenience. Some success has been achieved with spontaneous uptake of DNA or electroporation, techniques roughly comparable to the common methods used to transform bacteria. They are inefficient in animal cells, however, transforming only 1 in 100 to 10,000 cells. Microinjection—the injection of DNA directly into a nucleus, using a very fine nee- dle—has a high success rate for skilled practitioners, but the total number of cells that can be treated is small, because each must be injected individually. The most efficient and widely used methods for transforming animal cells rely on liposomes or viral vec- tors. Liposomes are small vesicles consisting of a lipid bilayer that encloses an aqueous compartment (see Fig. 11–4). Liposomes that enclose a recombinant DNA mol- ecule can be fused with the membranes of target cells to deliver DNA into the cell. The DNA sometimes reaches the nucleus, where it can integrate into a chro- mosome (mostly at random locations). Viral vectors are even more efficient at delivering DNA. Animal viruses have effective mechanisms for introducing their nucleic acids into cells, and several types also have mechanisms to integrate their DNA into a host-cell chromosome. Some of these, such as retroviruses (see Fig. 26–30) and adenoviruses, have been modified to serve as viral vectors to introduce foreign DNA into mammalian cells. The work on retroviral vectors illustrates some of the strategies being used (Fig. 9–32). When an engi- neered retrovirus enters a cell, its RNA genome is tran- scribed to DNA by reverse transcriptase and then inte- grated into the host genome by the enzyme viral integrase. Special regions of DNA are required for this Chapter 9 DNA-Based Information Technologies334 Reverse transcriptase converts RNA genome to double-stranded DNA. LTR H9274 gag pol env LTR Retroviral genome (single-stranded RNA) Viral genes are replaced with a foreign gene. LTR H9274 gag pol env LTR DNA Recombinant DNA is introduced into cells in tissue culture. LTR H9274 LTR Recombinant defective retroviral DNA RNA copies of recombinant viruses are produced in cells containing a helper virus and packaged into viral particles. Reverse transcriptase and integrase Retroviral RNA genome with foreign gene Retroviral genome with foreign gene is integrated into the target cell chromosome. Recombinant virus particles infect a target cell. FIGURE 9–32 Use of retroviral vectors in mammalian cell cloning. A typical retroviral genome (somewhat simplified here), engineered to carry a foreign gene (pink), is added to a host-cell tissue culture. The helper virus (not shown) lacks the packaging sequence, H9274 , so its RNA transcripts cannot be packaged into viral particles, but it provides the gag, pol, and env gene products needed to package the engineered retrovirus into functional viral particles. This enables the foreign gene in the recombinant retroviral genome to be introduced efficiently into the target cells. procedure: long terminal repeat (LTR) sequences to in- tegrate retroviral DNA into the host chromosome and the H9274 (psi) sequence to package the viral RNA in viral particles (see Fig. 26–30). The gag, pol, and env genes of the retroviral genome, required for retroviral replication and assem- bly of viral particles, can be replaced with foreign DNA. To assemble viruses that contain the recombinant ge- netic information, researchers must introduce the DNA into cultured cells that are simultaneously infected with a “helper virus” that has the genes to produce viral par- ticles but lacks the H9274 sequence required for packaging. Thus the recombinant DNA can be transcribed and its 8885d_c09_306-342 2/7/04 8:14 AM Page 334 mac76 mac76:385_reb: RNA packaged into viral particles. These particles can act as vectors to introduce the recombinant RNA into target cells. Viral reverse transcriptase and integrase en- zymes (produced by the helper virus) are also packaged in the viral particle and introduced into the target cells. Once the engineered viral genome is inside a cell, these enzymes create a DNA copy of the recombinant viral RNA genome and integrate it into a host chromosome. The integrated recombinant DNA then becomes a per- manent part of the target cell’s chromosome and is repli- cated with the chromosome at every cell division. The cell itself is not endangered by the integrated viral DNA, because the recombinant virus lacks the genes needed to produce RNA copies of its genome and package them into new viral particles. The use of recombinant retro- viruses is often the best method for introducing DNA into large numbers of mammalian cells. Each type of virus has different attributes, so sev- eral classes of animal viruses are being engineered as vectors to transform mammalian cells. Adenoviruses, for example, lack a mechanism for integrating DNA into a chromosome. Recombinant DNA introduced via an ade- noviral vector is therefore expressed for only a short time and then destroyed. This can be useful if the ob- jective is transient expression of a gene. Transformation of animal cells by any of the above techniques has its problems. Introduced DNA is gener- ally integrated into chromosomes at random locations. Even when the foreign DNA contains a sequence simi- lar to a sequence in a host chromosome, allowing tar- geting to that position, nonhomologous integrants still outnumber the targeted ones by several orders of mag- nitude. If these integration events disrupt essential genes, they can sometimes alter cellular functions (most cells are diploid or polyploid, however, so an integration usually leaves at least one unaffected copy of any given gene). A particularly poor outcome would involve an in- tegration event that inadvertently activated a gene that stimulated cell division, potentially creating a cancer cell. Although such an event was once thought to be rare, recent trials suggest it is a significant hazard (Box 9–2). Finally, the site of an integration can determine the level of expression of the integrated gene, because integrants are not transcribed equally well everywhere in the genome. Despite these challenges, the transformation of ani- mal cells has been used extensively to study chromosome structure and the function, regulation, and expression of genes. The successful introduction of recombinant DNA into an animal can be illustrated by an experiment that permanently altered an easily observable inheritable physical trait. Microinjection of DNA into the nuclei of fertilized mouse eggs can produce efficient transforma- tion (chromosomal integration). When the injected eggs are introduced into a female mouse and allowed to de- velop, the new gene is often expressed in some of the newborn mice. Those in which the germ line has been 9.4 Genome Alterations and New Products of Biotechnology 335 FIGURE 9–33 Cloning in mice. The gene for human growth hormone was introduced into the genome of the mouse on the right. Expres- sion of the gene resulted in the unusually large size of this mouse. altered can be identified by testing their offspring. By careful breeding of these mice, researchers can establish a transgenic mouse line in which all the mice are ho- mozygous for the new gene or genes. This technology was used to introduce into mice the gene for human growth hormone, under the control of an inducible promoter. When the mice were fed a diet that included the inducer, some of the mice that developed from injected embryos grew to an unusually large size (Fig. 9–33). Transgenic mice have now been produced with a wide range of ge- netic variations, including many relevant to human dis- eases and their control, pointing the way to human gene therapy (Box 9–2). A very similar approach is used to generate mice in which a particular gene has been inac- tivated (“knockout mice”), a way of establishing the func- tion of the inactivated gene. Creating a Transgenic Mouse New Technologies Promise to Expedite the Discovery of New Pharmaceuticals It is difficult to summarize all the ways in which genomics and proteomics might affect the de- velopment of pharmaceutical agents, but a few exam- ples illustrate the potential. Hypertension, congestive heart failure, hypercholesterolemia, and obesity are treated by pharmaceutical drugs that alter human phys- iology. Therapies are arrived at by identifying an enzyme or receptor involved in the process and discovering an inhibitor that interferes with its action. Proteomics will play an increasing role in identifying such potential drug targets. For example, the most potent vasoconstrictor known is the peptide hormone urotensin II. First dis- covered in fish spinal fluid, urotensin II is a small cyclic peptide, with 11 amino acid residues in humans and 12 or 13 in some other organisms. The vasoconstriction it induces can cause or exacerbate hypertension, conges- tive heart failure, and coronary artery disease. Some of the methods described in Section 9.3 for elucidating 8885d_c09_306-342 2/7/04 8:14 AM Page 335 mac76 mac76:385_reb: protein-protein interactions have been used to demon- strate that urotensin II is bound by a G-protein-coupled receptor called GPR14. As we shall see in Chapter 12, G proteins play an important role in many signaling pathways. However, GPR14 was an “orphan” receptor, in that human genome sequencing had identified it as a G-protein-coupled receptor, but with no known func- tion. The association of urotensin II with GPR14 now makes the latter protein a key target for drug therapies aimed at interfering with the action of urotensin II. 336 BOX 9–2 BIOCHEMISTRY IN MEDICINE The Human Genome and Human Gene Therapy As biotechnology gained momentum in the 1980s, a rational approach to the treatment of genetic diseases became increasingly attractive. In principle, DNA can be introduced into human cells to correct inherited genetic deficiencies. Genetic correction may even be targeted to a specific tissue by inoculating an individ- ual with a genetically engineered, tissue-specific virus carrying a payload of DNA to be incorporated into de- ficient cells. The goal is entrancing, but the research path is strewn with impediments. Altering chromosomal DNA entails substantial risk—a risk that cannot be quantified in the early stages of discovery. Consequently, early efforts at human gene therapy were directed at only a small subset of genetic diseases. Panels of scientists and ethicists developed a list of several conditions that should be satisfied to justify the risk involved, includ- ing the following. (1) The genetic defect must be a well-characterized, single-gene disorder. (2) Both the mutant and the normal gene must be cloned and se- quenced. (3) In the absence of a technique for elimi- nating the existing mutant gene, the functional gene must function well in the presence of the mutant gene. (4) Finally, and most important, the risks inherent in a new technology must be outweighed by the seri- ousness of the disease. Protocols for human clinical trials were submitted by scientists in several nations and reviewed for scientific rigor and ethical compli- ance by carefully selected advisory panels in each country; then human trials commenced. Early targets of gene therapy included cancer and genetic diseases affecting the immune system. Immu- nity is mediated by leukocytes (white blood cells) of several different types, all arising from undifferenti- ated stem cells in the bone marrow. These cells divide quickly and have special metabolic requirements. Dif- ferentiation can become blocked in several ways, re- sulting in a condition called severe combined immune deficiency (SCID). One form of SCID results from ge- netically inherited defects in the gene encoding adenosine deaminase (ADA), an enzyme involved in nucleotide biosynthesis (discussed in Chapter 22). Another form of SCID arises from a defect in a cell- surface receptor protein that binds chemical signals called cytokines, which trigger differentiation. In both cases, the progenitor stem cells cannot differentiate into the mature immune system cells, such as T and B lymphocytes (Chapter 5). Children with these rare hu- man diseases are highly susceptible to bacterial and vi- ral infections, and often suffer from a range of related physiological and neurological problems. In the absence of an effective therapy, the children must be confined in a sterile environment. About 20% of these children have a human leukocyte antigen (HLA)–identical sib- ling who can serve as a bone marrow transplant donor, a procedure that can cure the disease. The remaining children need a different approach. The first human gene therapy trial was carried out at the National Institutes of Health in Bethesda, Mary- land, in 1990. The patient was a four-year-old girl crip- pled by ADA deficiency. Bone marrow cells from the child were transformed with an engineered retrovirus containing a functional ADA gene; when the alteration of cells is done in this way—in the laboratory rather than in the living patient—the procedure is said to be done ex vivo. The treated cells were reintroduced into the patient’s marrow. Four years later, the child was leading a normal life, going to school, and even testi- fying about her experiences before Congress. However, her recovery cannot be uniquely attributed to gene therapy. Before the gene therapy clinical trials began, researchers had developed a new treatment for ADA deficiency, in which synthetic ADA was administered in a complex with polyethylene glycol (PEG). For many ADA-SCID patients, injection of the ADA-PEG com- plex allowed some immune system development, with weight gain and reduced infection, although not full immune reconstitution. The new gene therapy was risky, and withdrawing the inoculation treatment from patients in the gene therapy trial was judged unethi- cal. So trial participants received both treatments at once, making it unclear which treatment was primarily responsible for the positive clinical outcome. Never- theless, the clinical trial provided important informa- tion: it was feasible to transfer genes ex vivo to large numbers of leukocytes, and cells bearing the trans- ferred gene were still detectable years after treatment, suggesting that long-term correction was possible. In addition, the risk associated with use of the retroviral vectors appeared to be low. Through the 1990s, hundreds of human gene ther- apy clinical trials were carried out, targeting a variety of genetic diseases, but the results in most cases were 8885d_c09_306-342 2/7/04 8:14 AM Page 336 mac76 mac76:385_reb: Glu–Thr–Pro–Asp–Cys–S–S–Cys–Val – – – – Phe Tyr Trp–Lys Urotensin II Another objective of medical research is to identify new agents that can treat the diseases caused by hu- man pathogens. This now means identifying enzymatic targets in microbial pathogens that can be inactivated with a new drug. The ideal microbial target enzyme 337 discouraging. One major impediment proved to be the inefficiency of introducing new genes into cells. Trans- formation failed in many cells, and the number of transformed cells often proved insufficient to reverse the disorder. In the ADA trials, achieving a sufficient population of transformed cells was particularly diffi- cult, because of the ongoing ADA-PEG therapy. Nor- mally, stem cells with the correct ADA gene would have a growth advantage over the untreated cells, ex- panding their population and gradually predominating in the bone marrow. However, the injections of ADA- PEG in the same patients allowed the untransformed (ADA-deficient) cells to live and develop, and the transformed cells did not have the needed growth ad- vantage to expand their population at the expense of the others. A gene therapy trial initiated in 1999 was success- ful in correcting a form of SCID caused by defective cy- tokine receptors (in particular a subunit called H9253c), as reported in 2000 by physician researchers in France, Italy, and Britain. These researchers introduced the corrected gene for the H9253c cytokine–receptor subunit into CD34 H11001 cells. (The stem cells that give rise to im- mune system cells have a protein called CD34 on their surface; these cells can be separated from other bone marrow cells by antibodies to CD34.) The transformed cells were placed back into the patients’ bone marrow. In this trial, introduction of the corrected gene clearly conferred a growth advantage over the untreated cells. A functioning immune system was detected in four of the first five patients within 6 to 12 weeks, and levels of mature immune system T lymphocytes reached the levels found in age-matched control subjects (who did not have SCID) within 6 to 8 months. Immune system function was restored, and nearly 4 years later (mid- 2003) most of the children are leading normal lives. Similar results have been obtained with four additional patients. This provided dramatic confirmation that human gene therapy could cure a serious genetic disease. In early 2003 came a setback. One of the original four patients who had received cells with the correct cytokine receptor gene developed a severe form of leukemia. During the gene therapy treatment, one of the introduced retroviruses had by chance inserted it- self into a chromosome of one CD34 H11001 cell, resulting in abnormally high expression of a gene called LMO- 2. The affected cell differentiated into an immune sys- tem T cell, and the elevated expression of LMO-2 led to uncontrolled growth of the cell, giving rise to the leukemia. As of mid-2003 the patient had responded well to chemotherapy, but there may be more chap- ters to write. The incident shows that early worries about the risk associated with retroviral vectors were well founded. After a review of the gene therapy trial protocols, including consultations with ethicists and parents of children affected by these diseases, further gene therapy trials are still planned for children who are not candidates for bone marrow transplants. The reason is simple enough. The potential benefit to the children with these debilitating conditions has been judged to outweigh the demonstrated risk. Human gene therapy is not limited to genetic dis- eases. Cancer cells are being targeted by delivering genes for proteins that might destroy the cell or restore the normal control of cell division. Immune system cells associated with tumors, called tumor- infiltrating lymphocytes, can be genetically modified to produce tumor necrosis factor (TNF; see Fig. 12–50). When these lymphocytes are taken from a cancer pa- tient, modified, and reintroduced, the engineered cells target the tumor, and the TNF they produce causes tu- mor shrinkage. AIDS may also be treatable with gene therapy; DNA that encodes an RNA molecule comple- mentary to a vital HIV mRNA could be introduced into immune system cells (the targets of HIV). The RNA transcribed from the introduced DNA would pair with the HIV mRNA, preventing its translation and inter- fering with the virus’s life cycle. Alternatively, a gene could be introduced that encodes an inactive form of one subunit of a multisubunit HIV enzyme; with one nonfunctional subunit, the entire enzyme might be in- activated. Our growing understanding of the human genome and the genetic basis for some diseases brings the promise of early diagnosis and constructive interven- tion. As the early results demonstrate, however, the road to effective therapies will be a long one, with many detours. We need to learn more about cellular metabolism, more about how genes interact, and more about how to manage the dangers. The prospect of vanquishing life-destroying genetic defects and other debilitating diseases provides the motivation to press on. 8885d_c09_306-342 2/7/04 8:14 AM Page 337 mac76 mac76:385_reb: should be (1) essential to the pathogen cell’s survival, (2) well-conserved among a wide range of pathogens, and (3) absent or significantly different in humans. The task of identifying metabolic processes that are critical to microorganisms but absent in humans is made much easier by comparative genomics, augmented by the functional information available from genomics and proteomics. ■ Recombinant DNA Technology Yields New Products and Challenges The products of recombinant DNA technology range from proteins to engineered organisms. The technology can produce large amounts of commercially useful pro- teins, can design microorganisms for special tasks, and can engineer plants or animals with traits that are use- ful in agriculture or medicine. Some products of this technology have been approved for consumer or profes- sional use, and many more are in development. Genetic engineering has been transformed over a few years from a promising new technology to a multibillion-dollar in- dustry, with much of the growth occurring in the phar- maceutical industry. Some major classes of new products are listed in Table 9–3. Erythropoietin is typical of the newer products. This protein hormone (M r 51,000) stimulates erythrocyte production. People with diseases that com- promise kidney function often have a deficiency of this protein, resulting in anemia. Erythropoietin produced by recombinant DNA technology can be used to treat these individuals, reducing the need for repeated blood transfusions. ■ Other applications of this technology continue to emerge. Enzymes produced by recombinant DNA tech- nology are already used in the production of detergents, sugars, and cheese. Engineered proteins are being used as food additives to supplement nutrition, flavor, and fragrance. Microorganisms are being engineered with al- tered or entirely novel metabolic pathways to extract oil and minerals from ground deposits, to digest oil spills, and to detoxify hazardous waste dumps and sewage. En- gineered plants with improved resistance to drought, frost, pests, and disease are increasing crop yields and reducing the need for agricultural chemicals. Complete animals can be cloned by moving an entire nucleus and all of its genetic material to a prepared egg from which the nucleus has been removed. The extraordinary promise of modern biotechnol- ogy does not come without controversy. The cloning of mammals challenges societal mores and may be accom- panied by serious deficiencies in the health and longevity of the cloned animal. If useful pharmaceutical agents can be produced, so can toxins suitable for bio- logical warfare. The potential for hazards posed by the release of engineered plants and other organisms into Chapter 9 DNA-Based Information Technologies338 TABLE 9–3 Some Recombinant DNA Products in Medicine Product category Examples/uses Anticoagulants Tissue plasminogen activator (TPA); activates plasmin, an enzyme involved in dissolving clots; effective in treating heart attack patients. Blood factors Factor VIII; promotes clotting; it is deficient in hemophiliacs; treatment with factor VIII produced by recombinant DNA technology eliminates infection risks associated with blood transfusions. Colony-stimulating factors Immune system growth factors that stimulate leukocyte production; treatment of immune deficiencies and infections. Erythropoietin Stimulates erythrocyte production; treatment of anemia in patients with kidney disease. Growth factors Stimulate differentiation and growth of various cell types; promote wound healing. Human growth hormone Treatment of dwarfism. Human insulin Treatment of diabetes. Interferons Interfere with viral reproduction; used to treat some cancers. Interleukins Activate and stimulate different classes of leukocytes; possible uses in treatment of wounds, HIV infection, cancer, and immune deficiencies. Monoclonal antibodies Extraordinary binding specificity is used in: diagnostic tests; targeted transport of drugs, toxins, or radioactive compounds to tumors as a cancer therapy; many other applications. Superoxide dismutase Prevents tissue damage from reactive oxygen species when tissues briefly deprived of O 2 during surgery suddenly have blood flow restored. Vaccines Proteins derived from viral coats are as effective in “priming” an immune system as is the killed virus more traditionally used for vaccines, and are safer; first developed was the vaccine for hepatitis B. 8885d_c09_306-342 2/7/04 8:14 AM Page 338 mac76 mac76:385_reb: the biosphere continues to be monitored carefully. The full range of the long-term consequences of this tech- nology for our species and for the global environment is impossible to foresee, but will certainly demand our increasing understanding of both cellular metabolism and ecology. SUMMARY 9.4 Genome Alterations and New Products of Biotechnology ■ Advances in whole genome sequencing and genetic engineering methods are enhancing our ability to modify genomes in all species. ■ Cloning in plants, which makes use of the Ti plasmid vector from Agrobacterium, allows the introduction of new plant traits. ■ In animal cloning, researchers introduce foreign DNA primarily with the use of viral vectors or microinjection. These techniques can produce transgenic animals and provide new methods for human gene therapy. ■ The use of genomics and proteomics in basic and pharmaceutical research is greatly advancing the discovery of new drugs. Biotechnology is also generating an ever-expanding range of other products and technologies. Chapter 9 Further Reading 339 Key Terms cloning 306 vector 307 recombinant DNA 307 restriction endonucleases 307 DNA ligase 307 plasmid 311 bacterial artificial chromosome (BAC) 313 yeast artificial chromosome (YAC) 314 site-directed mutagenesis 316 fusion protein 317 genomics 317 genomic library 318 contig 318 sequence-tagged site (STS) 318 complementary DNA (cDNA) 318 cDNA library 318 expressed sequence tag (EST) 318 epitope tag 319 polymerase chain reaction (PCR) 319 DNA fingerprinting 322 restriction fragment length polymorphisms (RFLPs) 322 Southern blot 322 single nucleotide polymorphisms (SNPs) 324 proteome 325 proteomics 325 orthologs 326 synteny 326 DNA microarray 326 Ti plasmid 330 transgenic 335 Terms in bold are defined in the glossary. Further Reading General Jackson, D.A., Symons, R.H., & Berg, P. (1972) Biochemical method for inserting new genetic information into DNA of simian virus 40: circular SV40 DNA molecules containing lambda phage genes and the galactose operon of Escherichia coli. Proc. Natl. Acad. Sci. USA 69, 2904–2909. The first recombinant DNA experiment linking DNA from two species. Lobban, P.E. & Kaiser, A.D. (1973) Enzymatic end-to-end joining of DNA molecules. J. Mol. Biol. 78, 453–471. Report of the first recombinant DNA experiment. Sambrook, J., Fritsch, E.F., & Maniatis, T. (1989) Molecular Cloning: A Laboratory Manual, 2nd edn, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY. Although supplanted by more recent manuals, this three- volume set includes much useful background information on the biological, chemical, and physical principles underlying both classic and still-current techniques. Gene Cloning Arnheim, N. & Erlich, H. (1992) Polymerase chain reaction strategy. Annu. Rev. Biochem. 61, 131–156. Hofreiter, M., Serre, D., Poinar, H.N., Kuch, M., & Paabo, S. (2001) Ancient DNA. Nat. Rev. Genet. 2, 353–359. Successes and pitfalls in the retrieval of DNA from very old samples. Ivanov, P.L., Wadhams, M.J., Roby, R.K., Holland, M.M., Weedn, V.W., & Parsons, T.J. (1996) Mitochondrial DNA sequence heteroplasmy in the Grand Duke of Russia Georgij Romanov establishes the authenticity of the remains of Tsar Nicholas II. Nat. Genet. 12, 417–420. Lindahl, T. (1997) Facts and artifacts of ancient DNA. Cell 90, 1–3. Good description of how nucleic acid chemistry affects the retrieval of DNA in archaeology. Genomics Adams, M.D., Kelley, J.M., Gocayne, J.D., Dubnick, M., Polymeropoulos, M.H., Xiao, H., Merril, C.R., Wu, A., Olde, B., Moreno, R.F., et al. (1991) Complementary DNA sequencing: expressed sequence tags and Human Genome Project. Science 252, 1651–1656. The paper that introduced expressed sequence tags (ESTs). Bamshad, M. & Wooding, S.P. (2003) Signatures of natural selection in the human genome. Nat. Rev. Genet. 4, 99A–111A. Use of the human genome to trace human evolution. Brenner, S. (2004) Genes to genomics. Annu. Rev. Genet. 38, in press. 8885d_c09_306-342 2/7/04 8:14 AM Page 339 mac76 mac76:385_reb: Chapter 9 DNA-Based Information Technologies340 Carroll, S.B. (2003) Genetics and the making of Homo sapiens. Nature 422, 849–857. Clark, M.S. (1999) Comparative genomics: the key to understand- ing the Human Genome Project. Bioessays 21, 121–130. Useful background on some reasons for the importance of sequencing the genomes of many organisms. Collins, F.S., Green, E.D., Guttmacher, A.E., & Guyer, M.S. (2003) A vision for the future of genomics research. Nature 422, 835–847. A wide-ranging overview of the enormous potential of genomics research. Lander, E.S., Linton, L.M., Birren, B., Nusbaum, C., Zody, M.C., Baldwin, J., Devon, K., Dewar, K., Doyle, M., FitzHugh, W., et al. (2001) Initial sequencing and analysis of the human genome. Nature 409, 860–921. Discussion of the draft genome sequence put together by the international Human Genome Project. Many other useful arti- cles are to be found in this issue. Venter, J.C., Adams, M.D., Myers, E.W., Li, P.W., Mural, R.J., Sutton, G.G., Smith, H.O., Yandell, M., Evans, C.A., Holt, R.A., et al. (2001) The sequence of the human genome. Science 291, 1304–1351. Description of the draft of the human genome sequence pro- duced by Celera Corporation. Many other articles in the same issue provide insight and additional information. Proteomics Brown, P.O. & Botstein, D. (1999) Exploring the new world of the genome with DNA microarrays. Nat. Genet. 21, 33–37. Eisenberg, D., Marcotte, E.M., Xenarios, I., & Yeates, T.O. (2000) Protein function in the post-genomic era. Nature 405, 823–826. Pandey, A. & Mann, M. (2000) Proteomics to study genes and genomes. Nature 405, 837–846. An especially good description of the various strategies and methods used to identify proteins and their functions. Zhu, H., Bilgin, M., & Snyder, M. (2003) Proteomics. Annu. Rev. Biochem. 72, 783–812. Applying Biotechnology Foster, E.A., Jobling, M.A., Taylor, P.G., Donnelly, P., de Knijff, P., Mieremet, R., Zerjal, T., & Tyler-Smith, C. (1999) The Thomas Jefferson paternity case. Nature 397, 32. Last article of a series in an interesting case study of the uses of biotechnology to address historical questions. Hansen, G. & Wright M.S. (1999) Recent advances in the trans- formation of plants. Trends Plant Sci. 4, 226–231. Koopman, P., Gubbay, J., Vivian, N., Goodfellow, P., & Lovell-Badge, R. (1991) Male development of chromosomally female mice transgenic for Sry. Nature 351, 117–121. Recombinant DNA technology shows that a single gene directs development of chromosomally female mice into males. Lapham, E.V., Kozma, C., & Weiss, J. (1996) Genetic discrimi- nation: perspectives of consumers. Science 274, 621–624. The upside and downside of knowing what is in your genome. Mahowald, M.B., Verp, M.S., & Anderson, R.R. (1998) Genetic counseling: clinical and ethical challenges. Annu. Rev. Genet. 32, 547–559. Ohlstein, E.H., Ruffolo, R.R., Jr., & Elliott, J.D. (2000) Drug discovery in the next millennium. Annu. Rev. Pharmacol. Toxi- col. 40, 177–191. Palmiter, R.D., Brinster, R.L., Hammer, R.E., Trumbauer, M.E., Rosenfeld, M.G., Birnberg, N.C., & Evans, R.M. (1982) Dramatic growth of mice that develop from eggs microinjected with metallothionein-growth hormone fusion genes. Nature 300, 611–615. A description of how to make giant mice. Pfeifer, A. & Verma, I. M. (2001) Gene therapy: promises and problems. Annu. Rev. Genomics Hum. Genet. 2, 177–211. Thompson, J. & Donkersloot, J.A. (1992) N-(Carboxyalkyl) amino acids: occurrence, synthesis, and functions. Annu. Rev. Biochem. 61, 517–557. A summary of the structure and biological functions of opines. Wadhwa, P.D., Zielske, S.P., Roth, J.C., Ballas, C.B., Bowman, J.E., & Gerson, S.L. (2002) Cancer gene therapy: scientific basis. Annu. Rev. Med. 53, 437–452. 1. Cloning When joining two or more DNA fragments, a researcher can adjust the sequence at the junction in a vari- ety of subtle ways, as seen in the following exercises. (a) Draw the structure of each end of a linear DNA frag- ment produced by an EcoRI restriction digest (include those sequences remaining from the EcoRI recognition sequence). (b) Draw the structure resulting from the reaction of this end sequence with DNA polymerase I and the four de- oxynucleoside triphosphates (see Fig. 8–36). (c) Draw the sequence produced at the junction that arises if two ends with the structure derived in (b) are lig- ated (see Fig. 25–16). (d) Draw the structure produced if the structure derived in (a) is treated with a nuclease that degrades only single- stranded DNA. (e) Draw the sequence of the junction produced if an end with structure (b) is ligated to an end with structure (d). (f) Draw the structure of the end of a linear DNA frag- ment that was produced by a PvuII restriction digest (include those sequences remaining from the PvuII recognition sequence). (g) Draw the sequence of the junction produced if an end with structure (b) is ligated to an end with structure (f). (h) Suppose you can synthesize a short duplex DNA fragment with any sequence you desire. With this synthetic fragment and the procedures described in (a) through (g), design a protocol that would remove an EcoRI restriction site from a DNA molecule and incorporate a new BamHI restric- tion site at approximately the same location. (See Fig. 9–3.) (i) Design four different short synthetic double- stranded DNA fragments that would permit ligation of struc- ture (a) with a DNA fragment produced by a PstI restriction Problems 8885d_c09_306-342 2/7/04 8:14 AM Page 340 mac76 mac76:385_reb: Chapter 9 Problems 341 digest. In one of these fragments, design the sequence so that the final junction contains the recognition sequences for both EcoRI and PstI. In the second and third fragments, design the sequence so that the junction contains only the EcoRI and only the PstI recognition sequence, respectively. Design the sequence of the fourth fragment so that neither the EcoRI nor the PstI sequence appears in the junction. 2. Selecting for Recombinant Plasmids When cloning a foreign DNA fragment into a plasmid, it is often useful to insert the fragment at a site that interrupts a selectable marker (such as the tetracycline-resistance gene of pBR322). The loss of function of the interrupted gene can be used to identify clones containing recombinant plasmids with foreign DNA. With a bacteriophage H9261 vector it is not necessary to do this, yet one can easily distinguish vectors that incorporate large foreign DNA fragments from those that do not. How are these recombinant vectors identified? 3. DNA Cloning The plasmid cloning vector pBR322 (see Fig. 9–4) is cleaved with the restriction endonuclease PstI. An isolated DNA fragment from a eukaryotic genome (also produced by PstI cleavage) is added to the prepared vector and ligated. The mixture of ligated DNAs is then used to trans- form bacteria, and plasmid-containing bacteria are selected by growth in the presence of tetracycline. (a) In addition to the desired recombinant plasmid, what other types of plasmids might be found among the trans- formed bacteria that are tetracycline resistant? How can the types be distinguished? (b) The cloned DNA fragment is 1,000 bp long and has an EcoRI site 250 bp from one end. Three different recom- binant plasmids are cleaved with EcoRI and analyzed by gel electrophoresis, giving the patterns shown. What does each pattern say about the cloned DNA? Note that in pBR322, the PstI and EcoRI restriction sites are about 750 bp apart. The entire plasmid with no cloned insert is 4,361 bp. Size mark- ers in lane 4 have the number of nucleotides noted. 4. Identifying the Gene for a Protein with a Known Amino Acid Sequence Using Figure 27–7 to translate the genetic code, design a DNA probe that would allow you to iden- tify the gene for a protein with the following amino-terminal amino acid sequence. The probe should be 18 to 20 nucleotides long, a size that provides adequate specificity if there is suffi- cient homology between the probe and the gene. H 3 N H11001 –Ala–Pro–Met–Thr–Trp–Tyr–Cys–Met– Asp–Trp–Ile–Ala–Gly–Gly–Pro–Trp–Phe–Arg– Lys–Asn–Thr–Lys– 5. Designing a Diagnostic Test for a Genetic Disease Huntington’s disease (HD) is an inherited neurodegenerative disorder, characterized by the gradual, irreversible impair- ment of psychological, motor, and cognitive functions. Symp- toms typically appear in middle age, but onset can occur at almost any age. The course of the disease can last 15 to 20 years. The molecular basis of the disease is becoming better understood. The genetic mutation underlying HD has been traced to a gene encoding a protein (M r 350,000) of unknown function. In individuals who will not develop HD, a region of the gene that encodes the amino terminus of the protein has a sequence of CAG codons (for glutamine) that is repeated 6 to 39 times in succession. In individuals with adult-onset HD, this codon is typically repeated 40 to 55 times. In indi- viduals with childhood-onset HD, this codon is repeated more than 70 times. The length of this simple trinucleotide repeat indicates whether an individual will develop HD, and at ap- proximately what age the first symptoms will occur. A small portion of the amino-terminal coding sequence of the 3,143-codon HD gene is given below. The nucleotide sequence of the DNA is shown in black, the amino acid se- quence corresponding to the gene is shown in blue, and the CAG repeat is shaded. Using Figure 27–7 to translate the ge- netic code, outline a PCR-based test for HD that could be car- ried out using a blood sample. Assume the PCR primer must be 25 nucleotides long. By convention, unless otherwise spec- ified a DNA sequence encoding a protein is displayed with the coding strand (the sequence identical to the mRNA tran- scribed from the gene) on top such that it is read 5H11032 to 3H11032, left to right. Source: The Huntington’s Disease Collaborative Research Group. (1993) A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. Cell 72, 971–983. 6. Using PCR to Detect Circular DNA Molecules In a species of ciliated protist, a segment of genomic DNA is sometimes deleted. The deletion is a genetically programmed reaction associated with cellular mating. A researcher pro- poses that the DNA is deleted in a type of recombination called site-specific recombination, with the DNA on either end of the segment joined together and the deleted DNA ending up as a circular DNA reaction product. proposed reaction ATGGCGACCCTGGAAAAGCTGATGAAGGCCTTCGAGTCCCTCAAGTCCTTC M 307 1 ATLEKLMKAFESLKS CAGCAGTTCCAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAGCAG Q 358 18 QFQQQQQQQQQQQQQQ F CAGCAGCAGCAGCAGCAGCAGCAACAGCCGCCACCGCCGCCGCCGCCGCCG Q 409 35 QQQQQQQQPPPPPP PP CCGCCTCCTCAGCTTCCTCAGCCGCCGCCG P 460 52 PPQLPQPPP 750 Electrophoresis 1234 3,000 1,000 250 Nucleotide length 500 1,500 5,000 8885d_c09_306-342 2/7/04 8:14 AM Page 341 mac76 mac76:385_reb: Chapter 9 DNA-Based Information Technologies342 Suggest how the researcher might use the polymerase chain reaction (PCR) to detect the presence of the circular form of the deleted DNA in an extract of the protist. 7. RFLP Analysis for Paternity Testing DNA finger- printing and RFLP analysis are often used to test for pater- nity. A child inherits chromosomes from both mother and father, so DNA from a child displays restriction fragments de- rived from each parent. In the gel shown here, which child, if any, can be excluded as being the biological offspring of the putative father? Explain your reasoning. Lane M is the sam- ple from the mother, F from the putative father, and C1, C2, and C3 from the children. 8. Mapping a Chromosome Segment A group of over- lapping clones, designated A through F, is isolated from one region of a chromosome. Each of the clones is separately cleaved by a restriction enzyme and the pieces resolved by agarose gel electrophoresis, with the results shown in the fig- ure below. There are nine different restriction fragments in this chromosomal region, with a subset appearing in each clone. Using this information, deduce the order of the re- striction fragments in the chromosome. 9. Cloning in Plants The strategy outlined in Figure 9–28 employs Agrobacterium cells that contain two separate plasmids. Suggest why the sequences on the two plasmids are not combined on one plasmid. 10. DNA Fingerprinting and RFLP Analysis DNA is ex- tracted from the blood cells of two different humans, indi- viduals 1 and 2. In separate experiments, the DNA from each individual is cleaved by restriction endonucleases A, B, and C, and the fragments separated by electrophoresis. A hypo- thetical map of a 10,000 bp segment of a human chromosome is shown (1 kbp H11005 1,000 bp). Individual 2 has point muta- tions that eliminate restriction recognition sites B* and C*. You probe the gel with a radioactive oligonucleotide comple- mentary to the indicated sequence and expose a piece of x- ray film to the gel. Indicate where you would expect to see bands on the film. The lanes of the gel are marked in the ac- companying diagram. 11. Use of Photolithography to Make a DNA Microar- ray Figure 9–21 shows the first steps in the process of mak- ing a DNA microarray, or DNA chip, using photolithography. Describe the remaining steps needed to obtain the desired sequences (a different four-nucleotide sequence on each of the four spots) shown in the first panel of the figure. After each step, give the resulting nucleotide sequence attached at each spot. 12. Cloning in Mammals The retroviral vectors described in Figure 9–32 make possible the efficient integration of for- eign DNA into a mammalian genome. Explain how these vec- tors, which lack genes for replication and viral packaging (gag, pol, env), are assembled into infectious viral particles. Suggest why it is important that these vectors lack the repli- cation and packaging genes. C * B probe B * AA 0kbp B 1 C 2 345678 C 910 M 12 12 12 M 12 12 12 10 9 8 7 6 5 4 3 2 1 A B C ABC Electrophoresis ABCDEF Nine restriction fragments 1 2 3 4 5 6 7 8 9 Overlapping clones Electrophoresis M F C1 C3C2 8885d_c09_306-342 2/7/04 8:14 AM Page 342 mac76 mac76:385_reb: chapter B iological lipids are a chemically diverse group of com- pounds, the common and defining feature of which is their insolubility in water. The biological functions of the lipids are as diverse as their chemistry. Fats and oils are the principal stored forms of energy in many or- ganisms. Phospholipids and sterols are major structural elements of biological membranes. Other lipids, al- though present in relatively small quantities, play cru- cial roles as enzyme cofactors, electron carriers, light- absorbing pigments, hydrophobic anchors for proteins, “chaperones” to help membrane proteins fold, emulsi- fying agents in the digestive tract, hormones, and intracellular messengers. This chapter introduces rep- resentative lipids of each type, with emphasis on their chemical structure and physical properties. We discuss the energy-yielding oxidation of lipids in Chapter 17 and their synthesis in Chapter 21. 10.1 Storage Lipids The fats and oils used almost universally as stored forms of energy in living organisms are derivatives of fatty acids. The fatty acids are hydrocarbon derivatives, at about the same low oxidation state (that is, as highly reduced) as the hydrocarbons in fossil fuels. The cellu- lar oxidation of fatty acids (to CO 2 and H 2 O), like the controlled, rapid burning of fossil fuels in internal com- bustion engines, is highly exergonic. We introduce here the structures and nomenclature of the fatty acids most commonly found in living or- ganisms. Two types of fatty acid–containing compounds, triacylglycerols and waxes, are described to illustrate the diversity of structure and physical properties in this family of compounds. Fatty Acids Are Hydrocarbon Derivatives Fatty acids are carboxylic acids with hydrocarbon chains ranging from 4 to 36 carbons long (C 4 to C 36 ). In some fatty acids, this chain is unbranched and fully saturated (contains no double bonds); in others the chain con- tains one or more double bonds (Table 10–1). A few contain three-carbon rings, hydroxyl groups, or methyl- group branches. A simplified nomenclature for these compounds specifies the chain length and number of double bonds, separated by a colon; for example, the 16-carbon saturated palmitic acid is abbreviated 16:0, and the 18-carbon oleic acid, with one double bond, is 18:1. The positions of any double bonds are specified by superscript numbers following H9004 (delta); a 20-carbon fatty acid with one double bond between C-9 and C-10 (C-1 being the carboxyl carbon) and another between C-12 and C-13 is designated 20:2(H9004 9,12 ). The most commonly occurring fatty acids have even numbers of carbon atoms in an unbranched chain of 12 to 24 carbons (Table 10–1). As we shall see in Chapter 21, the even number of carbons results from the mode of LIPIDS 10.1 Storage Lipids 343 10.2 Structural Lipids in Membranes 348 10.3 Lipids as Signals, Cofactors, and Pigments 357 10.4 Working with Lipids 363 The fatty substance, separated from the salifiable bases, was dissolved in boiling alcohol. On cooling, it was obtained crystallized and very pure, and in this state it was examined. As it has not been hitherto described . . . I purpose to call it margarine, from the Greek word signifying pearl, because one of its characters is to have the appearance of mother of pearl, which it communicates to several of the combinations of which it forms with the salifiable bases. —Michel-Eugène Chevreul, article in Philosophical Magazine, 1814 10 343 8885d_c10_343-368 1/12/04 1:06 PM Page 343 mac76 mac76:385_reb: synthesis of these compounds, which involves conden- sation of two-carbon (acetate) units. There is also a common pattern in the location of double bonds; in most monounsaturated fatty acids the double bond is between C-9 and C-10 (H9004 9 ), and the other double bonds of polyunsaturated fatty acids are generally H9004 12 and H9004 15 . (Arachidonic acid is an exception to this generalization.) The double bonds of polyunsaturated fatty acids are almost never con- jugated (alternating single and double bonds, as in OCHUCHOCHUCHO), but are separated by a meth- ylene group: OCHUCHOCH 2 OCHUCHO. In nearly all naturally occurring unsaturated fatty acids, the dou- ble bonds are in the cis configuration. Trans fatty acids are produced by fermentation in the rumen of dairy an- imals and are obtained from dairy products and meat. They are also produced during hydrogenation of fish or vegetable oils. Because diets high in trans fatty acids correlate with increased blood levels of LDL (bad cho- lesterol) and decreased HDL (good cholesterol), it is generally recommended that one avoid large amounts of these fatty acids. Unfortunately, French fries, dough- nuts, and cookies tend to be high in trans fatty acids. The physical properties of the fatty acids, and of compounds that contain them, are largely determined by the length and degree of unsaturation of the hydro- carbon chain. The nonpolar hydrocarbon chain accounts for the poor solubility of fatty acids in water. Lauric acid (12:0, M r 200), for example, has a solubility in water of 0.063 mg/g—much less than that of glucose (M r 180), which is 1,100 mg/g. The longer the fatty acyl chain and the fewer the double bonds, the lower is the solubility Chapter 10 Lipids344 TABLE 10–1 Some Naturally Occurring Fatty Acids: Structure, Properties, and Nomenclature Solubility at 30 H11034C Carbon Common name Melting (mg/g solvent) skeleton Structure* Systematic name ? (derivation) point (H11034C) Water Benzene 12:0 CH 3 (CH 2 ) 10 COOH n-Dodecanoic acid Lauric acid 44.2 0.063 2,600 (Latin laurus, “laurel plant”) 14:0 CH 3 (CH 2 ) 12 COOH n-Tetradecanoic acid Myristic acid 53.9 0.024 874 (Latin Myristica, nutmeg genus) 16:0 CH 3 (CH 2 ) 14 COOH n-Hexadecanoic acid Palmitic acid 63.1 0.0083 348 (Latin palma, “palm tree”) 18:0 CH 3 (CH 2 ) 16 COOH n-Octadecanoic acid Stearic acid 69.6 0.0034 124 (Greek stear, “hard fat”) 20:0 CH 3 (CH 2 ) 18 COOH n-Eicosanoic acid Arachidic acid 76.5 (Latin Arachis, legume genus) 24:0 CH 3 (CH 2 ) 22 COOH n-Tetracosanoic acid Lignoceric acid 86.0 (Latin lignum, “wood” H11001 cera, “wax“) 16:1(H9004 9 )CH 3 (CH 2 ) 5 CHUCH(CH 2 ) 7 COOH cis-9-Hexadecenoic acid Palmitoleic acid 1–0.5 18:1(H9004 9 )CH 3 (CH 2 ) 7 CHUCH(CH 2 ) 7 COOH cis-9-Octadecenoic acid Oleic acid 13.4 (Latin oleum, “oil”) 18:2(H9004 9,12 )CH 3 (CH 2 ) 4 CHUCHCH 2 CHU cis-,cis-9,12-Octadecadienoic Linoleic acid 1–5 CH(CH 2 ) 7 COOH acid (Greek linon, “flax”) 18:3(H9004 9,12,15 )CH 3 CH 2 CHUCHCH 2 CHU cis-,cis-,cis-9,12,15- H9251-Linolenic acid H1100211 CHCH 2 CHUCH(CH 2 ) 7 COOH Octadecatrienoic acid 20:4(H9004 5,8,11,14 )CH 3 (CH 2 ) 4 CHUCHCH 2 CHU cis-,cis-,cis-,cis-5,8,11,14- Arachidonic acid H1100249.5 CHCH 2 CHUCHCH 2 CHU Icosatetraenoic acid CH(CH 2 ) 3 COOH *All acids are shown in their nonionized form. At pH 7, all free fatty acids have an ionized carboxylate. Note that numbering of carbon atoms begins at the carboxyl carbon. ? The prefix n- indicates the “normal” unbranched structure. For instance, “dodecanoic” simply indicates 12 carbon atoms, which could be arranged in a variety of branched forms; “n-dodecanoic” specifies the linear, unbranched form. For unsaturated fatty acids, the configuration of each double bond is indicated; in biological fatty acids the configuration is almost always cis. 8885d_c10_343-368 1/12/04 1:06 PM Page 344 mac76 mac76:385_reb: in water. The carboxylic acid group is polar (and ion- ized at neutral pH) and accounts for the slight solubil- ity of short-chain fatty acids in water. Melting points are also strongly influenced by the length and degree of unsaturation of the hydrocarbon chain. At room temperature (25 H11034C), the saturated fatty acids from 12:0 to 24:0 have a waxy consistency, whereas unsaturated fatty acids of these lengths are oily liquids. This difference in melting points is due to different de- grees of packing of the fatty acid molecules (Fig. 10–1). In the fully saturated compounds, free rotation around each carbon–carbon bond gives the hydrocarbon chain great flexibility; the most stable conformation is the fully extended form, in which the steric hindrance of neigh- boring atoms is minimized. These molecules can pack to- gether tightly in nearly crystalline arrays, with atoms all along their lengths in van der Waals contact with the atoms of neighboring molecules. In unsaturated fatty acids, a cis double bond forces a kink in the hydrocar- bon chain. Fatty acids with one or several such kinks cannot pack together as tightly as fully saturated fatty acids, and their interactions with each other are there- fore weaker. Because it takes less thermal energy to disorder these poorly ordered arrays of unsaturated fatty acids, they have markedly lower melting points than saturated fatty acids of the same chain length (Table 10–1). In vertebrates, free fatty acids (unesterified fatty acids, with a free carboxylate group) circulate in the blood bound noncovalently to a protein carrier, serum albumin. However, fatty acids are present in blood plasma mostly as carboxylic acid derivatives such as es- ters or amides. Lacking the charged carboxylate group, these fatty acid derivatives are generally even less sol- uble in water than are the free fatty acids. Triacylglycerols Are Fatty Acid Esters of Glycerol The simplest lipids constructed from fatty acids are the triacylglycerols, also referred to as triglycerides, fats, or neutral fats. Triacylglycerols are composed of three fatty acids each in ester linkage with a single glycerol (Fig. 10–2). Those containing the same kind of fatty acid 10.1 Storage Lipids 345 (a) Carboxyl Hydrocarbon group chain C H11002 OO (b) C H11002 O O Saturated fatty acids (c) (d) Mixture of saturated and unsaturated fatty acids FIGURE 10–1 The packing of fatty acids into stable aggregates. The extent of packing depends on the degree of saturation. (a) Two rep- resentations of the fully saturated acid stearic acid (stearate at pH 7) in its usual extended conformation. Each line segment of the zigzag represents a single bond between adjacent carbons. (b) The cis dou- ble bond (shaded) in oleic acid (oleate) does not permit rotation and introduces a rigid bend in the hydrocarbon tail. All other bonds in the chain are free to rotate. (c) Fully saturated fatty acids in the extended form pack into nearly crystalline arrays, stabilized by many hydro- phobic interactions. (d) The presence of one or more cis double bonds interferes with this tight packing and results in less stable aggregates. C O O CH 2 13 2 O C O H CH 2 O C O 1-Stearoyl, 2-linoleoyl, 3-palmitoyl glycerol, a mixed triacylglycerol Glycerol HO CH 2 OH H CH 2 OHC C FIGURE 10–2 Glycerol and a triacylglycerol. The mixed triacylglyc- erol shown here has three different fatty acids attached to the glyc- erol backbone. When glycerol has two different fatty acids at C-1 and C-3, the C-2 is a chiral center (p. 76). 8885d_c10_343-368 1/12/04 1:06 PM Page 345 mac76 mac76:385_reb: in all three positions are called simple triacylglycerols and are named after the fatty acid they contain. Simple triacylglycerols of 16:0, 18:0, and 18:1, for example, are tristearin, tripalmitin, and triolein, respectively. Most naturally occurring triacylglycerols are mixed; they con- tain two or more different fatty acids. To name these compounds unambiguously, the name and position of each fatty acid must be specified. Because the polar hydroxyls of glycerol and the polar carboxylates of the fatty acids are bound in ester linkages, triacylglycerols are nonpolar, hydrophobic mol- ecules, essentially insoluble in water. Lipids have lower specific gravities than water, which explains why mix- tures of oil and water (oil-and-vinegar salad dressing, for example) have two phases: oil, with the lower specific gravity, floats on the aqueous phase. Triacylglycerols Provide Stored Energy and Insulation In most eukaryotic cells, triacylglycerols form a sepa- rate phase of microscopic, oily droplets in the aqueous cytosol, serving as depots of metabolic fuel. In verte- brates, specialized cells called adipocytes, or fat cells, store large amounts of triacylglycerols as fat droplets that nearly fill the cell (Fig. 10–3a). Triacylglycerols are also stored as oils in the seeds of many types of plants, providing energy and biosynthetic precursors during seed germination (Fig. 10–3b). Adipocytes and germi- nating seeds contain lipases, enzymes that catalyze the hydrolysis of stored triacylglycerols, releasing fatty acids for export to sites where they are required as fuel. There are two significant advantages to using tria- cylglycerols as stored fuels, rather than polysaccharides such as glycogen and starch. First, because the carbon atoms of fatty acids are more reduced than those of sug- ars, oxidation of triacylglycerols yields more than twice as much energy, gram for gram, as the oxidation of car- bohydrates. Second, because triacylglycerols are hy- drophobic and therefore unhydrated, the organism that carries fat as fuel does not have to carry the extra weight of water of hydration that is associated with stored poly- saccharides (2 g per gram of polysaccharide). Humans have fat tissue (composed primarily of adipocytes) un- der the skin, in the abdominal cavity, and in the mam- mary glands. Moderately obese people with 15 to 20 kg of triacylglycerols deposited in their adipocytes could meet their energy needs for months by drawing on their fat stores. In contrast, the human body can store less than a day’s energy supply in the form of glycogen. Car- bohydrates such as glucose and glycogen do offer cer- tain advantages as quick sources of metabolic energy, one of which is their ready solubility in water. In some animals, triacylglycerols stored under the skin serve not only as energy stores but as insulation against low temperatures. Seals, walruses, penguins, and other warm-blooded polar animals are amply padded with triacylglycerols. In hibernating animals (bears, for example), the huge fat reserves accumulated before hibernation serve the dual purposes of insulation and energy storage (see Box 17–1). The low density of tri- acylglycerols is the basis for another remarkable func- tion of these compounds. In sperm whales, a store of triacylglycerols and waxes allows the animals to match the buoyancy of their bodies to that of their surround- ings during deep dives in cold water (Box 10–1). Many Foods Contain Triacylglycerols Most natural fats, such as those in vegetable oils, dairy products, and animal fat, are complex mixtures of sim- ple and mixed triacylglycerols. These contain a variety of fatty acids differing in chain length and degree of sat- uration (Fig. 10–4). Vegetable oils such as corn (maize) and olive oil are composed largely of triacylglycerols with unsaturated fatty acids and thus are liquids at room temperature. They are converted industrially into solid Chapter 10 Lipids346 8 m(a) H9262 3 H9262m (b) H9262 FIGURE 10–3 Fat stores in cells. (a) Cross section of four guinea pig adipocytes, showing huge fat droplets that virtually fill the cells. Also visible are several capillaries in cross section. (b) Cross section of a cotyledon cell from a seed of the plant Arabidopsis. The large dark structures are protein bodies, which are surrounded by stored oils in the light-colored oil bodies. 8885d_c10_343-368 1/12/04 1:06 PM Page 346 mac76 mac76:385_reb: fats by catalytic hydrogenation, which reduces some of their double bonds to single bonds and converts oth- ers to trans double bonds. Triacylglycerols containing only saturated fatty acids, such as tristearin, the major component of beef fat, are white, greasy solids at room temperature. When lipid-rich foods are exposed too long to the oxygen in air, they may spoil and become rancid. The unpleasant taste and smell associated with rancidity re- sult from the oxidative cleavage of the double bonds in 10.1 Storage Lipids 347 Fatty acids (% of total) Natural fats at 25 °C Olive oil, liquid Butter, soft solid Beef fat, hard solid C 16 and C 18 saturated C 16 and C 18 unsaturated C 4 to C 14 saturated 20 40 60 80 100 FIGURE 10–4 Fatty acid composition of three food fats. Olive oil, butter, and beef fat consist of mixtures of triacylglycerols, differing in their fatty acid composition. The melting points of these fats—and hence their physical state at room temperature (25H11034C)—are a direct function of their fatty acid composition. Olive oil has a high propor- tion of long-chain (C 16 and C 18 ) unsaturated fatty acids, which ac- counts for its liquid state at 25H11034C. The higher proportion of long-chain (C 16 and C 18 ) saturated fatty acids in butter increases its melting point, so butter is a soft solid at room temperature. Beef fat, with an even higher proportion of long-chain saturated fatty acids, is a hard solid. BOX 10–1 THE WORLD OF BIOCHEMISTRY Sperm Whales: Fatheads of the Deep Studies of sperm whales have uncovered another way in which triacylglycerols are biologically useful. The sperm whale’s head is very large, accounting for over one-third of its total body weight. About 90% of the weight of the head is made up of the spermaceti or- gan, a blubbery mass that contains up to 3,600 kg (about 4 tons) of spermaceti oil, a mixture of triacyl- glycerols and waxes containing an abundance of un- saturated fatty acids. This mixture is liquid at the normal resting body temperature of the whale, about 37 H11034C, but it begins to crystallize at about 31 H11034C and becomes solid when the temperature drops several more degrees. The probable biological function of spermaceti oil has been deduced from research on the anatomy and feeding behavior of the sperm whale. These mammals feed almost exclusively on squid in very deep water. In their feeding dives they descend 1,000 m or more; the deepest recorded dive is 3,000 m (almost 2 miles). At these depths, there are no competitors for the very plentiful squid; the sperm whale rests quietly, waiting for schools of squid to pass. For a marine animal to remain at a given depth without a constant swimming effort, it must have the same density as the surrounding water. The sperm whale undergoes changes in buoyancy to match the density of its surroundings—from the tropical ocean surface to great depths where the water is much colder and thus denser. The key is the freezing point of spermaceti oil. When the temperature of the oil is lowered several degrees during a deep dive, it con- geals or crystallizes and becomes denser. Thus the buoyancy of the whale changes to match the density of seawater. Various physiological mechanisms pro- mote rapid cooling of the oil during a dive. During the return to the surface, the congealed spermaceti oil warms and melts, decreasing its density to match that of the surface water. Thus we see in the sperm whale a remarkable anatomical and biochemical adaptation. The triacylglycerols and waxes synthesized by the sperm whale contain fatty acids of the necessary chain length and degree of unsaturation to give the sper- maceti oil the proper melting point for the animal’s diving habits. Unfortunately for the sperm whale population, spermaceti oil was at one time considered the finest lamp oil and continues to be commercially valuable as a lubricant. Several centuries of intensive hunting of these mammals have driven sperm whales onto the endangered species list. Spermaceti organ 8885d_c10_343-368 1/12/04 1:06 PM Page 347 mac76 mac76:385_reb: unsaturated fatty acids, which produces aldehydes and carboxylic acids of shorter chain length and therefore higher volatility. Waxes Serve as Energy Stores and Water Repellents Biological waxes are esters of long-chain (C 14 to C 36 ) saturated and unsaturated fatty acids with long-chain (C 16 to C 30 ) alcohols (Fig. 10–5). Their melting points (60 to 100 H11034C) are generally higher than those of tria- cylglycerols. In plankton, the free-floating microorgan- isms at the bottom of the food chain for marine animals, waxes are the chief storage form of metabolic fuel. Waxes also serve a diversity of other functions re- lated to their water-repellent properties and their firm consistency. Certain skin glands of vertebrates secrete waxes to protect hair and skin and keep it pliable, lu- bricated, and waterproof. Birds, particularly waterfowl, secrete waxes from their preen glands to keep their feathers water-repellent. The shiny leaves of holly, rhododendrons, poison ivy, and many tropical plants are coated with a thick layer of waxes, which prevents excessive evaporation of water and protects against parasites. Biological waxes find a variety of applications in the pharmaceutical, cosmetic, and other industries. Lanolin (from lamb’s wool), beeswax (Fig. 10–5), carnauba wax (from a Brazilian palm tree), and wax extracted from spermaceti oil (from whales; see Box 10–1) are widely used in the manufacture of lotions, ointments, and polishes. SUMMARY 10.1 Storage Lipids ■ Lipids are water-insoluble cellular components of diverse structure that can be extracted by nonpolar solvents. ■ Almost all fatty acids, the hydrocarbon components of many lipids, have an even number of carbon atoms (usually 12 to 24); they are either saturated or unsaturated, with double bonds almost always in the cis configuration. ■ Triacylglycerols contain three fatty acid molecules esterified to the three hydroxyl groups of glycerol. Simple triacylglycerols contain only one type of fatty acid; mixed triacylglycerols, two or three types. Triacylglycerols are primarily storage fats; they are present in many foods. 10.2 Structural Lipids in Membranes The central architectural feature of biological mem- branes is a double layer of lipids, which acts as a bar- rier to the passage of polar molecules and ions. Mem- brane lipids are amphipathic: one end of the molecule is hydrophobic, the other hydrophilic. Their hydropho- bic interactions with each other and their hydrophilic interactions with water direct their packing into sheets called membrane bilayers. In this section we describe five general types of membrane lipids: glycerophospho- lipids, in which the hydrophobic regions are composed of two fatty acids joined to glycerol; galactolipids and sulfolipids, which also contain two fatty acids esterified to glycerol, but lack the characteristic phosphate of phos- pholipids; archaebacterial tetraether lipids, in which two very long alkyl chains are ether-linked to glycerol at both ends; sphingolipids, in which a single fatty acid is joined to a fatty amine, sphingosine; and sterols, compounds characterized by a rigid system of four fused hydrocar- bon rings. The hydrophilic moieties in these amphipathic com- pounds may be as simple as a single OOH group at one end of the sterol ring system, or they may be much more complex. In glycerophospholipids and some sphin- golipids, a polar head group is joined to the hydropho- bic moiety by a phosphodiester linkage; these are the phospholipids. Other sphingolipids lack phosphate but have a simple sugar or complex oligosaccharide at their polar ends; these are the glycolipids (Fig. 10–6). Within these groups of membrane lipids, enormous diversity re- sults from various combinations of fatty acid “tails” and polar “heads.” The arrangement of these lipids in mem- branes, and their structural and functional roles therein, are considered in the next chapter. Chapter 10 Lipids348 FIGURE 10–5 Biological wax. (a) Triacontanoylpalmitate, the major component of beeswax, is an ester of palmitic acid with the alcohol triacontanol. (b) A honeycomb, constructed of beeswax, is firm at 25H11034C and completely impervious to water. The term “wax” originates in the Old English weax, meaning “the material of the honeycomb.” (b) CH 3 (CH 2 ) 14 C O O CH 2 (CH 2 ) 28 CH 3 Palmitic acid (a) 1-Triacontanol 8885d_c10_343-368 1/12/04 1:06 PM Page 348 mac76 mac76:385_reb: Glycerophospholipids Are Derivatives of Phosphatidic Acid Glycerophospholipids, also called phosphoglycerides, are membrane lipids in which two fatty acids are attached in ester linkage to the first and second carbons of glyc- erol, and a highly polar or charged group is attached through a phosphodiester linkage to the third carbon. Glycerol is prochiral; it has no asymmetric carbons, but attachment of phosphate at one end converts it into a chiral compound, which can be correctly named either L-glycerol 3-phosphate, D-glycerol 1-phosphate, or sn- glycerol 3-phosphate (Fig. 10–7). Glycerophospholipids are named as derivatives of the parent compound, phos- phatidic acid (Fig. 10–8), according to the polar alcohol in the head group. Phosphatidylcholine and phosphati- dylethanolamine have choline and ethanolamine in their polar head groups, for example. In all these compounds, the head group is joined to glycerol through a phos- phodiester bond, in which the phosphate group bears a negative charge at neutral pH. The polar alcohol may be negatively charged (as in phosphatidylinositol 4,5-bisphosphate), neutral (phosphatidylserine), or pos- itively charged (phosphatidylcholine, phosphatidylethan- olamine). As we shall see in Chapter 11, these charges contribute greatly to the surface properties of membranes. The fatty acids in glycerophospholipids can be any of a wide variety, so a given phospholipid (phospha- tidylcholine, for example) may consist of a number of molecular species, each with its unique complement of fatty acids. The distribution of molecular species is spe- cific for different organisms, different tissues of the same organism, and different glycerophospholipids in the same cell or tissue. In general, glycerophospholipids contain a C 16 or C 18 saturated fatty acid at C-1 and a C 18 to C 20 unsaturated fatty acid at C-2. With few ex- ceptions, the biological significance of the variation in fatty acids and head groups is not yet understood. Some Phospholipids Have Ether-Linked Fatty Acids Some animal tissues and some unicellular organisms are rich in ether lipids, in which one of the two acyl chains is attached to glycerol in ether, rather than ester, link- age. The ether-linked chain may be saturated, as in the alkyl ether lipids, or may contain a double bond between C-1 and C-2, as in plasmalogens (Fig. 10–9). Vertebrate heart tissue is uniquely enriched in ether lipids; about half of the heart phospholipids are plasmalogens. The membranes of halophilic bacteria, ciliated protists, and certain invertebrates also contain high proportions of 10.2 Structural Lipids in Membranes 349 Storage lipids (neutral) Membrane lipids (polar) Phospholipids Glycolipids Triacylglycerols Glycerophospholipids Sphingolipids Sphingolipids Alcohol Sphingosine Archaebacterial ether lipids Glycerol Glycerol Fatty acid Fatty acid Fatty acid Fatty acid Fatty acid Fatty acid Fatty acid Diphytanyl Fatty acid Fatty acid PO 4 PO 4 PO 4 PO 4 Choline Sphingosine Glycerol Glycerol Diphytanyl Glycerol (SO 4 ) Glycerol Galactolipids (sulfolipids) Mono- or oligosaccharide Mono- or disaccharide ( ether linkage) FIGURE 10–6 Some common types of storage and membrane lipids. All the lipid types shown here have either glycerol or sphingosine as the backbone (pink screen), to which are attached one or more long- chain alkyl groups (yellow) and a polar head group (blue). In triacyl- glycerols, glycerophospholipids, galactolipids, and sulfolipids, the alkyl groups are fatty acids in ester linkage. Sphingolipids contain a single fatty acid, in amide linkage to the sphingosine backbone. The membrane lipids of archaebacteria are variable; that shown here has two very long, branched alkyl chains, each end in ether linkage with a glycerol moiety. In phospholipids the polar head group is joined through a phosphodiester, whereas glycolipids have a direct glycosidic linkage between the head-group sugar and the backbone glycerol. 1 CH 2 OH 2 COHH 3 CH 2 O O H11002 O O H11002 P L-Glycerol 3-phosphate (sn-glycerol 3-phosphate) FIGURE 10–7 L-Glycerol 3-phosphate, the backbone of phospho- lipids. Glycerol itself is not chiral, as it has a plane of symmetry through C-2. However, glycerol can be converted to a chiral compound by adding a substituent such as phosphate to either of the OCH 2 OH groups; that is, glycerol is prochiral. One unambiguous nomenclature for glycerol phosphate is the DL system (described on p. 77), in which the isomers are named according to their stereochemical relationships to glyceraldehyde isomers. By this system, the stereoisomer of glycerol phosphate found in most lipids is correctly named either L-glycerol 3-phosphate or D-glycerol 1-phosphate. Another way to specify stereoisomers is the stereospecific numbering (sn) system, in which C-1 is, by definition, that group of the prochiral compound that oc- cupies the pro-S position. The common form of glycerol phosphate in phospholipids is, by this system, sn-glycerol 3-phosphate. 8885d_c10_349 1/16/04 8:18 AM Page 349 mac76 mac76:385_reb: ether lipids. The functional significance of ether lipids in these membranes is unknown; perhaps their resistance to the phospholipases that cleave ester-linked fatty acids from membrane lipids is important in some roles. At least one ether lipid, platelet-activating factor, is a potent molecular signal. It is released from leukocytes called basophils and stimulates platelet aggregation and the release of serotonin (a vasocon- strictor) from platelets. It also exerts a variety of effects on liver, smooth muscle, heart, uterine, and lung tissues and plays an important role in inflammation and the allergic response. ■ Chapter 10 Lipids350 Glycerophospholipid Head-group (general structure) substituent 2 C 1 CH 2 O C O 3 CH 2 O P O O H11002 O X H O O C Saturated fatty acid (e.g., palmitic acid) Unsaturated fatty acid (e.g., oleic acid) Name of X Formula of X Net charge (at pH 7) H H110021 Ethanolamine 0 Choline 0 Serine H110021 Glycerol H110021 myo-Inositol 4,5- bisphosphate H110024 Phosphatidyl- H110022 glycerol CH 2 CH 2 N H11001 H 3 CH 2 CH 2 N H11001 (CH 3 ) 3 CH 2 C COO H11002 H N H11001 H 3 H OH HO C C CH 2 HOH H 2 O P O O H11002 O CH 2 C CH 2 O C O R 2 H O C O R 1 OH H H H OP OP HH HO 2 HCHC 2 1 23 4 56 HC OH Name of glycerophospholipid Phosphatidic acid Phosphatidylethanolamine Phosphatidylcholine Phosphatidylserine Phosphatidylglycerol Phosphatidylinositol 4,5-bisphosphate Cardiolipin FIGURE 10–8 Glycerophospholipids. The common glycerophospho- lipids are diacylglycerols linked to head-group alcohols through a phosphodiester bond. Phosphatidic acid, a phosphomonoester, is the parent compound. Each derivative is named for the head-group alco- hol (X), with the prefix “phosphatidyl-.” In cardiolipin, two phospha- tidic acids share a single glycerol. 8885d_c10_343-368 1/12/04 1:06 PM Page 350 mac76 mac76:385_reb: Chloroplasts Contain Galactolipids and Sulfolipids The second group of membrane lipids are those that predominate in plant cells: the galactolipids, in which one or two galactose residues are connected by a gly- cosidic linkage to C-3 of a 1,2-diacylglycerol (Fig. 10–10; see also Fig. 10–6). Galactolipids are localized in the thylakoid membranes (internal membranes) of chloro- plasts; they make up 70% to 80% of the total membrane lipids of a vascular plant. They are probably the most abundant membrane lipids in the biosphere. Phosphate is often the limiting plant nutrient in soil, and perhaps the evolutionary pressure to conserve phosphate for more critical roles favored plants that made phosphate- free lipids. Plant membranes also contain sulfolipids, in which a sulfonated glucose residue is joined to a di- acylglycerol in glycosidic linkage. In sulfolipids, the sul- fonate on the head group bears a fixed negative charge like that of the phosphate group in phospholipids (Fig. 10–10). 10.2 Structural Lipids in Membranes 351 ether-linked alkene 2 C 1 CH 2 O C H C H H O C O 3 CH 2 O O P H11002 O O CH 2 CH 2 N H11001 (CH 3 ) 3 Plasmalogen choline O P O 3 C 2 C 1 CH 2 O CH 2 CH 2 H O C O CH 3 H 2 H11002 O O CH 2 CH 2 N H11001 (CH 3 ) 3 acetyl ester Platelet-activating factor ether-linked alkane choline FIGURE 10–9 Ether lipids. Plasmalogens have an ether-linked alkenyl chain where most glycerophospholipids have an ester-linked fatty acid (compare Fig. 10–8). Platelet-activating factor has a long ether-linked alkyl chain at C-1 of glycerol, but C-2 is ester-linked to acetic acid, which makes the compound much more water-soluble than most glyc- erophospholipids and plasmalogens. The head-group alcohol is choline in plasmalogens and in platelet-activating factor. Monogalactosyldiacylglycerol (MGDG) CH 2 OH CH 2 CH HO H O O O O C O O CCH 2 H H H H OH OH 6-Sulfo-6-deoxy-H9251-D-glucopyranosyldiacylglycerol (a sulfolipid) CH 2 CH 2 CH HO O O O O C SO O O H11002 O O CCH 2 H H H H H OH OH CH 2 OH CH 2 HO H O O H H H H OH OH Digalactosyldiacylglycerol (DGDG) CH 2 CH HO H O O O O C O O CCH 2 H H H H OH OH FIGURE 10–10 Three glycolipids of chloroplast membranes. In monogalactosyldiacylglycerols (MGDGs) and digalactosyldiacylglycerols (DGDGs), almost all the acyl groups are derived from linoleic acid (18:2(H9004 9,12 )) and the head groups are uncharged. In the sulfolipid 6-sulfo- 6-deoxy-H9251-D-glucopyranosyldiacylglycerol, the sulfonate carries a fixed negative charge. 8885d_c10_343-368 1/12/04 1:06 PM Page 351 mac76 mac76:385_reb: Archaebacteria Contain Unique Membrane Lipids The archaebacteria, most of which live in ecological niches with extreme conditions—high temperatures (boiling water), low pH, high ionic strength, for ex- ample—have membrane lipids containing long-chain (32 carbons) branched hydrocarbons linked at each end to glycerol (Fig. 10–11). These linkages are through ether bonds, which are much more stable to hydrolysis at low pH and high temperature than are the ester bonds found in the lipids of eubacteria and eukaryotes. In their fully extended form, these archaebacterial lipids are twice the length of phospholipids and sphingolipids and span the width of the surface membrane. At each end of the extended molecule is a polar head consisting of glycerol linked to either phosphate or sugar residues. The general name for these compounds, glycerol dialkyl glycerol tetraethers (GDGTs), reflects their unique structure. The glycerol moiety of the archaebacterial lipids is not the same stereoisomer as that in the lipids of eubacteria and eukaryotes; the central carbon is in the R configuration in archaebacteria, in the S configu- ration in the other kingdoms (Fig. 10–7). Sphingolipids Are Derivatives of Sphingosine Sphingolipids, the fourth large class of membrane lipids, also have a polar head group and two nonpolar tails, but unlike glycerophospholipids and galactolipids they contain no glycerol. Sphingolipids are composed of one molecule of the long-chain amino alcohol sphingo- sine (also called 4-sphingenine) or one of its derivatives, one molecule of a long-chain fatty acid, and a polar head group that is joined by a glycosidic linkage in some cases and by a phosphodiester in others (Fig. 10–12). Carbons C-1, C-2, and C-3 of the sphingosine mol- ecule are structurally analogous to the three carbons of glycerol in glycerophospholipids. When a fatty acid is attached in amide linkage to the ONH 2 on C-2, the re- sulting compound is a ceramide, which is structurally similar to a diacylglycerol. Ceramide is the structural parent of all sphingolipids. There are three subclasses of sphingolipids, all de- rivatives of ceramide but differing in their head groups: sphingomyelins, neutral (uncharged) glycolipids, and gangliosides. Sphingomyelins contain phosphocholine or phosphoethanolamine as their polar head group and are therefore classified along with glycerophospholipids as phospholipids (Fig. 10–6). Indeed, sphingomyelins resemble phosphatidylcholines in their general proper- ties and three-dimensional structure, and in having no net charge on their head groups (Fig. 10–13). Sphingo- myelins are present in the plasma membranes of animal cells and are especially prominent in myelin, a mem- branous sheath that surrounds and insulates the axons of some neurons—thus the name “sphingomyelins.” Glycosphingolipids, which occur largely in the outer face of plasma membranes, have head groups with one or more sugars connected directly to the OOH at C-1 of the ceramide moiety; they do not contain phos- phate. Cerebrosides have a single sugar linked to ce- ramide; those with galactose are characteristically found in the plasma membranes of cells in neural tissue, and those with glucose in the plasma membranes of cells in nonneural tissues. Globosides are neutral (uncharged) glycosphingolipids with two or more sugars, usually D- glucose, D-galactose, or N-acetyl-D-galactosamine. Cere- brosides and globosides are sometimes called neutral glycolipids, as they have no charge at pH 7. Gangliosides, the most complex sphingolipids, have oligosaccharides as their polar head groups and one or more residues of N-acetylneuraminic acid (Neu5Ac), a sialic acid (often simply called “sialic acid”), at the Chapter 10 Lipids352 OH 2 C CH 2 CH 2 CH 2 C H O H 2 C HC O CH 2 OH O HCOH P H9251Glc(H92521—?2)Gal-1 Glycerol Diphytanyl groups Glycerol phosphate Glycerol O H11002 OO O O 2 1 3 FIGURE 10–11 A typical membrane lipid of archaebacteria. In this diphytanyl tetraether lipid, the diphytanyl moieties (yellow) are long hydrocarbons composed of eight five-carbon isoprene groups con- densed end-to-end (on the condensation of isoprene units, see Fig. 21–36; also, compare the diphytanyl groups with the 20-carbon phy- tol side chain of chlorophylls in Fig. 19–40a). In this extended form, the diphytanyl groups are about twice the length of a 16-carbon fatty acid typically found in the membrane lipids of eubacteria and eu- karyotes. The glycerol moieties in the archaebacterial lipids are in the R configuration, in contrast to those of eubacteria and eukaryotes, which have the S configuration. Archaebacterial lipids differ in the sub- stituents on the glycerols. In the molecule shown here, one glycerol is linked to the disaccharide H9251-glucopyranosyl-(1n2)-H9252-galactofuranose; the other glycerol is linked to a glycerol phosphate head group. 8885d_c10_343-368 1/12/04 1:06 PM Page 352 mac76 mac76:385_reb: termini. Sialic acid gives gangliosides the negative charge at pH 7 that distinguishes them from globosides. Gangliosides with one sialic acid residue are in the GM (M for mono-) series, those with two are in the GD (D for di-) series, and so on (GT, three sialic acid residues; GQ, four). H OH COO H11002 O C OH H C OH H CH 2 OH HN C O CH 3 N-Acetylneuraminic acid (a sialic acid) (Neu5Ac) H H H HOH Sphingolipids at Cell Surfaces Are Sites of Biological Recognition When sphingolipids were discovered a century ago by the physician-chemist Johann Thudichum, their biolog- ical role seemed as enigmatic as the Sphinx, for which he therefore named them. In humans, at least 60 dif- ferent sphingolipids have been identified in cellular membranes. Many of these are especially prominent in the plasma membranes of neurons, and some are clearly recognition sites on the cell surface, but a specific func- tion for only a few sphingolipids has been discovered thus far. The carbohydrate moieties of certain sphin- golipids define the human blood groups and therefore determine the type of blood that individuals can safely receive in blood transfusions (Fig. 10–14). 10.2 Structural Lipids in Membranes 353 Sphingolipid Sphingosine Fatty acid (general structure) 2 C HO 3 CH CH CH (CH 2 ) 12 CH 3 1 CH 2 O X H N H C O Name of X Formula of XName of sphingolipid Ceramide PhosphocholineSphingomyelin Neutral glycolipids Glucosylcerebroside Glucose Di-, tri-, or tetrasaccharide Lactosylceramide (a globoside) Complex oligosaccharide Ganglioside GM2 P O O H11002 O CH 2 CH 2 N H11001 (CH 3 ) 3 CH 2 OH HO Glc GalGlc Gal GalNAc Neu5Ac H H OH H O OH H H H FIGURE 10–12 Sphingolipids. The first three carbons at the polar end of sphingosine are analogous to the three carbons of glycerol in glyc- erophospholipids. The amino group at C-2 bears a fatty acid in amide linkage. The fatty acid is usually saturated or monounsaturated, with 16, 18, 22, or 24 carbon atoms. Ceramide is the parent compound for this group. Other sphingolipids differ in the polar head group (X) attached at C-1. Gangliosides have very complex oligosaccharide head groups. Standard abbreviations for sugars are used in this figure: Glc, D-glucose; Gal, D-galactose; GalNAc, N-acetyl-D-galactosamine; Neu5Ac, N-acetylneuraminic acid (sialic acid). Johann Thudichum, 1829–1901 8885d_c10_343-368 1/12/04 1:06 PM Page 353 mac76 mac76:385_reb: Gangliosides are concentrated in the outer surface of cells, where they present points of recognition for ex- tracellular molecules or surfaces of neighboring cells. The kinds and amounts of gangliosides in the plasma membrane change dramatically during embryonic de- velopment. Tumor formation induces the synthesis of a new complement of gangliosides, and very low concen- trations of a specific ganglioside have been found to in- duce differentiation of cultured neuronal tumor cells. Investigation of the biological roles of diverse ganglio- sides remains fertile ground for future research. Phospholipids and Sphingolipids Are Degraded in Lysosomes Most cells continually degrade and replace their mem- brane lipids. For each hydrolyzable bond in a glyc- erophospholipid, there is a specific hydrolytic enzyme in the lysosome (Fig. 10–15). Phospholipases of the A type remove one of the two fatty acids, producing a lysophospholipid. (These esterases do not attack the ether link of plasmalogens.) Lysophospholipases remove the remaining fatty acid. Gangliosides are degraded by a set of lysosomal en- zymes that catalyze the stepwise removal of sugar units, finally yielding a ceramide. A genetic defect in any of these hydrolytic enzymes leads to the accumulation of gangliosides in the cell, with severe medical conse- quences (Box 10–2). Sterols Have Four Fused Carbon Rings Sterols are structural lipids present in the membranes of most eukaryotic cells. The characteristic structure of this fifth group of membrane lipids is the steroid nu- Chapter 10 Lipids354 CH 3 N CH 3 CH 3 H11001 C CH 2 O O O H11002 H 2 O C P H 2 CH NH O C OH H C Sphingomyelin NCH 3 CH 3 H11001 C CH 2 O O O H11002 H 2 O H 2 Phosphatidylcholine CH O C O C C O P H 2 O C CH 3 FIGURE 10–13 The similarities in shape and in molecular structure of phosphatidylcholine (a glycerophospholipid) and sphingomyelin (a sphingolipid) are clear when their space-filling and structural formu- las are drawn as here. Ceramide Gal O Antigen A Antigen B Antigen Sphingosine Fatty acid Glc Gal GalNAc Gal Fuc GalNAc FIGURE 10–14 Glycosphingolipids as determinants of blood groups. The human blood groups (O, A, B) are determined in part by the oligosaccharide head groups (blue) of these glycosphin- golipids. The same three oligosaccharides are also found attached to certain blood proteins of individuals of blood types O, A, and B, re- spectively. (Fuc represents the sugar fucose.) 8885d_c10_343-368 1/12/04 1:06 PM Page 354 mac76 mac76:385_reb: cleus, consisting of four fused rings, three with six car- bons and one with five (Fig. 10–16). The steroid nucleus is almost planar and is relatively rigid; the fused rings do not allow rotation about COC bonds. Cholesterol, the major sterol in animal tissues, is amphipathic, with a polar head group (the hydroxyl group at C-3) and a nonpolar hydrocarbon body (the steroid nucleus and the hydrocarbon side chain at C-17), about as long as a 16- carbon fatty acid in its extended form. Similar sterols are found in other eukaryotes: stigmasterol in plants and ergosterol in fungi, for example. Bacteria cannot syn- thesize sterols; a few bacterial species, however, can in- corporate exogenous sterols into their membranes. The sterols of all eukaryotes are synthesized from simple five- carbon isoprene subunits, as are the fat-soluble vitamins, quinones, and dolichols described in Section 10.3. In addition to their roles as membrane constituents, the sterols serve as precursors for a variety of products with specific biological activities. Steroid hormones, for example, are potent biological signals that regulate gene expression. Bile acids are polar derivatives of choles- terol that act as detergents in the intestine, emulsifying dietary fats to make them more readily accessible to di- gestive lipases. We return to cholesterol and other sterols in later chapters, to consider the structural role of cho- lesterol in biological membranes (Chapter 11), signal- ing by steroid hormones (Chapter 12), the remarkable biosynthetic pathway to cholesterol, and the transport of cholesterol by lipoprotein carriers (Chapter 21). SUMMARY 10.2 Structural Lipids in Membranes ■ The polar lipids, with polar heads and nonpolar tails, are major components of membranes. The most abundant are the glycerophospholipids, which contain fatty acids esterified to two of the hydroxyl groups of glycerol, and a second alcohol, the head group, esterified to the third hydroxyl of glycerol via a phosphodiester bond. Other polar lipids are the sterols. ■ Glycerophospholipids differ in the structure of their head group; common glycerophospholipids are phosphatidylethanolamine and phosphatidylcholine. The polar heads of the glycerophospholipids carry electric charges at pH near 7. ■ Chloroplast membranes are remarkably rich in galactolipids, composed of a diacylglycerol with 10.2 Structural Lipids in Membranes 355 Phospholipase A 1 Phospholipase C Phospholipase A 2 Phospholipase D O P O 3 C 2 C 1 CH 2 O C O H O C O H 2 O H11002 O H OH HOOH H H H OP OP HH FIGURE 10–15 The specificities of phospholipases. Phospholipases A 1 and A 2 hydrolyze the ester bonds of intact glycerophospholipids at C-1 and C-2 of glycerol, respectively. Phospholipases C and D each split one of the phosphodiester bonds in the head group. Some phospholipases act on only one type of glycerophospholipid, such as phosphatidylinositol 4,5-bisphosphate (shown here) or phosphatidyl- choline; others are less specific. When one of the fatty acids has been removed by a type A phospholipase, the second fatty acid is cleaved from the molecule by a lysophospholipase (not shown). Alkyl Polar Steroid side head nucleus chain 20 C 22 C 23 C 24 C 25 C 19 CH 3 H 27 CH 3 H 2 H 2 H 2 H 21 CH 3 AB C D HO 26 CH 3 18 CH 3 5 11 4 2 1 8 3 9 7 6 10 13 12 17 16 1514 FIGURE 10–16 Cholesterol. The stick structure of cholesterol is visi- ble through a transparent surface contour model of the molecule (from coordinates supplied by Dave Woodcock). In the chemical structure, the rings are labeled A through D to simplify reference to derivatives of the steroid nucleus, and the carbon atoms are numbered in blue. The C-3 hydroxyl group (pink in both representations) is the polar head group. For storage and transport of the sterol, this hydroxyl group con- denses with a fatty acid to form a sterol ester. HO CH 3 OH 17 C O NH CH 2 CH 2 SO 3 H11002 Taurocholic acid (a bile acid) OH CH 3 CH 3 8885d_c10_343-368 1/12/04 1:06 PM Page 355 mac76 mac76:385_reb: BOX 10–2 BIOCHEMISTRY IN MEDICINE Inherited Human Diseases Resulting from Abnormal Accumulations of Membrane Lipids The polar lipids of membranes undergo constant meta- bolic turnover, the rate of their synthesis normally counterbalanced by the rate of breakdown. The breakdown of lipids is promoted by hydrolytic en- zymes in lysosomes, each enzyme capable of hy- drolyzing a specific bond. When sphingolipid degra- dation is impaired by a defect in one of these enzymes (Fig. 1), partial breakdown products accumulate in the tissues, causing serious disease. For example, Niemann-Pick disease is caused by a rare genetic defect in the enzyme sphingomyelinase, which cleaves phosphocholine from sphingomyelin. Sphingomyelin accumulates in the brain, spleen, and liver. The disease becomes evident in infants, and causes mental retardation and early death. More com- mon is Tay-Sachs disease, in which ganglioside GM2 accumulates in the brain and spleen (Fig. 2) owing to lack of the enzyme hexosaminidase A. The symptoms of Tay-Sachs disease are progressive retardation in de- velopment, paralysis, blindness, and death by the age of 3 or 4 years. Genetic counseling can predict and avert many in- heritable diseases. Tests on prospective parents can detect abnormal enzymes, then DNA testing can de- termine the exact nature of the defect and the risk it poses for offspring. Once a pregnancy occurs, fetal cells obtained by sampling a part of the placenta (chorionic villus sampling) or the fluid surrounding the fetus (amniocentesis) can be tested in the same way. H9252-galactosidase H9251-galacto- sidase A H9252-galactosidase Ceramide GM1 Generalized gangliosidosis glucocerebrosidase Ceramide Ceramide Gaucher’s disease hexosaminidase A Ceramide Tay-Sachs disease GM2 hexosaminidase A and B Ceramide Sandhoff’s disease Globoside Ceramide Fabry’s disease ganglioside neuraminidase Ceramide GM3 sphingo- myelinase Ceramide Phosphocholine Sphingomyelin Phosphocholine Niemann-Pick disease Ceramide Glc Gal GalNAc Neu5Ac 1mH9262 FIGURE 1 Pathways for the breakdown of GM1, globoside, and sphingomyelin to ceramide. A defect in the enzyme hydrolyzing a particular step is indicated by H11538, and the disease that results from accumulation of the partial breakdown product is noted. FIGURE 2 Electron micrograph of a portion of a brain cell from an infant with Tay-Sachs disease, showing abnormal ganglioside deposits in the lysosomes. 8885d_c10_343-368 1/12/04 1:06 PM Page 356 mac76 mac76:385_reb: one or two linked galactose residues, and sulfolipids, diacylglycerols with a linked sulfonated sugar residue and thus a negatively charged head group. ■ Archaebacteria have unique membrane lipids, with long-chain alkyl groups ether-linked to glycerol at both ends and with sugar residues and/or phosphate joined to the glycerol to provide a polar or charged head group. These lipids are stable under the harsh conditions in which archaebacteria live. ■ The sphingolipids contain sphingosine, a long- chain aliphatic amino alcohol, but no glycerol. Sphingomyelin has, in addition to phosphoric acid and choline, two long hydrocarbon chains, one contributed by a fatty acid and the other by sphingosine. Three other classes of sphingolipids are cerebrosides, globosides, and gangliosides, which contain sugar components. ■ Sterols have four fused rings and a hydroxyl group. Cholesterol, the major sterol in animals, is both a structural component of membranes and precursor to a wide variety of steroids. 10.3 Lipids as Signals, Cofactors, and Pigments The two functional classes of lipids considered thus far (storage lipids and structural lipids) are major cellular components; membrane lipids make up 5% to 10% of the dry mass of most cells, and storage lipids more than 80% of the mass of an adipocyte. With some important exceptions, these lipids play a passive role in the cell; lipid fuels are stored until oxidized by enzymes, and membrane lipids form impermeable barriers around cells and cellular compartments. Another group of lipids, present in much smaller amounts, have active roles in the metabolic traffic as metabolites and mes- sengers. Some serve as potent signals—as hormones, carried in the blood from one tissue to another, or as in- tracellular messengers generated in response to an ex- tracellular signal (hormone or growth factor). Others function as enzyme cofactors in electron-transfer reactions in chloroplasts and mitochondria, or in the transfer of sugar moieties in a variety of glycosylation (addition of sugar) reactions. A third group consists of lipids with a system of conjugated double bonds: pig- ment molecules that absorb visible light. Some of these act as light-capturing pigments in vision and photosyn- thesis; others produce natural colorations, such as the orange of pumpkins and carrots and the yellow of ca- nary feathers. Specialized lipids such as these are de- rived from lipids of the plasma membrane or from the fat-soluble vitamins A, D, E, and K. We describe in this section a few of these biologically active lipids. In later chapters, their synthesis and biological roles are con- sidered in more detail. Phosphatidylinositols and Sphingosine Derivatives Act as Intracellular Signals Phosphatidylinositol and its phosphorylated derivatives act at several levels to regulate cell structure and me- tabolism (Fig. 10–17). Phosphatidylinositol 4,5-bisphos- phate (Fig. 10–8) in the cytoplasmic (inner) face of plasma membranes serves as a specific binding site for certain cytoskeletal proteins and for some soluble pro- teins involved in membrane fusion during exocytosis. It also serves as a reservoir of messenger molecules that are released inside the cell in response to extracellular signals interacting with specific receptors on the outer surface of the plasma membrane. The signals act through a series of steps (Fig. 10–17) that begins with enzymatic removal of a phospholipid head group and ends with ac- tivation of an enzyme (protein kinase C). For example, when the hormone vasopressin binds to plasma mem- brane receptors on the epithelial cells of the renal col- lecting duct, a specific phospholipase C is activated. Phospholipase C hydrolyzes the bond between glyc- erol and phosphate in phosphatidylinositol 4,5-bisphos- phate, releasing two products: inositol 1,4,5-trisphos- phate (IP 3 ), which is water-soluble, and diacylglycerol, which remains associated with the plasma membrane. IP 3 triggers release of Ca 2H11001 from the endoplasmic retic- ulum, and the combination of diacylglycerol and elevated cytosolic Ca 2H11001 activates the enzyme protein kinase C. 10.3 Lipids as Signals, Cofactors, and Pigments 357 Phosphatidylinositol phosphorylation 2ATP in plasma in plasma membrane 2ADP Phosphatidylinositol 4,5-bisphosphate hormone-sensitive H 2 O phospholipase C membrane DiacylglycerolInositol 1,4,5-trisphosphate Activation of protein kinase C Release of intracellular Ca 2H11001 Regulation of other enzymes (by protein phosphorylation) Regulation of other enzymes (by Ca 2H11001 ) FIGURE 10–17 Phosphatidylinositols in cellular regulation. Phos- phatidylinositol 4,5-bisphosphate in the plasma membrane is hy- drolyzed by a specific phospholipase C in response to hormonal signals. Both products of hydrolysis act as intracellular messengers. 8885d_c10_343-368 1/12/04 1:06 PM Page 357 mac76 mac76:385_reb: This enzyme catalyzes the transfer of a phosphoryl group from ATP to a specific residue in one or more tar- get proteins, thereby altering their activity and conse- quently the cell’s metabolism. This signaling mechanism is described more fully in Chapter 12 (see Fig. 12–19). Inositol phospholipids also serve as points of nu- cleation for certain supramolecular complexes involved in signaling or in exocytosis. Proteins that contain cer- tain structural motifs, called PH and PX domains (for pleckstrin homology and Phox homology, respectively), bind phosphatidylinositols in the membrane with high specificity and affinity, initiating the formation of mul- tienzyme complexes at the membrane’s cytosolic sur- face. A number of proteins bind specifically to phos- phatidylinositol 3,4,5-trisphosphate, and the formation of this phospholipid in response to extracellular signals brings the proteins together at the surface of the plasma membrane (see Fig. 12–8). Membrane sphingolipids also can serve as sources of intracellular messengers. Both ceramide and sphin- gomyelin (Fig. 10–12) are potent regulators of protein kinases, and ceramide or its derivatives are known to be involved in the regulation of cell division, differentiation, migration, and programmed cell death (also called apop- tosis; see Chapter 12). Eicosanoids Carry Messages to Nearby Cells Eicosanoids are paracrine hormones, substances that act only on cells near the point of hormone synthesis instead of being transported in the blood to act on cells in other tissues or organs. These fatty acid derivatives have a variety of dramatic effects on verte- brate tissues. They are known to be involved in repro- ductive function; in the inflammation, fever, and pain associated with injury or disease; in the formation of blood clots and the regulation of blood pressure; in gas- tric acid secretion; and in a variety of other processes important in human health or disease. All eicosanoids are derived from arachidonic acid (20:4(H9004 5,8,11,14 )) (Fig. 10–18), the 20-carbon polyun- saturated fatty acid from which they take their gen- Chapter 10 Lipids358 Arachidonic acid 1 C O OH CH 3 O C O O H11002 O H CH 3 Prostaglandin E 1 (PGE 1 ) C O O H11002 CH 3 Leukotriene A 4 O CH 3 OH O NSAIDs C O O H11002 8 12 O HO 8 12 8 11 5 14 Thromboxane A 2 (b) Eicosanoids C C C O X H 2 H O C O H 2 O C O Membrane Polar phospholipid head group (a) Phospholipase A 2 851114 FIGURE 10–18 Arachidonic acid and some eicosanoid de- rivatives. (a) In response to hormonal signals, phospholipase A 2 cleaves arachidonic acid–containing membrane phospholipids to release arachidonic acid (arachidonate at pH 7), the precursor to var- ious eicosanoids. (b) These compounds include prostaglandins such as PGE 1 , in which C-8 and C-12 of arachidonate are joined to form the characteristic five-membered ring. In thromboxane A 2 , the C-8 and C-12 are joined and an oxygen atom is added to form the six- membered ring. Leukotriene A 4 has a series of three conjugated dou- ble bonds. Nonsteroidal antiinflammatory drugs (NSAIDs) such as aspirin and ibuprofen block the formation of prostaglandins and throm- boxanes from arachidonate by inhibiting the enzyme cyclooxygenase (prostaglandin H 2 synthase). 8885d_c10_343-368 1/12/04 1:06 PM Page 358 mac76 mac76:385_reb: eral name (Greek eikosi, “twenty”). There are three classes of eicosanoids: prostaglandins, thromboxanes, and leukotrienes. Prostaglandins (PG) contain a five-carbon ring originating from the chain of arachidonic acid. Their name derives from the prostate gland, the tissue from which they were first isolated by Bengt Samuelsson and Sune Bergstr?m. Two groups of prostaglandins were originally defined: PGE, for ether-soluble, and PGF, for phosphate ( fosfat in Swedish) buffer–soluble. Each group contains numerous subtypes, named PGE 1 , PGE 2 , and so forth. Prostaglandins act in many tissues by regulating the syn- thesis of the intracellular messenger 3H11032,5H11032-cyclic AMP (cAMP). Because cAMP mediates the action of diverse hormones, the prostaglandins affect a wide range of cellular and tissue functions. Some prostaglandins stim- ulate contraction of the smooth muscle of the uterus during menstruation and labor. Others affect blood flow to specific organs, the wake-sleep cycle, and the re- sponsiveness of certain tissues to hormones such as epinephrine and glucagon. Prostaglandins in a third group elevate body temperature (producing fever) and cause inflammation and pain. The thromboxanes have a six-membered ring con- taining an ether. They are produced by platelets (also called thrombocytes) and act in the formation of blood clots and the reduction of blood flow to the site of a clot. The nonsteroidal antiinflammatory drugs (NSAIDs)— aspirin, ibuprofen, and meclofenamate, for example— were shown by John Vane to inhibit the enzyme prostaglandin H 2 synthase (also called cyclooxygenase or COX), which catalyzes an early step in the pathway from arachidonate to prostaglandins and thromboxanes (Fig. 10–18; see also Box 21–2). Leukotrienes, first found in leukocytes, contain three conjugated double bonds. They are powerful bio- logical signals. For example, leukotriene D 4 , derived from leukotriene A 4 , induces contraction of the muscle lining the airways to the lung. Overproduction of leukotrienes causes asthmatic attacks, and leukotriene synthesis is one target of antiasthmatic drugs such as prednisone. The strong contraction of the smooth mus- cles of the lung that occurs during anaphylactic shock is part of the potentially fatal allergic reaction in indi- viduals hypersensitive to bee stings, penicillin, or other agents. ■ Steroid Hormones Carry Messages between Tissues Steroids are oxidized derivatives of sterols; they have the sterol nucleus but lack the alkyl chain attached to ring D of cholesterol, and they are more po- lar than cholesterol. Steroid hormones move through the bloodstream (on protein carriers) from their site of production to target tissues, where they enter cells, bind to highly specific receptor proteins in the nucleus, and trigger changes in gene expression and metabolism. Be- cause hormones have very high affinity for their recep- tors, very low concentrations of hormones (nanomolar or less) are sufficient to produce responses in target tis- sues. The major groups of steroid hormones are the male and female sex hormones and the hormones produced by the adrenal cortex, cortisol and aldosterone (Fig. 10–19). Prednisone and prednisolone are steroid drugs with potent antiinflammatory activities, mediated in part by the inhibition of arachidonate release by phospholi- pase A 2 (Fig. 10–18) and consequent inhibition of the 10.3 Lipids as Signals, Cofactors, and Pigments 359 John Vane, Sune Bergstr?m, and Bengt Samuelsson C CH 2 OH O Cortisol C O H 2 O H O Aldosterone H O O OH Testosterone Estradiol H 3 H OH HC 3 HC 3 HC 3 HC 3 HC 3 O O O OH C CH HO C CH 2 OH O Prednisolone Prednisone O HC 3 HC 3 OH HO C CH 2 OH O O HC 3 HC 3 OH O C FIGURE 10–19 Steroids derived from cholesterol. Testos- terone, the male sex hormone, is produced in the testes. Estra- diol, one of the female sex hormones, is produced in the ovaries and placenta. Cortisol and aldosterone are hormones synthesized in the cortex of the adrenal gland; they regulate glucose metabolism and salt excretion, respectively. Prednisolone and prednisone are synthetic steroids used as antiinflammatory agents. 8885d_c10_343-368 1/12/04 1:06 PM Page 359 mac76 mac76:385_reb: synthesis of leukotrienes, prostaglandins, and throm- boxanes. They have a variety of medical applications, including the treatment of asthma and rheumatoid arthritis. ■ Plants Use Phosphatidylinositols, Steroids, and Eicosanoidlike Compounds in Signaling Vascular plants contain phosphatidylinositol 4,5-bisphos- phate, as well as the phospholipase that releases IP 3 , and they use IP 3 to regulate the intracellular concen- tration of Ca 2H11001 . Brassinolide and the related group of brassinosteroids are potent growth regulators in plants, increasing the rate of stem elongation and influencing the orientation of cellulose microfibrils in the cell wall during growth. Jasmonate, derived from the fatty acid 18:3(H9004 9,12,15 ) in membrane lipids, is chemically similar to the eicosanoids of animal tissues and also serves as a powerful signal, triggering the plant’s defenses in re- sponse to insect-inflicted damage. The methyl ester of jasmonate gives the characteristic fragrance of jasmine oil, which is widely used in the perfume industry. Vitamins A and D Are Hormone Precursors During the first third of the twentieth century, a major focus of research in physiological chem- istry was the identification of vitamins, compounds that are essential to the health of humans and other verte- brates but cannot be synthesized by these animals and must therefore be obtained in the diet. Early nutritional O COO H11002 Jasmonate Brassinolide (a brassinosteroid) HO H O O HO OH OH Chapter 10 Lipids360 7-Dehydrocholesterol 1,25-Dihydroxycholecalciferol (1,25-dihydroxyvitamin D 3 ) CH 3 CH 3 CH 3 CH 3 CH 3 HO 7 64 1 53 210 9 8 HO CH 2 7 6 4 5 3 2 1 CH 3 CH 3 CH 3 CH 3 25 OH 25 HO CH 2 7 6 4 5 3 2 1 CH 3 CH 3 CH 3 CH 3 OH UV light 2 steps (in skin) (a) Cholecalciferol (vitamin D 3 ) 1 step in the liver 1 step in the kidney FIGURE 10–20 Vitamin D 3 production and metabolism. (a) Cholecalciferol (vitamin D 3 ) is produced in the skin by UV irradiation of 7-dehydrocholesterol, which breaks the bond shaded pink. In the liver, a hydroxyl group is added at C-25 (pink); in the kidney, a second hydroxylation at C-1 (pink) produces the active hormone, 1,25-dihydroxycholecalciferol. This hormone regulates the metabolism of Ca 2H11001 in kidney, intestine, and bone. (b) Dietary vitamin D prevents rickets, a disease once common in cold climates where heavy clothing blocks the UV component of sunlight necessary for the production of vitamin D 3 in skin. On the left is a 2 1 ?2-year-old boy with severe rickets; on the right, the same boy at age 5, after 14 months of vitamin D therapy. Before vitamin D treatment After 14 months of vitamin D treatment (b) 8885d_c10_343-368 1/12/04 1:06 PM Page 360 mac76 mac76:385_reb: 10.3 Lipids as Signals, Cofactors, and Pigments 361 H9252-Carotene Vitamin A 1 point of cleavage (retinol) C oxidation of alcohol to aldehyde 11-cis-Retinal Neuronal all-trans-Retinal signal to brain CH 3 CH 3 CH 3 CH 3 CH 3 CH 3 12 CH 3 CH 3 CH 3 CH 3 CH 2 OH CH 3 15 11 CH 3 CH 3 CH 3 CH 3 2 7 6 12 C 11 11 CH 3 CH 3 CH 3 CH 3 CH 3 CH 3 CH 3 CH 3 CH 3 CH 3 visible light OH OH (visual pigment) (a) (b) (e) (d) oxidation of aldehyde to acid Retinoic acid Hormonal signal to epithelial cells (c) FIGURE 10–21 Vitamin A 1 and its precursor and derivatives. (a) H9252-Carotene is the precursor of vitamin A 1 . Isoprene struc- tural units are set off by dashed red lines. Cleavage of H9252-carotene yields two molecules of vitamin A 1 (retinol) (b). Oxidation at C-15 converts retinol to the aldehyde, retinal (c), and further oxidation produces retinoic acid (d), a hormone that regulates gene expression. Retinal combines with the protein opsin to form rhodopsin (not shown), a vi- sual pigment widespread in nature. In the dark, retinal of rhodopsin is in the 11-cis form (c). When a rhodopsin molecule is excited by visible light, the 11-cis-retinal undergoes a series of photochemical re- actions that convert it to all-trans-retinal (e), forcing a change in the shape of the entire rhodopsin molecule. This transformation in the rod cell of the vertebrate retina sends an electrical signal to the brain that is the basis of visual transduction, a topic we address in more detail in Chapter 12. studies identified two general classes of such com- pounds: those soluble in nonpolar organic solvents (fat- soluble vitamins) and those that could be extracted from foods with aqueous solvents (water-soluble vitamins). Eventually the fat-soluble group was resolved into the four vitamin groups A, D, E, and K, all of which are iso- prenoid compounds synthesized by the condensation of multiple isoprene units. Two of these (D and A) serve as hormone precursors. Vitamin D 3 , also called cholecalciferol, is nor- mally formed in the skin from 7-dehydrocholesterol in a photochemical reaction driven by the UV component of sunlight (Fig. 10–20). Vitamin D 3 is not itself biolog- ically active, but it is converted by enzymes in the liver and kidney to 1,25-dihydroxycholecalciferol, a hormone that regulates calcium uptake in the intestine and cal- cium levels in kidney and bone. Deficiency of vitamin D CH 3 C Isoprene CH CH 2 CH 2 leads to defective bone formation and the disease rick- ets, for which administration of vitamin D produces a dramatic cure. Vitamin D 2 (ergocalciferol) is a com- mercial product formed by UV irradiation of the ergos- terol of yeast. Vitamin D 2 is structurally similar to D 3 , with slight modification to the side chain attached to the sterol D ring. Both have the same biological effects, and D 2 is commonly added to milk and butter as a dietary supplement. Like steroid hormones, the product of vi- tamin D metabolism, 1,25-dihydroxycholecalciferol, reg- ulates gene expression—for example, turning on the synthesis of an intestinal Ca 2H11001 -binding protein. Vitamin A (retinol) in its various forms functions as a hormone and as the visual pigment of the verte- brate eye (Fig. 10–21). Acting through receptor proteins in the cell nucleus, the vitamin A derivative retinoic acid regulates gene expression in the development of ep- ithelial tissue, including skin. Retinoic acid is the active ingredient in the drug tretinoin (Retin-A), used in the treatment of severe acne and wrinkled skin. The vita- min A derivative retinal is the pigment that initiates the 8885d_c10_343-368 1/12/04 1:06 PM Page 361 mac76 mac76:385_reb: response of rod and cone cells of the retina to light, producing a neuronal signal to the brain. This role of retinal is described in detail in Chapter 12. Vitamin A was first isolated from fish liver oils; liver, eggs, whole milk, and butter are good dietary sources. In vertebrates, H9252-carotene, the pigment that gives car- rots, sweet potatoes, and other yellow vegetables their characteristic color, can be enzymatically converted to vitamin A. Deficiency of vitamin A leads to a variety of symptoms in humans, including dryness of the skin, eyes, and mucous membranes; retarded development and growth; and night blindness, an early symptom com- monly used in diagnosing vitamin A deficiency. ■ Vitamins E and K and the Lipid Quinones Are Oxidation-Reduction Cofactors Vitamin E is the collective name for a group of closely related lipids called tocopherols, all of which contain a substituted aromatic ring and a long iso- prenoid side chain (Fig. 10–22a). Because they are hy- drophobic, tocopherols associate with cell membranes, lipid deposits, and lipoproteins in the blood. Tocopherols are biological antioxidants. The aromatic ring reacts with and destroys the most reactive forms of oxygen radicals and other free radicals, protecting unsaturated fatty acids from oxidation and preventing oxidative Chapter 10 Lipids362 FIGURE 10–22 Some other biologically active isoprenoid com- pounds or derivatives. Isoprene structural units are set off by dashed red lines. In most mammalian tissues, ubiquinone (also called coen- zyme Q) has 10 isoprene units. Dolichols of animals have 17 to 21 isoprene units (85 to 105 carbon atoms), bacterial dolichols have 11, and those of plants and fungi have 14 to 24. HO CH 3 O CH 2 CH 2 CH 2 C CH 3 H CH 2 CH 2 CH 2 C CH 3 H CH 2 CH 2 CH 2 C CH 3 H CH 3 OCH 3 CH 2 CH C CH 3 CH 2 CH 2 CH 2 C CH 3 H CH 22 CH 2 CH 2 C CH 3 H CH 3 O OH C CH H 2 C O CH 3 CH 3 O O CH 3 CH 2 CH C CH 3 CH 2 CH 2 CH C CH 3 CH 2 n CH 2 CH C CH 3 CH 3 CH 3 O CH 2 CH C CH 3 CH 2 HO CH 2 CH 2 C CH 3 H CH 2 CH 2 CH C CH 3 CH 2 n CH 2 CH C CH 3 CH 3 CH 3 CH 3 O CH 3 O O O CH 3 O CH 3 CH 2 CH C CH 3 CH 2 n CH 2 CH C CH 3 CH 3 H20899H20898 H20898 H20899 H20899 H20898 H20899H20898 (b) Vitamin K 1 : a blood-clotting cofactor (phylloquinone) (d) Ubiquinone: a mitochondrial electron carrier (coenzyme Q) (n H11005 4 to 8) (e) Plastoquinone: a chloroplast electron carrier (n H11005 4 to 8) (f) Dolichol: a sugar carrier (n H11005 9 to 22) (a) Vitamin E: an antioxidant (c) Warfarin: a blood anticoagulant 8885d_c10_343-368 1/12/04 1:06 PM Page 362 mac76 mac76:385_reb: damage to membrane lipids, which can cause cell fragility. Tocopherols are found in eggs and vegetable oils and are especially abundant in wheat germ. Labo- ratory animals fed diets depleted of vitamin E develop scaly skin, muscular weakness and wasting, and steril- ity. Vitamin E deficiency in humans is very rare; the prin- cipal symptom is fragile erythrocytes. The aromatic ring of vitamin K (Fig. 10–22b) un- dergoes a cycle of oxidation and reduction during the formation of active prothrombin, a blood plasma protein essential in blood clot formation. Prothrombin is a pro- teolytic enzyme that splits peptide bonds in the blood protein fibrinogen to convert it to fibrin, the insoluble fibrous protein that holds blood clots together. Henrik Dam and Edward A. Doisy independently discovered that vitamin K deficiency slows blood clotting, which can be fatal. Vitamin K deficiency is very uncommon in humans, aside from a small percentage of infants who suffer from hemorrhagic disease of the newborn, a po- tentially fatal disorder. In the United States, newborns are routinely given a 1 mg injection of vitamin K. Vita- min K 1 (phylloquinone) is found in green plant leaves; a related form, vitamin K 2 (menaquinone), is formed by bacteria residing in the vertebrate intestine. Warfarin (Fig. 10–22c) is a synthetic compound that inhibits the formation of active prothrombin. It is par- ticularly poisonous to rats, causing death by internal bleeding. Ironically, this potent rodenticide is also an in- valuable anticoagulant drug for treating humans at risk for excessive blood clotting, such as surgical patients and those with coronary thrombosis. ■ Ubiquinone (also called coenzyme Q) and plasto- quinone (Fig. 10–22d, e) are isoprenoids that function as lipophilic electron carriers in the oxidation-reduction reactions that drive ATP synthesis in mitochondria and chloroplasts, respectively. Both ubiquinone and plasto- quinone can accept either one or two electrons and ei- ther one or two protons (see Fig. 19–54). Dolichols Activate Sugar Precursors for Biosynthesis During assembly of the complex carbohydrates of bac- terial cell walls, and during the addition of polysaccha- ride units to certain proteins (glycoproteins) and lipids (glycolipids) in eukaryotes, the sugar units to be added are chemically activated by attachment to isoprenoid al- cohols called dolichols (Fig. 10–22f). These compounds have strong hydrophobic interactions with membrane lipids, anchoring the attached sugars to the membrane, where they participate in sugar-transfer reactions. SUMMARY 10.3 Lipids as Signals, Cofactors, and Pigments ■ Some types of lipids, although present in relatively small quantities, play critical roles as cofactors or signals. ■ Phosphatidylinositol bisphosphate is hydrolyzed to yield two intracellular messengers, diacylglycerol and inositol 1,4,5-trisphosphate. Phosphatidylinositol 3,4,5-trisphosphate is a nucleation point for supramolecular protein complexes involved in biological signaling. ■ Prostaglandins, thromboxanes, and leukotrienes (the eicosanoids), derived from arachidonate, are extremely potent hormones. ■ Steroid hormones, derived from sterols, serve as powerful biological signals, such as the sex hormones. ■ Vitamins D, A, E, and K are fat-soluble compounds made up of isoprene units. All play essential roles in the metabolism or physiology of animals. Vitamin D is precursor to a hormone that regulates calcium metabolism. Vitamin A furnishes the visual pigment of the vertebrate eye and is a regulator of gene expression during epithelial cell growth. Vitamin E functions in the protection of membrane lipids from oxidative damage, and vitamin K is essential in the blood-clotting process. ■ Ubiquinones and plastoquinones, also isoprenoid derivatives, function as electron carriers in mitochondria and chloroplasts, respectively. ■ Dolichols activate and anchor sugars on cellular membranes for use in the synthesis of certain complex carbohydrates, glycolipids, and glycoproteins. 10.4 Working with Lipids In exploring the biological role of lipids in cells and tis- sues, it is essential to know which lipids are present and in what proportions. Because lipids are insoluble in wa- ter, their extraction and subsequent fractionation re- quire the use of organic solvents and some techniques 10.4 Working with Lipids 363 Henrik Dam, 1895–1976 Edward A. Doisy, 1893–1986 8885d_c10_343-368 1/12/04 1:06 PM Page 363 mac76 mac76:385_reb: not commonly used in the purification of water-soluble molecules such as proteins and carbohydrates. In gen- eral, complex mixtures of lipids are separated by dif- ferences in the polarity or solubility of the components in nonpolar solvents. Lipids that contain ester- or amide- linked fatty acids can be hydrolyzed by treatment with acid or alkali or with highly specific hydrolytic enzymes (phospholipases, glycosidases) to yield their component parts for analysis. Some methods commonly used in lipid analysis are shown in Figure 10–23 and discussed below. Lipid Extraction Requires Organic Solvents Neutral lipids (triacylglycerols, waxes, pigments, and so forth) are readily extracted from tissues with ethyl ether, chloroform, or benzene, solvents that do not per- mit lipid clustering driven by hydrophobic interactions. Membrane lipids are more effectively extracted by more polar organic solvents, such as ethanol or methanol, which reduce the hydrophobic interactions among lipid molecules while also weakening the hydrogen bonds and electrostatic interactions that bind membrane lipids to membrane proteins. A commonly used extractant is a mixture of chloroform, methanol, and water, initially in volume proportions (1:2:0.8) that are miscible, produc- ing a single phase. After tissue is homogenized in this solvent to extract all lipids, more water is added to the resulting extract and the mixture separates into two phases, methanol/water (top phase) and chloroform (bottom phase). The lipids remain in the chloroform layer, and more polar molecules such as proteins and sugars partition into the methanol/water layer. Chapter 10 Lipids364 Concentration Elution time homogenized in chloroform/methanol/water Water Methanol/water Chloroform Adsorption chromatography (d) (e) (f) (c) NaOH/methanol Fatty acyl methyl esters Polar lipids Charged lipids Neutral lipids Gas-liquid chromatography High- performance liquid chromatography Tissue 18:0 16:1 14:0 16:0 (b) Thin-layer chromatography 123456789 (a) FIGURE 10–23 Common procedures in the extraction, separation, and identification of cellular lipids. (a) Tissue is homogenized in a chloroform/methanol/water mixture, which on addition of water and removal of unextractable sediment by centrifugation yields two phases. Different types of extracted lipids in the chloroform phase may be sep- arated by (b) adsorption chromatography on a column of silica gel, through which solvents of increasing polarity are passed, or (c) thin- layer chromatography (TLC), in which lipids are carried up a silica gel- coated plate by a rising solvent front, less polar lipids traveling farther than more polar or charged lipids. TLC with appropriate solvents can also be used to separate closely related lipid species; for example, the charged lipids phosphatidylserine, phosphatidylglycerol, and phos- phatidylinositol are easily separated by TLC. For the determination of fatty acid composition, a lipid fraction containing ester-linked fatty acids is transesterified in a warm aque- ous solution of NaOH and methanol (d), producing a mixture of fatty acyl methyl esters. These methyl esters are then separated on the ba- sis of chain length and degree of saturation by (e) gas-liquid chro- matography (GLC) or (f ) high-performance liquid chromatography (HPLC). Precise determination of molecular mass by mass spectrom- etry allows unambiguous identification of individual lipids. 8885d_c10_343-368 1/12/04 1:06 PM Page 364 mac76 mac76:385_reb: Adsorption Chromatography Separates Lipids of Different Polarity Complex mixtures of tissue lipids can be fractionated by chromatographic procedures based on the different polarities of each class of lipid. In adsorption chro- matography (Fig. 10–23b), an insoluble, polar material such as silica gel (a form of silicic acid, Si(OH) 4 ) is packed into a glass column, and the lipid mixture (in chloroform solution) is applied to the top of the col- umn. (In high-performance liquid chromatography, the column is of smaller diameter and solvents are forced through the column under high pressure.) The polar lipids bind tightly to the polar silicic acid, but the neu- tral lipids pass directly through the column and emerge in the first chloroform wash. The polar lipids are then eluted, in order of increasing polarity, by washing the column with solvents of progressively higher polarity. Uncharged but polar lipids (cerebrosides, for example) are eluted with acetone, and very polar or charged lipids (such as glycerophospholipids) are eluted with methanol. Thin-layer chromatography on silicic acid employs the same principle (Fig. 10–23c). A thin layer of silica gel is spread onto a glass plate, to which it adheres. A small sample of lipids dissolved in chloroform is applied near one edge of the plate, which is dipped in a shallow container of an organic solvent or solvent mixture—all of which is enclosed within a chamber saturated with the solvent vapor. As the solvent rises on the plate by capillary action, it carries lipids with it. The less polar lipids move farthest, as they have less tendency to bind to the silicic acid. The separated lipids can be detected by spraying the plate with a dye (rhodamine) that flu- oresces when associated with lipids or by exposing the plate to iodine fumes. Iodine reacts reversibly with the double bonds in fatty acids, such that lipids containing unsaturated fatty acids develop a yellow or brown color. A number of other spray reagents are also useful in de- tecting specific lipids. For subsequent analysis, regions containing separated lipids can be scraped from the plate and the lipids recovered by extraction with an or- ganic solvent. Gas-Liquid Chromatography Resolves Mixtures of Volatile Lipid Derivatives Gas-liquid chromatography separates volatile compo- nents of a mixture according to their relative tenden- cies to dissolve in the inert material packed in the chro- matography column and to volatilize and move through the column, carried by a current of an inert gas such as helium. Some lipids are naturally volatile, but most must first be derivatized to increase their volatility (that is, lower their boiling point). For an analysis of the fatty acids in a sample of phospholipids, the lipids are first heated in a methanol/HCl or methanol/NaOH mixture, which converts fatty acids esterified to glycerol into their methyl esters (in a process of transesterification; Fig. 10–23d). These fatty acyl methyl esters are then loaded onto the gas-liquid chromatography column, and the column is heated to volatilize the compounds. Those fatty acyl esters most soluble in the column material par- tition into (dissolve in) that material; the less soluble lipids are carried by the stream of inert gas and emerge first from the column. The order of elution depends on the nature of the solid adsorbant in the column and on the boiling point of the components of the lipid mixture. Using these techniques, mixtures of fatty acids of vari- ous chain lengths and various degrees of unsaturation can be completely resolved (Fig. 10–23e). Specific Hydrolysis Aids in Determination of Lipid Structure Certain classes of lipids are susceptible to degradation under specific conditions. For example, all ester-linked fatty acids in triacylglycerols, phospholipids, and sterol esters are released by mild acid or alkaline treatment, and somewhat harsher hydrolysis conditions release amide-bound fatty acids from sphingolipids. Enzymes that specifically hydrolyze certain lipids are also useful in the determination of lipid structure. Phospholipases A, C, and D (Fig. 10–15) each split particular bonds in phospholipids and yield products with characteristic sol- ubilities and chromatographic behaviors. Phospholipase C, for example, releases a water-soluble phosphoryl al- cohol (such as phosphocholine from phosphatidyl- choline) and a chloroform-soluble diacylglycerol, each of which can be characterized separately to determine the structure of the intact phospholipid. The combina- tion of specific hydrolysis with characterization of the products by thin-layer, gas-liquid, or high-performance liquid chromatography often allows determination of a lipid structure. Mass Spectrometry Reveals Complete Lipid Structure To establish unambiguously the length of a hydrocarbon chain or the position of double bonds, mass spectral analysis of lipids or their volatile derivatives is invalu- able. The chemical properties of similar lipids (for ex- ample, two fatty acids of similar length unsaturated at different positions, or two isoprenoids with different numbers of isoprene units) are very much alike, and their positions of elution from the various chromato- graphic procedures often do not distinguish between them. When the effluent from a chromatography column is sampled by mass spectrometry, however, the compo- nents of a lipid mixture can be simultaneously separated and identified by their unique pattern of fragmentation (Fig. 10–24). 10.4 Working with Lipids 365 8885d_c10_343-368 1/12/04 1:06 PM Page 365 mac76 mac76:385_reb: Chapter 10 Lipids366 Key Terms fatty acid 343 triacylglycerol 345 lipases 346 phospholipid 348 glycolipid 348 glycerophospholipid 349 ether lipid 349 plasmalogen 349 galactolipid 351 sphingolipid 352 ceramide 352 sphingomyelin 352 glycosphingolipid 352 cerebroside 352 globoside 352 neutral glycolipids 352 gangliosides 352 sterols 354 cholesterol 355 prostaglandins 359 thromboxanes 359 leukotrienes 359 vitamin 360 vitamin D 3 361 cholecalciferol 361 vitamin A (retinol) 361 vitamin E 362 tocopherols 362 vitamin K 363 dolichol 363 Terms in bold are defined in the glossary. Abundance (%) 90 80 70 60 60 80 100 120 140 160 55 67 92 108 123 151 164 178 206 220 260 234 274 300 314 356 371 92 108 164 206 234 274 314 342 178 220 260 300 328 356 328 M + N 180 200 220 240 260 280 300 320 340 360 380 m/z 50 40 30 20 10 H C H O O C FIGURE 10–24 Determination of the structure of a fatty acid by mass spectrometry. The fatty acid is first converted to a derivative that min- imizes migration of the double bonds when the molecule is fragmented by electron bombardment. The derivative shown here is a picolinyl ester of linoleic acid—18:2(H9004 9,12 ) (M r 371)—in which the alcohol is picolinol (red). When bombarded with a stream of electrons, this mol- ecule is volatilized and converted to a parent ion (M H11001 ; M r 371), in which the N atom bears the positive charge, and a series of smaller fragments produced by breakage of COC bonds in the fatty acid. The mass spectrometer separates these charged fragments according to their mass/charge ratio (m/z). (To review the principles of mass spec- trometry, see Box 3–2.) The prominent ions at m/z H11005 92, 108, 151, and 164 contain the pyridine ring of the picolinol and various fragments of the carboxyl group, showing that the compound is indeed a picolinyl ester. The molecular ion (m/z H11005 371) confirms the presence of a C-18 fatty acid with two double bonds. The uniform series of ions 14 atomic mass units (amu) apart represents loss of each successive methyl and methylene group from the right end of the molecule (C-18 of the fatty acid), until the ion at m/z H11005 300 is reached. This is followed by a gap of 26 amu for the carbons of the terminal double bond, at m/z H11005 274; a further gap of 14 amu for the C-11 methylene group, at m/z H11005 260, and so forth. By this means the entire structure is determined, although these data alone do not reveal the configuration (cis or trans) of the double bonds. SUMMARY 10.4 Working with Lipids ■ In the determination of lipid composition, the lipids are first extracted from tissues with organic solvents and separated by thin-layer, gas-liquid, or high-performance liquid chromatography. ■ Phospholipases specific for one of the bonds in a phospholipid can be used to generate simpler compounds for subsequent analysis. ■ Individual lipids are identified by their chromatographic behavior, their susceptibility to hydrolysis by specific enzymes, or mass spectrometry. 8885d_c10_343-368 1/12/04 1:06 PM Page 366 mac76 mac76:385_reb: Chapter 10 Problems 367 Further Reading General Gurr, M.I. & Harwood, J.L. (1991) Lipid Biochemistry: An Introduction, 4th edn, Chapman & Hall, London. A good general resource on lipid structure and metabolism, at the intermediate level. Vance, D.E. & Vance, J.E. (eds) (2002) Biochemistry of Lipids, Lipoproteins, and Membranes, New Comprehensive Biochemistry, Vol. 36, Elsevier Science Publishing Co., Inc., New York. An excellent collection of reviews on various aspects of lipid structure, biosynthesis, and function. Structural Lipids in Membranes Bogdanov, M. & Dowhan, W. (1999) Lipid-assisted protein folding. J. Biol. Chem. 274, 36,827–36,830. A minireview of the role of membrane lipids in the folding of membrane proteins. De Rosa, M. & Gambacorta, A. (1988) The lipids of archaebac- teria. Prog. Lipid Res. 27, 153–175. Dowhan, W. (1997) Molecular basis for membrane phospholipid diversity: why are there so many lipids? Annu. Rev. Biochem. 66, 199–232. Gravel, R.A., Kaback, M.M., Proia, R., Sandhoff, K., Suzuki, K., & Suzuki, K. (2001) The GM 2 gangliosidoses. In The Metabolic and Molecular Bases of Inherited Disease, 8th edn (Scriver, C.R., Sly, W.S., Childs, B., Beaudet, A.L., Valle, D., Kinzler, K.W., & Vogelstein, B., eds), pp. 3827–3876, McGraw-Hill, Inc., New York. This article is one of many in a four-volume set that contains definitive descriptions of the clinical, biochemical, and genetic aspects of hundreds of human metabolic diseases—an authori- tative source and fascinating reading. Hoekstra, D. (ed.) (1994) Cell Lipids, Current Topics in Membranes, Vol. 4, Academic Press, Inc., San Diego. Lipids as Signals, Cofactors, and Pigments Bell, R.M., Exton, J.H., & Prescott, S.M. (eds) (1996) Lipid Second Messengers, Handbook of Lipid Research, Vol. 8, Plenum Press, New York. Binkley, N.C. & Suttie, J.W. (1995) Vitamin K nutrition and osteoporosis. J. Nutr. 125, 1812–1821. Brigelius-Flohé, R. & Traber, M.G. (1999) Vitamin E: function and metabolism. FASEB J. 13, 1145–1155. Chojnacki, T. & Dallner, G. (1988) The biological role of dolichol. Biochem. J. 251, 1–9. Clouse, S.D. (2002) Brassinosteroid signal transduction: clarifying the pathway from ligand perception to gene expression. Mol. Cell 10, 973–982. Lemmon, M.A. & Ferguson, K.M. (2000) Signal-dependent membrane targeting by pleckstrin homology (PH) domains. Biochem. J. 350, 1–18. Prescott, S.M., Zimmerman, G.A., Stafforini, D.M., & McIntyre, T.M. (2000) Platelet-activating factor and related lipid mediators. Annu. Rev. Biochem. 69, 419–445. Schneiter, R. (1999) Brave little yeast, please guide us to Thebes: sphingolipid function in S. cerevisiae. BioEssays 21, 1004–1010. Suttie, J.W. (1993) Synthesis of vitamin K-dependent proteins. FASEB J. 7, 445–452. Vermeer, C. (1990) H9253-Carboxyglutamate-containing proteins and the vitamin K-dependent carboxylase. Biochem. J. 266, 625–636. Describes the biochemical basis for the requirement of vitamin K in blood clotting and the importance of carboxylation in the synthesis of the blood-clotting protein thrombin. Viitala, J. & J?rnefelt, J. (1985) The red cell surface revisited. Trends Biochem. Sci. 10, 392–395. Includes discussion of the human A, B, and O blood type deter- minants. Weber, H. (2002) Fatty acid-derived signals in plants. Trends Plant Sci. 7, 217–224. Zittermann, A. (2001) Effects of vitamin K on calcium and bone metabolism. Curr. Opin. Clin. Nutr. Metab. Care 4, 483–487. Working with Lipids Christie, W.W. (1998) Gas chromatography-mass spectrometry methods for structural analysis of fatty acids. Lipids 33, 343–353. A detailed description of the methods used to obtain data such as those presented in Figure 10–24. Christie, W.W. (2003) Lipid Analysis, 3rd edn, The Oily Press, Bridgwater, England. Hamilton, R.J. & Hamilton, S. (eds) (1992) Lipid Analysis: A Practical Approach, IRL Press at Oxford University Press, New York. This text, though out of print, is available as part of the IRL Press Practical Approach Series on CD-ROM, from Oxford University Press (www.oup-usa/acadsci/pasbooks.html). Matsubara, T. & Hagashi, A. (1991) FAB/mass spectrometry of lipids. Prog. Lipid Res. 30, 301–322. An advanced discussion of the identification of lipids by fast atom bombardment (FAB) mass spectrometry, a powerful tech- nique for structure determination. 1. Operational Definition of Lipids How is the defini- tion of “lipid” different from the types of definitions used for other biomolecules that we have considered, such as amino acids, nucleic acids, and proteins? 2. Melting Points of Lipids The melting points of a se- ries of 18-carbon fatty acids are: stearic acid, 69.6 H11034C; oleic acid, 13.4 H11034C; linoleic acid, H110025 H11034C; and linolenic acid, H1100211 H11034C. (a) What structural aspect of these 18-carbon fatty acids Problems 8885d_c10_367 1/16/04 8:18 AM Page 367 mac76 mac76:385_reb: Chapter 10 Lipids368 can be correlated with the melting point? Provide a molecu- lar explanation for the trend in melting points. (b) Draw all the possible triacylglycerols that can be constructed from glycerol, palmitic acid, and oleic acid. Rank them in order of increasing melting point. (c) Branched-chain fatty acids are found in some bac- terial membrane lipids. Would their presence increase or de- crease the fluidity of the membranes (that is, give them a lower or higher melting point)? Why? 3. Preparation of Béarnaise Sauce During the prepa- ration of béarnaise sauce, egg yolks are incorporated into melted butter to stabilize the sauce and avoid separation. The stabilizing agent in the egg yolks is lecithin (phosphatidyl- choline). Suggest why this works. 4. Hydrophobic and Hydrophilic Components of Mem- brane Lipids A common structural feature of membrane lipids is their amphipathic nature. For example, in phos- phatidylcholine, the two fatty acid chains are hydrophobic and the phosphocholine head group is hydrophilic. For each of the following membrane lipids, name the components that serve as the hydrophobic and hydrophilic units: (a) phos- phatidylethanolamine; (b) sphingomyelin; (c) galactosyl- cerebroside; (d) ganglioside; (e) cholesterol. 5. Alkali Lability of Triacylglycerols A common pro- cedure for cleaning the grease trap in a sink is to add a prod- uct that contains sodium hydroxide. Explain why this works. 6. The Action of Phospholipases The venom of the Eastern diamondback rattler and the Indian co- bra contains phospholipase A 2 , which catalyzes the hydroly- sis of fatty acids at the C-2 position of glycerophospholipids. The phospholipid breakdown product of this reaction is lysolecithin (lecithin is phosphatidylcholine). At high con- centrations, this and other lysophospholipids act as deter- gents, dissolving the membranes of erythrocytes and lysing the cells. Extensive hemolysis may be life-threatening. (a) Detergents are amphipathic. What are the hy- drophilic and hydrophobic portions of lysolecithin? (b) The pain and inflammation caused by a snake bite can be treated with certain steroids. What is the basis of this treatment? (c) Though high levels of phospholipase A 2 can be deadly, this enzyme is necessary for a variety of normal meta- bolic processes. What are these processes? 7. Intracellular Messengers from Phosphatidylinosi- tols When the hormone vasopressin stimulates cleavage of phosphatidylinositol 4,5-bisphosphate by hormone-sensitive phospholipase C, two products are formed. What are they? Compare their properties and their solubilities in water, and predict whether either would diffuse readily through the cytosol. 8. Storage of Fat-Soluble Vitamins In contrast to water-soluble vitamins, which must be a part of our daily diet, fat-soluble vitamins can be stored in the body in amounts suf- ficient for many months. Suggest an explanation for this dif- ference, based on solubilities. 9. Hydrolysis of Lipids Name the products of mild hy- drolysis with dilute NaOH of (a) 1-stearoyl-2,3-dipalmitoyl- glycerol; (b) 1-palmitoyl-2-oleoylphosphatidylcholine. 10. Effect of Polarity on Solubility Rank the following in order of increasing solubility in water: a triacylglycerol, a diacylglycerol, and a monoacylglycerol, all containing only palmitic acid. 11. Chromatographic Separation of Lipids A mixture of lipids is applied to a silica gel column, and the column is then washed with increasingly polar solvents. The mixture consists of phosphatidylserine, phosphatidylethanolamine, phosphatidylcholine, cholesteryl palmitate (a sterol ester), sphingomyelin, palmitate, n-tetradecanol, triacylglycerol, and cholesterol. In what order do you expect the lipids to elute from the column? Explain your reasoning. 12. Identification of Unknown Lipids Johann Thu- dichum, who practiced medicine in London about 100 years ago, also dabbled in lipid chemistry in his spare time. He iso- lated a variety of lipids from neural tissue, and characterized and named many of them. His carefully sealed and labeled vials of isolated lipids were rediscovered many years later. (a) How would you confirm, using techniques not avail- able to Thudichum, that the vials labeled “sphingomyelin” and “cerebroside” actually contain these compounds? (b) How would you distinguish sphingomyelin from phosphatidylcholine by chemical, physical, or enzymatic tests? 13. Ninhydrin to Detect Lipids on TLC Plates Ninhy- drin reacts specifically with primary amines to form a purplish-blue product. A thin-layer chromatogram of rat liver phospholipids is sprayed with ninhydrin, and the color is al- lowed to develop. Which phospholipids can be detected in this way? 8885d_c10_343-368 1/12/04 1:06 PM Page 368 mac76 mac76:385_reb: chapter T he first cell probably came into being when a mem- brane formed, enclosing a small volume of aqueous solution and separating it from the rest of the universe. Membranes define the external boundaries of cells and regulate the molecular traffic across that boundary (Fig. 11–1); in eukaryotic cells, they divide the internal space into discrete compartments to segregate processes and components. They organize complex reaction se- quences and are central to both biological energy con- servation and cell-to-cell communication. The biological activities of membranes flow from their remarkable physical properties. Membranes are flexible, self-sealing, and selectively permeable to polar solutes. Their flexi- bility permits the shape changes that accompany cell growth and movement (such as amoeboid movement). With their ability to break and reseal, two membranes can fuse, as in exocytosis, or a single membrane-enclosed compartment can undergo fission to yield two sealed compartments, as in endocytosis or cell division, without creating gross leaks through cellular surfaces. Because membranes are selectively permeable, they retain certain compounds and ions within cells and within specific cel- lular compartments, while excluding others. Membranes are not merely passive barriers. They in- clude an array of proteins specialized for promoting or catalyzing various cellular processes. At the cell surface, transporters move specific organic solutes and inorganic ions across the membrane; receptors sense extracellular signals and trigger molecular changes in the cell; adhe- sion molecules hold neighboring cells together. Within the cell, membranes organize cellular processes such as the synthesis of lipids and certain proteins, and the en- ergy transductions in mitochondria and chloroplasts. Because membranes consist of just two layers of mole- cules, they are very thin—essentially two-dimensional. Intermolecular collisions are far more probable in this two-dimensional space than in three-dimensional space, so the efficiency of enzyme-catalyzed processes organ- ized within membranes is vastly increased. 11 369 BIOLOGICAL MEMBRANES AND TRANSPORT Good fences make good neighbors. —Robert Frost, “Mending Wall,” in North of Boston, 1914 11.1 The Composition and Architecture of Membranes 370 11.2 Membrane Dynamics 380 11.3 Solute Transport across Membranes 389 Membrane bilayer FIGURE 11–1 Biological membranes. Viewed in cross section, all cell membranes share a characteristic trilaminar appearance. When an erythrocyte is stained with osmium tetroxide and viewed with an elec- tron microscope, the plasma membrane appears as a three-layer struc- ture, 5 to 8 nm (50 to 80 ?) thick. The trilaminar image consists of two electron-dense layers (the osmium, bound to the inner and outer surfaces of the membrane) separated by a less dense central region. 8885d_c11_369-420 2/7/04 6:58 AM Page 369 mac76 mac76:385_reb: In this chapter we first describe the composition of cellular membranes and their chemical architecture— the molecular structures that underlie their biological functions. Next, we consider the remarkable dynamic features of membranes, in which lipids and proteins move relative to each other. Cell adhesion, endocytosis, and the membrane fusion accompanying neurotrans- mitter secretion illustrate the dynamic role of membrane proteins. We then turn to the protein-mediated passage of solutes across membranes via transporters and ion channels. In later chapters we discuss the role of mem- branes in signal transduction (Chapters 12 and 23), energy transduction (Chapter 19), lipid synthesis (Chapter 21), and protein synthesis (Chapter 27). 11.1 The Composition and Architecture of Membranes One approach to understanding membrane function is to study membrane composition—to determine, for ex- ample, which components are common to all mem- branes and which are unique to membranes with specific functions. So before describing membrane structure and function we consider the molecular com- ponents of membranes: proteins and polar lipids, which account for almost all the mass of biological membranes, and carbohydrates, present as part of glycoproteins and glycolipids. Each Type of Membrane Has Characteristic Lipids and Proteins The relative proportions of protein and lipid vary with the type of membrane (Table 11–1), reflecting the di- versity of biological roles. For example, certain neurons have a myelin sheath, an extended plasma membrane that wraps around the cell many times and acts as a pas- sive electrical insulator. The myelin sheath consists primarily of lipids, whereas the plasma membranes of bacteria and the membranes of mitochondria and chloroplasts, the sites of many enzyme-catalyzed processes, contain more protein than lipid (in mass per total mass). For studies of membrane composition, the first task is to isolate a selected membrane. When eukaryotic cells are subjected to mechanical shear, their plasma mem- branes are torn and fragmented, releasing cytoplasmic components and membrane-bounded organelles such as mitochondria, chloroplasts, lysosomes, and nuclei. Plasma membrane fragments and intact organelles can be isolated by centrifugal techniques described in Chapter 1 (see Fig. 1–8). Chemical analyses of membranes isolated from var- ious sources reveal certain common properties. Each kingdom, each species, each tissue or cell type, and the organelles of each cell type have a characteristic set of membrane lipids. Plasma membranes, for example, are enriched in cholesterol and contain no detectable cardiolipin (Fig. 11–2); in the inner mitochondrial mem- brane of the hepatocyte, this distribution is reversed: very low cholesterol and high cardiolipin. Cardiolipin is essential to the function of certain proteins of the inner mitochondrial membrane. Cells clearly have mecha- nisms to control the kinds and amounts of membrane lipids they synthesize and to target specific lipids to particular organelles. In many cases, we can surmise the adaptive advantages of distinct combinations of mem- brane lipids; in other cases, the functional significance of these combinations is as yet unknown. The protein composition of membranes from dif- ferent sources varies even more widely than their lipid composition, reflecting functional specialization. In a rod cell of the vertebrate retina, one portion of the cell is highly specialized for the reception of light; more than 90% of the plasma membrane protein in this region is the light-absorbing glycoprotein rhodopsin. The less- specialized plasma membrane of the erythrocyte has about 20 prominent types of proteins as well as scores of minor ones; many of these are transporters, each moving a specific solute across the membrane. The plasma membrane of Escherichia coli contains hun- Chapter 11 Biological Membranes and Transport370 TABLE 11–1 Major Components of Plasma Membranes in Various Organisms Components (% by weight) Protein Phospholipid Sterol Sterol type Other lipids Human myelin sheath 30 30 19 Cholesterol Galactolipids, plasmalogens Mouse liver 45 27 25 Cholesterol — Maize leaf 47 26 7 Sitosterol Galactolipids Yeast 52 7 4 Ergosterol Triacylglycerols, steryl esters Paramecium (ciliated protist) 56 40 4 Stigmasterol — E. coli 75 25 0 — — Note: Values do not add up to 100% in every case, because there are components other than protein, phospholipids, and sterol; plants, for example, have high levels of glycolipids. 8885d_c11_369-420 2/7/04 6:58 AM Page 370 mac76 mac76:385_reb: dreds of different proteins, including transporters and many enzymes involved in energy-conserving metabo- lism, lipid synthesis, protein export, and cell division. The outer membrane of E. coli, which encloses the plasma membrane, has a different function (protection) and a different set of proteins. Some membrane proteins are covalently linked to complex arrays of carbohydrate. For example, in gly- cophorin, a glycoprotein of the erythrocyte plasma membrane, 60% of the mass consists of complex oligosaccharide units covalently attached to specific amino acid residues. Ser, Thr, and Asn residues are the most common points of attachment (see Fig. 7–31). At the other end of the scale is rhodopsin of the rod cell plasma membrane, which contains just one hexasac- charide. The sugar moieties of surface glycoproteins influence the folding of the proteins, as well as their sta- bilities and intracellular destinations, and they play a significant role in the specific binding of ligands to gly- coprotein surface receptors (see Fig. 7–37). Some membrane proteins are covalently attached to one or more lipids, which serve as hydrophobic an- chors that hold the proteins to the membrane, as we shall see. All Biological Membranes Share Some Fundamental Properties Membranes are impermeable to most polar or charged solutes, but permeable to nonpolar compounds; they are 5 to 8 nm (50 to 80 ?) thick and appear trilaminar when viewed in cross section with the electron microscope (Fig. 11–1). The combined evidence from electron mi- croscopy and studies of chemical composition, as well as physical studies of permeability and the motion of in- dividual protein and lipid molecules within membranes, led to the development of the fluid mosaic model for the structure of biological membranes (Fig. 11–3). Phospholipids form a bilayer in which the nonpolar re- gions of the lipid molecules in each layer face the core of the bilayer and their polar head groups face outward, interacting with the aqueous phase on either side. Pro- teins are embedded in this bilayer sheet, held by hy- drophobic interactions between the membrane lipids and hydrophobic domains in the proteins. Some proteins protrude from only one side of the membrane; others have domains exposed on both sides. The orientation of proteins in the bilayer is asymmetric, giving the mem- brane “sidedness”: the protein domains exposed on one side of the bilayer are different from those exposed on the other side, reflecting functional asymmetry. The in- dividual lipid and protein units in a membrane form a fluid mosaic with a pattern that, unlike a mosaic of ce- ramic tile and mortar, is free to change constantly. The membrane mosaic is fluid because most of the interac- tions among its components are noncovalent, leaving individual lipid and protein molecules free to move lat- erally in the plane of the membrane. We now look at some of these features of the fluid mosaic model in more detail and consider the experi- mental evidence that supports the basic model but has necessitated its refinement in several ways. A Lipid Bilayer Is the Basic Structural Element of Membranes Glycerophospholipids, sphingolipids, and sterols are vir- tually insoluble in water. When mixed with water, they spontaneously form microscopic lipid aggregates in a phase separate from their aqueous surroundings, clus- tering together, with their hydrophobic moieties in con- tact with each other and their hydrophilic groups in- teracting with the surrounding water. Recall that lipid clustering reduces the amount of hydrophobic surface 11.1 The Composition and Architecture of Membranes 371 FIGURE 11–2 Lipid composition of the plasma membrane and or- ganelle membranes of a rat hepatocyte. The functional specialization of each membrane type is reflected in its unique lipid composition. Cholesterol is prominent in plasma membranes but barely detectable in mitochondrial membranes. Cardiolipin is a major component of the inner mitochondrial membrane but not of the plasma membrane. Phosphatidylserine, phosphatidylinositol, and phosphatidylglycerol are relatively minor components (yellow) of most membranes but serve critical functions; phosphatidylinositol and its derivatives, for exam- ple, are important in signal transductions triggered by hormones. Sphingolipids, phosphatidylcholine, and phosphatidylethanolamine are present in most membranes, but in varying proportions. Glycolipids, which are major components of the chloroplast mem- branes of plants, are virtually absent from animal cells. 0 Plasma Lysosomal Nuclear Rat hepatocyte membrane type Rough ER Smooth ER Golgi Inner mitochondrial Outer mitochondrial 20 40 Percent membrane lipid Cholesterol 60 80 100 Cardiolipin Minor lipids Sphingolipids Phosphatidylcholine Phosphatidylethanolamine 8885d_c11_369-420 2/7/04 6:58 AM Page 371 mac76 mac76:385_reb: Lipid bilayer Peripheral protein Integral protein (single trans- membrane helix) Peripheral protein covalently linked to lipid Oligosaccharide chains of glycoprotein Integral protein (multiple trans- membrane helices) Phospholipid polar heads Sterol Glycolipid Outside Inside FIGURE 11–3 Fluid mosaic model for membrane structure. The fatty acyl chains in the interior of the membrane form a fluid, hydropho- bic region. Integral proteins float in this sea of lipid, held by hydrophobic interactions with their nonpolar amino acid side chains. Both proteins and lipids are free to move laterally in the plane of the bilayer, but movement of either from one face of the bilayer to the other is restricted. The carbohydrate moieties attached to some proteins and lipids of the plasma membrane are exposed on the ex- tracellular surface of the membrane. exposed to water and thus minimizes the number of mol- ecules in the shell of ordered water at the lipid-water interface (see Fig. 2–7), resulting in an increase in en- tropy. Hydrophobic interactions among lipid molecules provide the thermodynamic driving force for the for- mation and maintenance of these clusters. Depending on the precise conditions and the nature of the lipids, three types of lipid aggregates can form when amphipathic lipids are mixed with water (Fig. 11–4). Micelles are spherical structures that contain anywhere from a few dozen to a few thousand amphi- pathic molecules. These molecules are arranged with Chapter 11 Biological Membranes and Transport372 Individual units are wedge-shaped (cross section of head greater than that of side chain) Individual units are cylindrical (cross section of head equals that of side chain) (c) Liposome(b) Bilayer(a) Micelle Aqueous cavity FIGURE 11–4 Amphipathic lipid aggregates that form in water. (a) In micelles, the hydrophobic chains of the fatty acids are sequestered at the core of the sphere. There is virtually no water in the hydrophobic interior. (b) In an open bilayer, all acyl side chains except those at the edges of the sheet are protected from interaction with water. (c) When a two-dimensional bilayer folds on itself, it forms a closed bilayer, a three-dimensional hollow vesicle (liposome) enclosing an aqueous cavity. 8885d_c11_369-420 2/7/04 6:58 AM Page 372 mac76 mac76:385_reb: their hydrophobic regions aggregated in the interior, where water is excluded, and their hydrophilic head groups at the surface, in contact with water. Micelle for- mation is favored when the cross-sectional area of the head group is greater than that of the acyl side chain(s), as in free fatty acids, lysophospholipids (phospholipids lacking one fatty acid), and detergents such as sodium dodecyl sulfate (SDS; p. 92). A second type of lipid aggregate in water is the bilayer, in which two lipid monolayers (leaflets) form a two-dimensional sheet. Bilayer formation occurs most readily when the cross-sectional areas of the head group and acyl side chain(s) are similar, as in glycerophos- pholipids and sphingolipids. The hydrophobic portions in each monolayer, excluded from water, interact with each other. The hydrophilic head groups interact with water at each surface of the bilayer. Because the hy- drophobic regions at its edges (Fig. 11–4b) are tran- siently in contact with water, the bilayer sheet is rela- tively unstable and spontaneously forms a third type of aggregate: it folds back on itself to form a hollow sphere, a vesicle or liposome (Fig. 11–4c). By forming vesicles, bilayers lose their hydrophobic edge regions, achieving maximal stability in their aqueous environment. These bilayer vesicles enclose water, creating a separate aque- ous compartment. It is likely that the precursors to the first living cells resembled liposomes, their aqueous con- tents segregated from the rest of the world by a hy- drophobic shell. Biological membranes are constructed of lipid bi- layers 3 nm (30 ?) thick, with proteins protruding on each side. The hydrocarbon core of the membrane, made up of the OCH 2 O and OCH 3 of the fatty acyl groups, is about as nonpolar as decane, and liposomes formed in the laboratory from pure lipids are essentially imper- meable to polar solutes, as are biological membranes (although the latter, as we shall see, are permeable to solutes for which they have specific transporters). Plasma membrane lipids are asymmetrically dis- tributed between the two monolayers of the bilayer, al- though the asymmetry, unlike that of membrane pro- teins, is not absolute. In the plasma membrane of the erythrocyte, for example, choline-containing lipids (phosphatidylcholine and sphingomyelin) are typically found in the outer (extracellular or exoplasmic) leaflet (Fig. 11–5), whereas phosphatidylserine, phosphatidyl- ethanolamine, and the phosphatidylinositols are much more common in the inner (cytoplasmic) leaflet. Changes in the distribution of lipids between plasma membrane leaflets have biological consequences. For example, only when the phosphatidylserine in the plasma membrane moves into the outer leaflet is a platelet able to play its role in formation of a blood clot. For many other cells types, phosphatidylserine expo- sure on the outer surface marks a cell for destruction by programmed cell death. Peripheral Membrane Proteins Are Easily Solubilized Membrane proteins may be divided operationally into two groups (Fig. 11–6). Integral proteins are very firmly associated with the membrane, removable only by agents that interfere with hydrophobic interactions, such as detergents, organic solvents, or denaturants. Peripheral proteins associate with the membrane through electrostatic interactions and hydrogen bond- ing with the hydrophilic domains of integral proteins and with the polar head groups of membrane lipids. They can be released by relatively mild treatments that interfere with electrostatic interactions or break hy- drogen bonds; a commonly used agent is carbonate at high pH. Peripheral proteins may serve as regulators of membrane-bound enzymes or may limit the mobility of integral proteins by tethering them to intracellular structures. Many Membrane Proteins Span the Lipid Bilayer Membrane protein topology (localization relative to the lipid bilayer) can be determined with reagents that react with protein side chains but cannot cross membranes—polar chemical reagents that react with primary amines of Lys residues, for example, or enzymes 11.1 The Composition and Architecture of Membranes 373 Phosphatidylinositol 4-phosphate Phosphatidylinositol Phosphatidylinositol 4,5-bisphosphate Phosphatidic acid 5 Phosphatidylserine 15 Sphingomyelin 23 Phosphatidylcholine 27 Phosphatidyl- ethanolamine 30 100 Inner monolayer Outer monolayer 0 Distribution in membrane Membrane phospholipid Percent of total membrane phospholipid 100 FIGURE 11–5 Asymmetric distribution of phospholipids between the inner and outer monolayers of the erythrocyte plasma membrane. The distribution of a specific phospholipid is determined by treating the intact cell with phospholipase C, which cannot reach lipids in the inner monolayer (leaflet) but removes the head groups of lipids in the outer monolayer. The proportion of each head group released provides an estimate of the fraction of each lipid in the outer monolayer. 8885d_c11_369-420 2/7/04 6:58 AM Page 373 mac76 mac76:385_reb: like trypsin that cleave proteins but cannot cross the membrane. The human erythrocyte is convenient for such studies because it has no membrane-bounded or- ganelles; the plasma membrane is the only membrane present. If a membrane protein in an intact erythrocyte reacts with a membrane-impermeant reagent, that pro- tein must have at least one domain exposed on the outer (extracellular) face of the membrane. Trypsin is found to cleave extracellular domains but does not affect do- mains buried within the bilayer or exposed on the inner surface only, unless the plasma membrane is broken to make these domains accessible to the enzyme. Experiments with such topology-specific reagents show that the erythrocyte glycoprotein glycophorin spans the plasma membrane. Its amino-terminal domain (bearing the carbohydrate chains) is on the outer sur- face and is cleaved by trypsin. The carboxyl terminus protrudes on the inside of the cell, where it cannot re- act with impermeant reagents. Both the amino-terminal and carboxyl-terminal domains contain many polar or charged amino acid residues and are therefore quite hydrophilic. However, a segment in the center of the protein (residues 75 to 93) contains mainly hydropho- bic amino acid residues, suggesting that glycophorin has a transmembrane segment arranged as shown in Figure 11–7. One further fact may be deduced from the results of experiments with glycophorin: its disposition in the membrane is asymmetric. Similar studies of other mem- brane proteins show that each has a specific orientation Chapter 11 Biological Membranes and Transport374 Peripheral protein Ca 2+ H11545 H11545 H11546H11546 H11545 detergent phospholipase C GPI-linked protein Protein-glycan Integral protein (hydrophobic domain coated with detergent) change in pH; chelating agent; urea; CO 3 2– FIGURE 11–6 Peripheral and integral proteins. Membrane proteins can be operationally distinguished by the conditions required to re- lease them from the membrane. Most peripheral proteins are released by changes in pH or ionic strength, removal of Ca 2H11001 by a chelating agent, or addition of urea or carbonate. Integral proteins are ex- tractable with detergents, which disrupt the hydrophobic interactions with the lipid bilayer and form micelle-like clusters around individual protein molecules. Integral proteins covalently attached to a mem- brane lipid, such as a glycosyl phosphatidylinositol (GPI; see Fig. 11–14), can be released by treatment with phospholipase C. Inside Outside Amino terminus Carboxyl terminus Leu Ser Thr Thr Glu Val Ala Met His Thr Thr Thr Ser Ser Ser Val Ser Lys Ser TyrIle Ser SerGln Thr Asn AspThr HisLys ArgAsp Thr Tyr Ala Ala Thr Pro Arg Ala His Glu Val Ser Glu Ile Ser Val Arg Thr Val Tyr Pro Pro Glu GluGluThr Glu Glu Arg Val Gln Leu Ala His Phe Ser Pro Glu Glu Ile Thr Leu Ile Ile Phe Gly Val Met Ala Gly Val Ile Gly Thr Ile Leu Leu Ile Ser Tyr Gly Ile ArgArg LeuIle Lys Lys Ser Pro Ser Asp Val Lys Pro Leu Pro Ser Pro Asp Val Thr Asp Pro Leu Ser Ser Val Glu Ile Glu AsnPro Glu Thr SerAsp Gln His 1 60 74 95 131 FIGURE 11–7 Transbilayer disposition of glycophorin in an erythrocyte. One hydrophilic domain, containing all the sugar residues, is on the outer surface, and another hydrophilic domain pro- trudes from the inner face of the membrane. Each red hexagon rep- resents a tetrasaccharide (containing two Neu5Ac (sialic acid), Gal, and GalNAc) O-linked to a Ser or Thr residue; the blue hexagon rep- resents an oligosaccharide chain N-linked to an Asn residue. The rel- ative size of the oligosaccharide units is larger than shown here. A segment of 19 hydrophobic residues (residues 75 to 93) forms an H9251 helix that traverses the membrane bilayer (see Fig. 11–11a). The seg- ment from residues 64 to 74 has some hydrophobic residues and prob- ably penetrates into the outer face of the lipid bilayer, as shown. 8885d_c11_369-420 2/7/04 6:58 AM Page 374 mac76 mac76:385_reb: in the bilayer; one domain of a transmembrane protein always faces out, the other always faces in. Further- more, glycoproteins of the plasma membrane are in- variably situated with their sugar residues on the outer surface of the cell. As we shall see, the asymmetric arrangement of membrane proteins results in functional asymmetry. All the molecules of a given ion pump, for example, have the same orientation in the membrane and therefore pump in the same direction. Integral Proteins Are Held in the Membrane by Hydrophobic Interactions with Lipids The firm attachment of integral proteins to membranes is the result of hydrophobic interactions between mem- brane lipids and hydrophobic domains of the protein. Some proteins have a single hydrophobic sequence in the middle (as in glycophorin) or at the amino or car- boxyl terminus. Others have multiple hydrophobic se- quences, each of which, when in the H9251-helical confor- mation, is long enough to span the lipid bilayer (Fig. 11–8). The same techniques used to determine the three-dimensional structures of soluble proteins can, in principle, be applied to membrane proteins. In practice, however, membrane proteins have until recently proved difficult to crystallize. New techniques are overcoming this obstacle, and crystallographic structures of mem- brane proteins are regularly becoming available, yield- ing deep insights into membrane events at the molecu- lar level. One of the best-studied membrane-spanning pro- teins, bacteriorhodopsin, has seven very hydrophobic in- ternal sequences and crosses the lipid bilayer seven times. Bacteriorhodopsin is a light-driven proton pump densely packed in regular arrays in the purple mem- brane of the bacterium Halobacterium salinarum. X-ray crystallography reveals a structure with seven H9251- helical segments, each traversing the lipid bilayer, con- nected by nonhelical loops at the inner and outer face of the membrane (Fig. 11–9). In the amino acid se- quence of bacteriorhodopsin, seven segments of about 20 hydrophobic residues can be identified, each seg- ment just long enough to form an H9251 helix that spans the bilayer. Hydrophobic interactions between the nonpolar amino acids and the fatty acyl groups of the membrane lipids firmly anchor the protein in the membrane. The seven helices are clustered together and oriented not quite perpendicular to the bilayer plane, providing a transmembrane pathway for proton movement. As we shall see in Chapter 12, this pattern of seven hy- drophobic membrane-spanning helices is a common mo- tif in membrane proteins involved in signal reception. The photosynthetic reaction center of a purple bac- terium was the first membrane protein structure solved by crystallography. Although a more complex membrane protein than bacteriorhodopsin, it is constructed on the same principles. The reaction center has four protein subunits, three of which contain H9251-helical segments that span the membrane (Fig. 11–10). These segments are rich in nonpolar amino acids, their hydrophobic side chains oriented toward the outside of the molecule where they interact with membrane lipids. The archi- tecture of the reaction center protein is therefore the inverse of that seen in most water-soluble proteins, in 11.1 The Composition and Architecture of Membranes 375 Inside Outside Type I Type II Type III Type IV Type VI Type V – OOC NH 3 COO – H 3 N + + FIGURE 11–8 Integral membrane proteins. For known proteins of the plasma membrane, the spatial relationships of protein domains to the lipid bilayer fall into six categories. Types I and II have only one trans- membrane helix; the amino-terminal domain is outside the cell in type I proteins and inside in type II. Type III proteins have multiple trans- membrane helices in a single polypeptide. In type IV proteins, trans- membrane domains of several different polypeptides assemble to form a channel through the membrane. Type V proteins are held to the bilayer primarily by covalently linked lipids (see Fig. 11–14), and type VI proteins have both transmembrane helices and lipid (GPI) anchors. In this figure, and in figures throughout the book, we represent transmembrane protein segments in their most likely conformations: as H9251 helices of six to seven turns. Sometimes these helices are shown simply as cylinders. As relatively few membrane protein structures have been deduced by x-ray crystallography, our representation of the extramembrane domains is arbitrary and not necessarily to scale. 8885d_c11_369-420 2/7/04 6:58 AM Page 375 mac76 mac76:385_reb: which hydrophobic residues are buried within the protein core and hydrophilic residues are on the surface (recall the structures of myoglobin and hemo- globin, for example). In Chapter 19 we will encounter several complex membrane proteins having multiple transmembrane helical segments in which hydrophobic chains are positioned to interact with the lipid bilayer. The Topology of an Integral Membrane Protein Can Be Predicted from Its Sequence Determination of the three-dimensional structure of a membrane protein, or its topology, is generally much more difficult than determining its amino acid sequence, which can be accomplished by sequencing the protein or its gene. Thousands of sequences are known for mem- brane proteins, but relatively few three-dimensional structures have been established by crystallography or NMR spectroscopy. The presence of unbroken sequences of more than 20 hydrophobic residues in a membrane protein is commonly taken as evidence that these se- quences traverse the lipid bilayer, acting as hydropho- bic anchors or forming transmembrane channels. Virtu- ally all integral proteins have at least one such sequence. Application of this logic to entire genomic sequences leads to the conclusion that in many species, 10% to 20% of all proteins are integral membrane proteins. What can we predict about the secondary structure of the membrane-spanning portions of integral proteins? An H9251-helical sequence of 20 to 25 residues is just long enough to span the thickness (30 ?) of the lipid bilayer (recall that the length of an H9251 helix is 1.5 ? (0.15 nm) per amino acid residue). A polypeptide chain sur- rounded by lipids, having no water molecules with which to hydrogen-bond, will tend to form H9251 helices or H9252 sheets, in which intrachain hydrogen bonding is maxi- mized. If the side chains of all amino acids in a helix are nonpolar, hydrophobic interactions with the surround- ing lipids further stabilize the helix. Several simple methods of analyzing amino acid se- quences yield reasonably accurate predictions of sec- ondary structure for transmembrane proteins. The rel- ative polarity of each amino acid has been determined experimentally by measuring the free-energy change ac- companying the movement of that amino acid side chain from a hydrophobic solvent into water. This free energy of transfer ranges from very exergonic for charged or polar residues to very endergonic for amino acids with aromatic or aliphatic hydrocarbon side chains. The overall hydrophobicity of a sequence of amino acids is estimated by summing the free energies of transfer for Chapter 11 Biological Membranes and Transport376 FIGURE 11–9 Bacteriorhodopsin, a membrane-spanning protein. (PDB ID 2AT9) The single polypeptide chain folds into seven hy- drophobic H9251 helices, each of which traverses the lipid bilayer roughly perpendicular to the plane of the membrane. The seven transmem- brane helices are clustered, and the space around and between them is filled with the acyl chains of membrane lipids. The light-absorbing pigment retinal (see Fig. 10–21) is buried deep in the membrane in contact with several of the helical segments (not shown). The helices are colored to correspond with the hydropathy plot in Figure 11–11b. Amino terminus Carboxyl terminus Inside Outside Inside Outside FIGURE 11–10 Three-dimensional structure of the photosynthetic reaction center of Rhodopseudomonas viridis, a purple bacterium. This was the first integral membrane protein to have its atomic struc- ture determined by x-ray diffraction methods (PDB ID 1PRC). Eleven H9251-helical segments from three of the four subunits span the lipid bi- layer, forming a cylinder 45 ? (4.5 nm) long; hydrophobic residues on the exterior of the cylinder interact with lipids of the bilayer. In this ribbon representation, residues that are part of the transmembrane he- lices are shown in yellow. The prosthetic groups (light-absorbing pig- ments and electron carriers; see Fig. 19–45) are red. 8885d_c11_369-420 2/7/04 6:58 AM Page 376 mac76 mac76:385_reb: the residues in the sequence, which yields a hydropa- thy index for that region (see Table 3–1). To scan a polypeptide sequence for potential membrane-spanning segments, an investigator calculates the hydropathy in- dex for successive segments (called windows) of a given size, from 7 to 20 residues. For a window of seven residues, for example, the indices for residues 1 to 7, 2 to 8, 3 to 9, and so on, are plotted as in Figure 11–11 (plotted for the middle residue in each window— residue 4 for residues 1 to 7, for example). A region with more than 20 residues of high hydropathy index is presumed to be a transmembrane segment. When the sequences of membrane proteins of known three- dimensional structure are scanned in this way, we find a reasonably good correspondence between predicted and known membrane-spanning segments. Hydropathy analysis predicts a single hydrophobic helix for gly- cophorin (Fig. 11–11a) and seven transmembrane segments for bacteriorhodopsin (Fig. 11–11b)—in agreement with experimental studies. On the basis of their amino acid sequences and hy- dropathy plots, many of the transport proteins de- scribed in this chapter are believed to have multiple membrane-spanning helical regions—that is, they are type III or type IV integral proteins (Fig. 11–8). When predictions are consistent with chemical studies of protein localization (such as those described above for glycophorin and bacteriorhodopsin), the assumption that hydrophobic regions correspond to membrane- spanning domains is much better justified. A further remarkable feature of many transmem- brane proteins of known structure is the presence of Tyr and Trp residues at the interface between lipid and water (Fig. 11–12). The side chains of these residues apparently serve as membrane interface anchors, able to interact simultaneously with the central lipid phase and the aqueous phases on either side of the membrane. 11.1 The Composition and Architecture of Membranes 377 FIGURE 11–11 Hydropathy plots. Hydropathy index (see Table 3–1) is plotted against residue number for two integral membrane proteins. The hydropathy index for each amino acid residue in a sequence of defined length (called the window) is used to calculate the average hydropathy for the residues in that window. The horizontal axis shows the residue number in the middle of the window. (a) Glycophorin from human erythrocytes has a single hydrophobic sequence between residues 75 and 93 (yellow); compare this with Figure 11–7. (b) Bac- teriorhodopsin, known from independent physical studies to have seven transmembrane helices (see Fig. 11–9), has seven hydrophobic regions. Note, however, that the hydropathy plot is ambiguous in the region of segments 6 and 7. Physical studies have confirmed that this region has two transmembrane segments. Hydrophobic Hydrophilic H110023 0 3 50 100 150 50 100 150 200 200 250 25010 1 2 3 4 5 6 7 10 Hydropathy index (b) Bacteriorhodopsin Residue number Hydropathy index (a) Glycophorin Hydrophobic Hydrophilic H110023 0 3 0 50 100 0 50 100 130 130 Residue number K + channel Maltoporin Outer membrane phospholipase A OmpX Phosphoporin E FIGURE 11–12 Tyr and Trp residues of membrane proteins cluster- ing at the water-lipid interface. The detailed structures of these five integral membrane proteins are known from crystallographic studies. The K H11001 channel (PDB ID 1BL8) is from the bacterium Streptomyces lividans (see Fig. 11–48); maltoporin (PDB ID 1AF6), outer membrane phospholipase A (PDB ID 1QD5), OmpX (PDB ID 1QJ9), and phos- phoporin E (PDB ID 1PHO) are proteins of the outer membrane of E. coli. Residues of Tyr (orange) and Trp (red) are found predominantly where the nonpolar region of acyl chains meets the polar head group region. Charged residues (Lys, Arg, Glu, Asp) are shown in blue; they are found almost exclusively in the aqueous phases. 8885d_c11_369-420 2/7/04 6:58 AM Page 377 mac76 mac76:385_reb: FepA OmpLA Maltoporin TolC Top view a-Hemolysin toxin The hydrophobic domains of some integral mem- brane proteins penetrate only one leaflet of the bilayer. Cyclooxygenase, the target of aspirin action, is an ex- ample; its hydrophobic helices do not span the whole membrane but interact strongly with the acyl groups on one side of the bilayer (see Box 21–2, Fig. 1a). Not all integral membrane proteins are composed of transmembrane H9251 helices. Another structural motif common in membrane proteins is the H9252 barrel (see Fig. 4–20d), in which 20 or more transmembrane segments form H9252 sheets that line a cylinder (Fig. 11–13). The same factors that favor H9251-helix formation in the hydrophobic interior of a lipid bilayer also stabilize H9252 barrels. When no water molecules are available to hydrogen-bond with the carbonyl oxygen and nitrogen of the peptide bond, maximal intrachain hydrogen bonding gives the most stable conformation. Planar H9252 sheets do not maximize these interactions and are generally not found in the membrane interior; H9252 barrels do allow all possible hydrogen bonds and are apparently common among membrane proteins. Porins, proteins that allow certain polar solutes to cross the outer membrane of gram- negative bacteria such as E. coli, have many-stranded H9252 barrels lining the polar transmembrane passage. A polypeptide is more extended in the H9252 confor- mation than in an H9251 helix; just seven to nine residues of H9252 conformation are needed to span a membrane. Recall that in the H9252 conformation, alternating side chains project above and below the sheet (see Fig. 4–7). In H9252 strands of membrane proteins, every second residue in the membrane-spanning segment is hydrophobic and in- teracts with the lipid bilayer; aromatic side chains are commonly found at the lipid-protein interface. The other residues may or may not be hydrophilic. The hy- dropathy plot is not useful in predicting transmembrane segments for proteins with H9252 barrel motifs, but as the database of known H9252 barrel motifs increases, sequence- based predictions of transmembrane H9252 conformations have become feasible. For example, a number of outer membrane proteins of gram-negative bacteria (Fig. 11–13) have been correctly predicted, by sequence analysis, to contain H9252 barrels. Covalently Attached Lipids Anchor Some Membrane Proteins Some membrane proteins contain one or more cova- lently linked lipids of several types: long-chain fatty acids, isoprenoids, sterols, or glycosylated derivatives of phosphatidylinositol, GPI (Fig. 11–14). The attached lipid provides a hydrophobic anchor that inserts into the lipid bilayer and holds the protein at the membrane sur- face. The strength of the hydrophobic interaction be- tween a bilayer and a single hydrocarbon chain linked to a protein is barely enough to anchor the protein se- curely, but many proteins have more than one attached Chapter 11 Biological Membranes and Transport378 FIGURE 11–13 Membrane proteins with H9252-barrel structure. Five ex- amples are shown, viewed in the plane of the membrane; The first four are from the E. coli outer membrane. FepA (PDB ID 1FEP), in- volved in iron uptake, has 22 membrane-spanning H9252 strands. OmpLA (derived from PDB ID 1QD5), a phospholipase, is a 12-stranded H9252 barrel that exists as a dimer in the membrane. Maltoporin (derived from PDB ID 1MAL), a maltose transporter, is a trimer, each monomer constructed of 16 H9252 strands. TolC (PDB ID 1EK9), another transporter, has three separate subunits, each contributing four H9252 strands in this 12-stranded barrel. The Staphylococcus aureus H9251-hemolysin toxin (PDB ID 7AHL; top view below) is composed of seven identical sub- units, each contributing one hairpin-shaped pair of H9252 strands to the 14-stranded barrel. 8885d_c11_369-420 2/7/04 6:58 AM Page 378 mac76 mac76:385_reb: lipid moiety. Other interactions, such as ionic attractions between positively charged Lys residues in the protein and negatively charged lipid head groups, probably con- tribute to the stability of the attachment. The associa- tion of these lipid-linked proteins with the membrane is certainly weaker than that for integral membrane proteins and is, in at least some cases, reversible. But treatment with alkaline carbonate does not release GPI-linked proteins, which are therefore, by the work- ing definition, integral proteins. Beyond merely anchoring a protein to the mem- brane, the attached lipid may have a specific role. In the plasma membrane, proteins with GPI anchors are ex- clusively on the outer face and are confined within clus- ters, as we shall see below, whereas other types of lipid- linked proteins (with farnesyl or geranylgeranyl groups attached; Fig. 11–14) are exclusively on the inner face. Attachment of a specific lipid to a newly synthesized membrane protein therefore has a targeting function, directing the protein to its correct membrane location. 11.1 The Composition and Architecture of Membranes 379 Inside Outside Palmitoyl group on internal Cys (or Ser) COO H11002 CH 2 N H O O C O O NH C N-Myristoyl group on amino-terminal Gly Farnesyl (or geranylgeranyl) group on carboxyl-terminal Cys GPI anchor on carboxyl terminus CH 2 CH 3 O CH O COO H11002 NH 3 CH 2 S O C C C C S Cys H11001 O Inositol O GlcNAc CH 2 CH 2 CH 2 CH 2 C O NH Man O CH 2 O O H11002 O P O H11002 O O O P Man Man Man H11001 NH 3 H11001 NH 3 FIGURE 11–14 Lipid-linked membrane proteins. Covalently attached lipids anchor membrane proteins to the lipid bilayer. A palmitoyl group is shown attached by thioester linkage to a Cys residue; an N-myristoyl group is generally attached to an amino-terminal Gly; the farnesyl and geranylgeranyl groups attached to carboxyl-terminal Cys residues are isoprenoids of 15 and 20 carbons, respectively. These three lipid-protein assemblies are found only on the inner face of the plasma membrane. Glycosyl phosphatidylinositol (GPI) anchors are derivatives of phosphatidylinositol in which the inositol bears a short oligosaccharide covalently joined to the carboxyl- terminal residue of a protein through phosphoethanolamine. GPI-linked proteins are always on the extracellular face of the plasma membrane. 8885d_c11_369-420 2/7/04 6:58 AM Page 379 mac76 mac76:385_reb: SUMMARY 11.1 The Composition and Architecture of Membranes ■ Biological membranes define cellular boundaries, divide cells into discrete compartments, organize complex reaction sequences, and act in signal reception and energy transformations. ■ Membranes are composed of lipids and proteins in varying combinations particular to each species, cell type, and organelle. The fluid mosaic model describes features common to all biological membranes. The lipid bilayer is the basic structural unit. Fatty acyl chains of phospholipids and the steroid nucleus of sterols are oriented toward the interior of the bilayer; their hydrophobic interactions stabilize the bilayer but give it flexibility. ■ Peripheral proteins are loosely associated with the membrane through electrostatic interactions and hydrogen bonds or by covalently attached lipid anchors. Integral proteins associate firmly with membranes by hydrophobic interactions between the lipid bilayer and their nonpolar amino acid side chains, which are oriented toward the outside of the protein molecule. ■ Some membrane proteins span the lipid bilayer several times, with hydrophobic sequences of about 20 amino acid residues forming transmembrane H9251 helices. Detection of such hydrophobic sequences in proteins can be used to predict their secondary structure and transmembrane disposition. Multistranded H9252 barrels are also common in integral membrane proteins. Tyr and Trp residues of transmembrane proteins are commonly found at the lipid-water interface. ■ The lipids and proteins of membranes are inserted into the bilayer with specific sidedness; thus membranes are structurally and function- ally asymmetric. Many membrane proteins contain covalently attached oligosaccharides. Plasma membrane glycoproteins are always oriented with the carbohydrate-bearing domain on the extracellular surface. 11.2 Membrane Dynamics One remarkable feature of all biological membranes is their flexibility—their ability to change shape without losing their integrity and becoming leaky. The basis for this property is the noncovalent interactions among lipids in the bilayer and the motions allowed to individual lipids because they are not covalently anchored to one another. We turn now to the dynamics of membranes: the motions that occur and the transient structures al- lowed by these motions. Acyl Groups in the Bilayer Interior Are Ordered to Varying Degrees Although the lipid bilayer structure is quite stable, its individual phospholipid and sterol molecules have some freedom of motion (Fig. 11–15). The structure and flex- ibility of the lipid bilayer depend on temperature and on the kinds of lipids present. At relatively low temper- atures, the lipids in a bilayer form a semisolid gel phase, in which all types of motion of individual lipid molecules are strongly constrained; the bilayer is paracrystalline (Fig. 11–15a). At relatively high temperatures, individual hydrocarbon chains of fatty acids are in constant motion produced by rotation about the carbon–carbon bonds of the long acyl side chains. In this liquid-disordered state, or fluid state (Fig. 11–15b), the interior of the bilayer is more fluid than solid and the bilayer is like a sea of constantly moving lipid. At intermediate temper- atures, the lipids exist in a liquid-ordered state; there is less thermal motion in the acyl chains of the lipid bi- layer, but lateral movement in the plane of the bilayer still takes place. These differences in bilayer state are easily observed in liposomes composed of a single lipid, Chapter 11 Biological Membranes and Transport380 Heat produces thermal motion of side chains (gel → fluid transition) (a) Paracrystalline state (gel) (b) Fluid state FIGURE 11–15 Two states of bilayer lipids. (a) In the paracrystalline state, or gel phase, polar head groups are uniformly arrayed at the surface, and the acyl chains are nearly motionless and packed with regular geometry; (b) in the liquid disordered state, or fluid state, acyl chains undergo much thermal motion and have no regular organiza- tion. Intermediate between these extremes is the liquid-ordered state, in which individual phospholipid molecules can diffuse laterally but the acyl groups remain extended and more or less ordered. 8885d_c11_380 2/11/04 12:13 PM Page 380 mac76 mac76:385_reb: but biological membranes contain many lipids with a va- riety of fatty acyl chains and thus do not show sharp phase changes with temperature. At temperatures in the physiological range (about 20 to 40 H11034C), long-chain saturated fatty acids (such as 16:0 and 18:0) pack well into a liquid-ordered array, but the kinks in unsaturated fatty acids (see Fig. 10–1) in- terfere with this packing, favoring the liquid-disordered state. Shorter-chain fatty acyl groups have the same ef- fect. The sterol content of a membrane (which varies greatly with organism and organelle; Table 11–1) is an- other important determinant of lipid state. The rigid pla- nar structure of the steroid nucleus, inserted between fatty acyl side chains, reduces the freedom of neigh- boring fatty acyl chains to move by rotation about their carbon–carbon bonds, forcing acyl chains into their fully extended conformation. The presence of sterols there- fore reduces the fluidity in the core of the bilayer, thus favoring the liquid-ordered phase, and increases the thickness of the lipid leaflet (as described below). Cells regulate their lipid composition to achieve a constant membrane fluidity under various growth conditions. For example, bacteria synthesize more un- saturated fatty acids and fewer saturated ones when cul- tured at low temperatures than when cultured at higher temperatures (Table 11–2). As a result of this adjust- ment in lipid composition, membranes of bacteria cultured at high or low temperatures have about the same degree of fluidity. Transbilayer Movement of Lipids Requires Catalysis At physiological temperature, transbilayer—or “flip- flop”—diffusion of a lipid molecule from one leaflet of the bilayer to the other (Fig. 11–16a) occurs very slowly if at all in most membranes. Transbilayer movement requires that a polar or charged head group leave its 11.2 Membrane Dynamics 381 Percentage of total fatty acids * 10 H11034C 20 H11034C 30 H11034C 40 H11034C Myristic acid (14:0) 4 4 4 8 Palmitic acid (16:0) 18 25 29 48 Palmitoleic acid (16:1) 26 24 23 9 Oleic acid (18:1) 38 34 30 12 Hydroxymyristic acid 13 10 10 8 Ratio of unsaturated to saturated ? 2.9 2.0 1.6 0.38 Source: Data from Marr, A.G. & Ingraham, J.L. (1962) Effect of temperature on the composition of fatty acids in Escherichia coli. J. Bacteriol. 84, 1260. * The exact fatty acid composition depends not only on growth temperature but on growth stage and growth medium composition. ? Ratios calculated as the total percentage of 16:1 plus 18:1 divided by the total percentage of 14:0 plus 16:0. Hydroxymyristic acid was omitted from this calculation. TABLE 11–2 Fatty Acid Composition of E. coli Cells Cultured at Different Temperatures Flippase (a) Uncatalyzed transverse (“flip-flop”) diffusion (b) Transverse diffusion catalyzed by flippase (c) Uncatalyzed lateral diffusion fast (t in seconds) very fast (1 mm/s) very slow (t in days) 1 2 1 2 FIGURE 11–16 Motion of single phospholipids in a bilayer. (a) Move- ment from one leaflet to the other is very slow, unless (b) catalyzed by a flippase; in contrast, lateral diffusion within the leaflet (c) is very rapid and requires no protein catalysis. 8885d_c11_369-420 2/7/04 6:58 AM Page 381 mac76 mac76:385_reb: Cell Fluorescent probe on lipids React cell with fluorescent probe to label lipids View surface with fluorescence microscope Measure rate of fluorescence return With time, unbleached phospholipids diffuse into bleached area Intense laser beam bleaches small area aqueous environment and move into the hydrophobic interior of the bilayer, a process with a large, positive free-energy change. There are, however, situations in which such movement is essential. For example, during synthesis of the bacterial plasma membrane, phospho- lipids are produced on the inside surface of the mem- brane and must undergo flip-flop diffusion to enter the outer leaflet of the bilayer. Similar transbilayer diffusion must also take place in eukaryotic cells as membrane lipids synthesized in one organelle move from the inner to the outer leaflet and into other organelles. A family of proteins, the flippases (Fig. 11–16b), facilitates flip- flop diffusion, providing a transmembrane path that is energetically more favorable and much faster than the uncatalyzed movement. Lipids and Proteins Diffuse Laterally in the Bilayer Individual lipid molecules can move laterally in the plane of the membrane by changing places with neighboring lipid molecules (Fig. 11–16c). A molecule in one mono- layer, or leaflet, of the lipid bilayer—the outer leaflet of the erythrocyte plasma membrane, for example—can diffuse laterally so fast that it circumnavigates the ery- throcyte in seconds. This rapid lateral diffusion within the plane of the bilayer tends to randomize the posi- tions of individual molecules in a few seconds. Lateral diffusion can be shown experimentally by attaching fluorescent probes to the head groups of lipids and using fluorescence microscopy to follow the probes over time (Fig. 11–17). In one technique, a small region (5 H9262m 2 ) of a cell surface with fluorescence-tagged lipids is bleached by intense laser radiation so that the irradi- ated patch no longer fluoresces when viewed in the much dimmer light of the fluorescence microscope. However, within milliseconds, the region recovers its fluo- rescence as unbleached lipid molecules diffuse into the bleached patch and bleached lipid molecules diffuse away from it. The rate of f luorescence recovery after photobleaching, or FRAP, is a measure of the rate of lateral diffusion of the lipids. Using the FRAP technique, Chapter 11 Biological Membranes and Transport382 FIGURE 11–17 Measurement of lateral diffusion rates of lipids by fluorescence recovery after photobleaching (FRAP). The lipids in the outer leaflet of the plasma membrane are labeled by reaction with a membrane-impermeant fluorescent probe (red), so the surface is uni- formly labeled when viewed with a fluorescence microscope. A small area is bleached by irradiation with an intense laser beam, leaving that area nonfluorescent. With the passage of time, labeled lipid mol- ecules diffuse into the bleached region, and it again becomes fluo- rescent. From the time course of fluorescence return to this area, the diffusion coefficient for the labeled lipid is determined. The rates are typically high; a lipid moving at this speed could circumnavigate E. coli in one second. (The FRAP method can also be used to meas- ure the lateral diffusion of membrane proteins.) researchers have shown that some membrane lipids dif- fuse laterally by up to 1 H9262m/s. Another technique, single particle tracking, allows one to follow the movement of a single lipid molecule in the plasma membrane on a much shorter time scale. Results from these studies confirm the rapid lateral diffusion within small, discrete regions of the cell sur- 8885d_c11_369-420 2/7/04 6:58 AM Page 382 mac76 mac76:385_reb: face and show that movement from one such region to a nearby region is inhibited; lipids behave as though corralled by fences that they can occasionally jump (Fig. 11–18). Many membrane proteins seem to be afloat in a sea of lipids. Like membrane lipids, these proteins are free to diffuse laterally in the plane of the bilayer and are in constant motion, as shown by the FRAP technique with fluorescence-tagged surface proteins. Some membrane proteins associate to form large aggregates (“patches”) on the surface of a cell or organelle in which individual protein molecules do not move relative to one another; for example, acetylcholine receptors (see Fig. 11–51) form dense patches on neuron plasma membranes at synapses. Other membrane proteins are anchored to internal structures that prevent their free diffusion. In the erythrocyte membrane, both glycophorin and the chloride-bicarbonate exchanger (p. 395) are tethered to spectrin, a filamentous cytoskeletal protein (Fig. 11–19). One possible explanation for the pattern of lat- eral diffusion of lipid molecules shown in Figure 11–18 is that membrane proteins immobilized by their associ- ation with spectrin are the “fences” that define the re- gions of relatively unrestricted lipid motion. Sphingolipids and Cholesterol Cluster Together in Membrane Rafts We have seen that diffusion of membrane lipids from one bilayer leaflet into the other is very slow unless cat- alyzed, and that the different lipid species of the plasma membrane are asymmetrically distributed in the two leaflets of the bilayer (Fig. 11–5). Even within a single leaflet, the lipid distribution is not random. Glycosphin- golipids (cerebrosides and gangliosides), which typically contain long-chain saturated fatty acids, form transient clusters in the outer leaflet that largely exclude glycero- phospholipids, which typically contain one unsaturated fatty acyl group and a shorter saturated fatty acyl group. The long, saturated acyl groups of sphingolipids can form more compact, more stable associations with the long ring system of cholesterol than can the shorter, often unsaturated, chains of phospholipids. The cholesterol- sphingolipid microdomains in the outer monolayer of the plasma membrane, visible with atomic-force mi- croscopy (Box 11–1), are slightly thicker and more ordered (less fluid) than neighboring microdomains rich in phospholipids (Fig. 11–20) and are more difficult 11.2 Membrane Dynamics 383 1 μm Finish Start Chloride-bicarbonate exchange proteins Glycophorin Plasma membrane Ankyrin Spectrin Junctional complex (actin) Path of single lipid molecule Outside Inside FIGURE 11–19 Restricted motion of the erythrocyte chloride- bicarbonate exchanger and glycophorin. The proteins span the mem- brane and are tethered to spectrin, a cytoskeletal protein, by another protein, ankyrin, limiting their lateral mobilities. Ankyrin is anchored in the membrane by a covalently bound palmitoyl side chain (see Fig. 11–14). Spectrin, a long, filamentous protein, is cross-linked at junc- tional complexes containing actin. A network of cross-linked spectrin molecules attached to the cytoplasmic face of the plasma membrane stabilizes the membrane against deformation. This network of an- chored membrane proteins may be the “corral” suggested by the ex- periment shown in Figure 11–18; the lipid tracks shown here are con- fined to subregions defined by the tethered membrane proteins. FIGURE 11–18 Hop diffusion of individual lipid molecules. The motion of a single fluorescent lipid molecule in a cell surface is recorded on video by fluorescence microscopy, with a time resolu- tion of 25 μs (equivalent to 40,000 frames/s). The track shown here represents a molecule followed for 56 ms (a total of 2,250 frames); the trace begins in the purple area and continues through blue, green, and orange. The pattern of movement indicates rapid diffusion within a confined region (about 250 nm in diameter, shown by a single color), with occasional hops into an adjoining region. This finding suggests that the lipids are corralled by molecular fences that they occasion- ally jump. 8885d_c11_383 2/11/04 12:25 PM Page 383 mac76 mac76:385_reb: to dissolve with nonionic detergents; they behave like liquid-ordered sphingolipid rafts adrift in a sea of liq- uid-disordered phospholipids. These lipid rafts are remarkably enriched in two classes of integral membrane proteins: those anchored to the membrane by two covalently attached long-chain saturated fatty acids (two palmitoyl groups or a pal- mitoyl and a myristoyl group) and GPI-anchored proteins (Fig. 11–14). Presumably these lipid anchors, like the acyl chains of sphingolipids, form more stable associa- tions with the cholesterol and long acyl groups in rafts than with the surrounding phospholipids. (It is notable Chapter 11 Biological Membranes and Transport384 BOX 11–1 WORKING IN BIOCHEMISTRY Atomic Force Microscopy to Visualize Membrane Proteins In atomic force microscopy (AFM), the sharp tip of a microscopic probe attached to a flexible cantilever is drawn across an uneven surface such as a membrane (Fig. 1). Electrostatic and van der Waals interactions between the tip and the sample produce a force that moves the probe up and down (in the z dimension) as it encounters hills and valleys in the sample. A laser beam reflected from the cantilever detects motions of as little as 1 ?. In one type of atomic force microscope, the force on the probe is held constant (relative to a standard force, on the order of piconewtons) by a feedback circuit that causes the platform holding the sample to rise or fall to keep the force constant. A se- ries of scans in the x and y dimensions (the plane of the membrane) yields a three-dimensional contour map of the surface with resolution near the atomic scale—0.1 nm in the vertical dimension, 0.5 to 1.0 nm in the lateral dimensions. The membrane rafts shown in Figure 11–20b were visualized by this technique. In favorable cases, AFM can be used to study single membrane protein molecules. Single mole- cules of bacteriorhodopsin in the purple membranes of the bacterium Halobacterium salinarum (see Fig. 11–9) are seen as highly regular structures (Fig. 2a). When a number of images of individual units are superimposed with the help of a computer, the real parts of the image reinforce each other and the noise in individual images is averaged out, yielding a high- resolution image of the protein (inset in Fig. 2a). AFM of purified E. coli aquaporin, reconstituted into lipid bilayers and viewed as if from the outside of a cell, shows the fine details of the protein’s periplas- mic domains (Fig. 2b). And AFM reveals that F o , the proton-driven rotor of the chloroplast ATP synthase (p. 742), is composed of many subunits (14 in Fig. 2c) arranged in a circle. Laser Cantilever Laser light detector (detects cantilever deflection) Platform moves to maintain constant pressure on cantilever tip. Excursions in the z dimension are plotted as a function of x, y. x y z Sample + – 10 nm 2 nm FIGURE 2 FIGURE 1 (a) (b) (c) 8885d_c11_384 2/11/04 12:13 PM Page 384 mac76 mac76:385_reb: that other lipid-linked proteins, those with covalently attached isoprenyl groups such as farnesyl, are not preferentially associated with the outer leaflet of sphingolipid/cholesterol rafts (Fig. 11–20a).) The “raft” and “sea” domains of the plasma membrane are not rigidly separated; membrane proteins can move into and out of lipid rafts on a time scale of seconds. But in the shorter time scale (microseconds) more relevant to many membrane-mediated biochemical processes, many of these proteins reside primarily in a raft. We can estimate the fraction of the cell surface occupied by rafts from the fraction of the plasma mem- 11.2 Membrane Dynamics 385 brane that resists detergent solubilization, which can be as high as 50% in some cases: the rafts cover half of the ocean (Fig. 11–20b). Indirect measurements in cultured fibroblasts suggest a diameter of roughly 50 nm for an individual raft, which corresponds to a patch containing a few thousand sphingolipids and perhaps 10 to 50 membrane proteins. Because most cells express more than 50 different kinds of plasma membrane proteins, it is likely that a single raft contains only a subset of mem- brane proteins and that this segregation of membrane proteins is functionally significant. For a process that involves interaction of two membrane proteins, their presence in a single raft would hugely increase the likelihood of their collision. Certain membrane recep- tors and signaling proteins, for example, appear to be segregated together in membrane rafts. Experiments show that signaling through these proteins can be dis- rupted by manipulations that deplete the plasma mem- brane of cholesterol and destroy lipid rafts. Caveolins Define a Special Class of Membrane Rafts Caveolin is an integral membrane protein with two globular domains connected by a hairpin-shaped hydrophobic domain, which binds the protein to the cy- toplasmic leaflet of the plasma membrane. Three palmi- toyl groups attached to the carboxyl-terminal globular domain further anchor it to the membrane. Caveolin (actually, a family of related caveolins) binds cholesterol in the membrane, and the presence of caveolin forces the associated lipid bilayer to curve inward, forming caveolae (“little caves”) in the surface of the cell (Fig. 11–21). Caveolae are unusual rafts: they involve both leaflets of the bilayer—the cytoplasmic leaflet, from which the caveolin globular domains project, and the ex- oplasmic leaflet, a typical sphingolipid/cholesterol raft with associated GPI-anchored proteins. Caveolae are im- plicated in a variety of cellular functions, including mem- brane trafficking within cells and the transduction of external signals into cellular responses. The receptors for insulin and other growth factors, as well as certain GTP-binding proteins and protein kinases associated with transmembrane signaling, appear to be localized in rafts and perhaps in caveolae. We discuss some possi- ble roles of rafts in signaling in Chapter 12. Certain Integral Proteins Mediate Cell-Cell Interactions and Adhesion Several families of integral proteins in the plasma mem- brane provide specific points of attachment between cells, or between a cell and extracellular matrix proteins. Integrins are heterodimeric proteins (two unlike sub- units, H9251 and H9252) anchored to the plasma membrane by a single hydrophobic transmembrane helix in each sub- unit (Fig. 11–22; see also Fig. 7–30). The large extra- cellular domains of the H9251 and H9252 subunits combine to form a specific binding site for extracellular proteins Prenylated protein Caveolin Cholesterol Raft, enriched in sphingolipids, cholesterol GPI-linked protein Doubly acylated protein Inside Outside (a) Acyl groups (palmitoyl, myristoyl) FIGURE 11–20 Microdomains (rafts) in the plasma membrane. (a) Stable associations of sphingolipids and cholesterol in the outer leaflet produce a microdomain, slightly thicker than other membrane regions, that is enriched with specific types of membrane proteins. GPI-linked proteins are commonly found in the outer leaflet of such rafts, and proteins with one or several covalently attached long-chain acyl groups are common in the inner leaflet. Caveolin is especially common in inwardly curved rafts called caveolae (see Fig. 11–21). Proteins with attached prenyl groups (such as Ras; see Fig. 12–6) tend to be excluded from rafts. (b) The greater thickness of raft regions can be visualized by atomic force microscopy (see Box 11–1). In this view of a membrane region, we can see the rafts protruding from a lipid bilayer ocean; in the rafts, sharp peaks represent GPI-linked proteins. Note that these peaks are found almost exclusively in rafts. (b) 8885d_c11_369-420 2/7/04 6:58 AM Page 385 mac76 mac76:385_reb: such as collagen and fibronectin. As there are 18 dif- ferent H9251 subunits and at least 8 different H9252 subunits, a wide variety of specificities may be generated from var- ious combinations of H9251 and H9252. One common determinant of integrin binding in several extracellular partners of integrins is the sequence Arg–Gly–Asp (RGD). Integrins are not merely adhesives; they serve as receptors and signal transducers, conveying in- formation across the plasma membrane in both direc- tions. Integrins regulate many processes, including platelet aggregation at the site of a wound, tissue repair, the activity of immune cells, and the invasion of tissue by a tumor. Mutation in an integrin gene encoding the H9252 subunit known as CD18 is the cause of leukocyte ad- hesion deficiency in humans, a rare genetic disease in which leukocytes fail to pass out of blood vessels to reach sites of infection (see Fig. 7–33). Infants with a severe defect in CD18 commonly die of infections be- fore the age of two. ■ At least three other families of plasma membrane proteins are also involved in surface adhesion (Fig. 11–22). Cadherins undergo homophilic (“with same kind”) interactions with identical cadherins in an adja- cent cell. Immunoglobulin-like proteins can undergo either homophilic interactions with their identical coun- terparts on another cell or heterophilic interactions with an integrin on a neighboring cell. Selectins have ex- tracellular domains that, in the presence of Ca 2H11001 , bind specific polysaccharides on the surface of an adjacent cell. Selectins are present primarily in the various types of blood cells and in the endothelial cells that line blood vessels (see Fig. 7–33). They are an essential part of the blood-clotting process. Chapter 11 Biological Membranes and Transport386 Caveola Inside Outside Plasma membrane Caveolin dimer (six fatty acyl moieties) FIGURE 11–21 Caveolin forces inward curvature in membranes. The protein caveolin has a central hydrophobic domain and three long- chain acyl groups on each monomeric unit, which hold the molecule to the inside of the plasma membrane. When a number of caveolin dimers are concentrated in a small region (a raft), they force a curva- ture in the lipid bilayer, forming a caveola. FIGURE 11–22 Four examples of integral protein types that function in cell-cell interactions. Integrins consist of H9251 and H9252 transmembrane polypeptides; their extra- cellular domains combine to form binding sites for divalent metal ions and proteins of the extracellular matrix (such as collagen and fibronectin) or for specific surface proteins of other cells. Cadherin has four extracellular Ca 2H11001 -binding domains, the most distal of which contains the site that binds to cadherin on another cell surface. N-CAM (neuronal cell adhesion molecule) is one of a family of immunoglobulin-like proteins that mediate Ca 2H11001 -independent interactions with surface proteins of nearby cells. Selectins bind tightly to carbohydrate moieties in neighboring cells; this binding is Ca 2H11001 -dependent. Outside Inside Plasma membrane Ligand-binding region Adhesive domain Immunoglobulin-like domains SelectinN-CAMCadherinIntegrin Ca 2+2H11001 Ca 2+ Ca 2+ Ca 2+ Ca 2+ 2H11001 Ca 2+2H11001 2H11001 2H11001 2H11001 S S S S S S S S S S b a Lectin domain (binds carbohydrates) 8885d_c11_369-420 2/7/04 6:58 AM Page 386 mac76 mac76:385_reb: Integral proteins play a role in many other cellular processes. They serve as transporters and ion channels (discussed in Section 11.3) and as receptors for hor- mones, neurotransmitters, and growth factors (Chap- ter 12). They are central to oxidative phosphorylation and photosynthesis (Chapter 19) and to cell-cell and antigen-cell recognition in the immune system (Chap- ter 5). Integral proteins are also important players in the membrane fusion that accompanies exocytosis, en- docytosis, and the entry of many types of viruses into host cells. Membrane Fusion Is Central to Many Biological Processes A remarkable feature of the biological membrane is its ability to undergo fusion with another membrane with- out losing its continuity. Although membranes are sta- ble, they are by no means static. Within the eukaryotic endomembrane system (which includes the nuclear membrane, endoplasmic reticulum, Golgi, and various small vesicles), the membranous compartments con- stantly reorganize. Vesicles bud from the endoplasmic reticulum to carry newly synthesized lipids and proteins to other organelles and to the plasma membrane. Exo- cytosis, endocytosis, cell division, fusion of egg and sperm cells, and entry of a membrane-enveloped virus into its host cell all involve membrane reorganization in which the fundamental operation is fusion of two mem- brane segments without loss of continuity (Fig. 11–23). Specific fusion of two membranes requires that (1) they recognize each other; (2) their surfaces become closely apposed, which requires the removal of water mol- ecules normally associated with the polar head groups of lipids; (3) their bilayer structures become locally dis- rupted, resulting in fusion of the outer leaflet of each membrane (hemifusion); and (4) their bilayers fuse to form a single continuous bilayer. Receptor mediated en- docytosis, or regulated secretion, also requires that (5) the fusion process is triggered at the appropriate time or in response to a specific signal. Integral proteins called fusion proteins mediate these events, bringing about specific recognition and a transient local distortion of the bilayer structure that favors membrane fusion. (Note that these fusion proteins are unrelated to the products of two fused genes, also called fusion proteins, discussed in Chapter 9.) Two cases of membrane fusion are especially well studied: the entry into a host cell of an enveloped virus such as influenza virus, and the release of neurotrans- mitters by exocytosis. Both processes involve com- plexes of fusion proteins that undergo dramatic confor- mational changes. The influenza virus is surrounded by a membrane containing, among other proteins, many molecules of the hemagglutination (HA) protein (named for its abil- ity to cause erythrocytes to clump together). The virus enters a host cell by inducing endocytosis, which en- closes the virus in an endosome, a small membrane vesicle with a pH of about 5 (Fig. 11–24). At this pH, a conformational change in the HA protein occurs, ex- posing a sequence within the HA protein called the fusion peptide and enabling the protein to penetrate the endosomal membrane. The endosomal membrane and the viral membrane are now connected through the HA protein. Next, the HA protein bends at its middle to form a hairpin shape, bringing its two ends together. This pulls the two membranes into close apposition and causes fusion of the viral membrane and the endosomal membrane. The HA protein functions as a trimer (Fig. 11–24). In its low-pH form, three HA domains at the closed end of the hairpin twist about each other to form a stable, coiled structure. The fusion process involves an intermediate stage (hemifusion) in which the outer leaflet of the viral membrane is fused with the inner leaflet of the endosomal membrane, while the other two leaflets maintain their continuity. At the point of hemi- fusion, the lipid bilayer must be temporarily disorgan- ized, presumably caused by the HA fusion peptide 11.2 Membrane Dynamics 387 Budding of vesicles from Golgi complex Fusion of endosome and lysosome Viral infection Fusion of sperm and egg Fusion of small vacuoles (plants) Separation of two plasma membranes at cell division Exocytosis Endocytosis FIGURE 11–23 Membrane fusion. The fusion of two membranes is central to a variety of cellular processes involving both organelles and the plasma membrane. 8885d_c11_369-420 2/7/04 6:58 AM Page 387 mac76 mac76:385_reb: Cytosol Secretory vesicle Neurotransmitter molecules v-SNARE t-SNARE SNAP 25 Plasma membrane v-SNARE and t-SNARE bind to each other, zipping up from the amino termini and drawing the two membranes together. Zipping causes curvature and lateral tension on bilayers, favoring hemifusion between outer leaflets and causing formation of an energetically unfavorable void space. Inner leaflets of both membranes come into contact. Complete fusion creates a fusion pore. Pore widens; vesicle contents are released outside cell. Neurotransmitter-filled vesicle approaches plasma membrane. Unstable void space Chapter 11 Biological Membranes and Transport388 FIGURE 11–24 Fusion induced by the hemagglutinin (HA) protein during viral infection. HA protein is exposed on the membrane sur- face of the influenza virus. When the virus moves from the neutral pH of the interstitial fluid to the low-pH compartment (endosome) in the host cell, HA undergoes dramatic shape changes that mediate fusion of the viral and endosomal membranes, releasing the viral contents into the cytoplasm. Host cell Virus Endosome HA protein in pH 7 form has fusion peptides buried. HA protein (trimer) Fusion peptide HA hairpins Low pH of endosome triggers extension of HA fusion peptides, which insert into endosomal membrane. HA folds into hairpins, drawing viral and endosomal membranes together. HA fusion peptide creates local disruption of bilayer, and hemifusion occurs; outer monolayer of virus fuses with inner monolayer of endosome. Complete fusion allows viral contents to enter cytoplasm. Virus binds sialic acid receptors on host surface. Virus triggers endocytosis; becomes enclosed in an endosome. FIGURE 11–25 Fusion during neurotransmitter release at a synapse. The membrane of the secretory vesicle contains the v-SNARE synap- tobrevin (red). The target (plasma) membrane contains the t-SNAREs syntaxin (blue) and SNAP25 (violet). When a local increase in [Ca 2H11001 ] signals release of neurotransmitter, the v-SNARE, SNAP25, and t-SNARE interact, forming a coiled bundle of four H9251 helices, pulling the two membranes together and disrupting the bilayer locally, which leads to membrane fusion and neurotransmitter release. 8885d_c11_369-420 2/7/04 6:58 AM Page 388 mac76 mac76:385_reb: domains. Complete fusion results in release of the viral contents into the host cell cytoplasm. Neurotransmitters are released at synapses when intracellular vesicles loaded with neurotransmitter fuse with the plasma membrane. This process involves a family of proteins called SNARES (Fig. 11–25). SNAREs in the cytoplasmic face of the intracellular vesicles are called v-SNAREs; those in the target membranes with which the vesicles fuse (the plasma membrane during exocytosis) are t-SNAREs. Two other proteins, SNAP25 and NSF, are also involved. During fusion, v- and t-SNAREs bind to each other and undergo a structural change that produces a bundle of long thin rods made up of helices from both v- and t-SNARES and two helices from SNAP25 (Fig. 11–25). The two SNAREs initially interact at their ends, then zip up into the bundle of helices. This structural change pulls the two membranes into contact and ini- tiates the fusion of their lipid bilayers. The complex of SNAREs and SNAP25 is the target of the powerful Clostridium botulinum toxin, a pro- tease that cleaves specific bonds in these proteins, pre- venting neurotransmission and causing the death of the organism. Because of its very high specificity for these proteins, purified botulinum toxin has served as a pow- erful tool for dissecting the mechanism of neurotrans- mitter release in vivo and in vitro. SUMMARY 11.2 Membrane Dynamics ■ Lipids in a biological membrane can exist in liquid-ordered or liquid-disordered states; in the latter state, thermal motion of acyl chains makes the interior of the bilayer fluid. Fluidity is affected by temperature, fatty acid composition, and sterol content. ■ Flip-flop diffusion of lipids between the inner and outer leaflets of a membrane is very slow except when specifically catalyzed by flippases. ■ Lipids and proteins can diffuse laterally within the plane of the membrane, but this mobility is limited by interactions of membrane proteins with internal cytoskeletal structures and interactions of lipids with lipid rafts. One class of lipid rafts consists of sphingolipids and cholesterol with a subset of membrane proteins that are GPI-linked or attached to several long-chain fatty acyl moieties. ■ Caveolin is an integral membrane protein that associates with the inner leaflet of the plasma membrane, forcing it to curve inward to form caveolae, probably involved in membrane transport and signaling. ■ Integrins are transmembrane proteins of the plasma membrane that act both to attach cells to each other and to carry messages between the extracellular matrix and the cytoplasm. ■ Specific proteins mediate the fusion of two membranes, which accompanies processes such as viral invasion and endocytosis and exocytosis. 11.3 Solute Transport across Membranes Every living cell must acquire from its surroundings the raw materials for biosynthesis and for energy produc- tion, and must release to its environment the byprod- ucts of metabolism. A few nonpolar compounds can dissolve in the lipid bilayer and cross the membrane unassisted, but for polar or charged compounds or ions, a membrane protein is essential for transmembrane movement. In some cases a membrane protein simply facilitates the diffusion of a solute down its concentra- tion gradient, but transport often occurs against a gra- dient of concentration, electrical charge, or both, in which case solutes must be “pumped” in a process that requires energy (Fig. 11–26). The energy may come directly from ATP hydrolysis or may be supplied in the form of movement of another solute down its electro- chemical gradient with enough energy to carry another solute up its gradient. Ions may also move across mem- branes via ion channels formed by proteins, or they may be carried across by ionophores, small molecules that mask the charge of the ions and allow them to diffuse through the lipid bilayer. With very few exceptions, the traffic of small molecules across the plasma membrane is mediated by proteins such as transmembrane chan- nels, carriers, or pumps. Within the eukaryotic cell, dif- ferent compartments have different concentrations of metabolic intermediates and products and of ions, and these, too, must move across intracellular membranes in tightly regulated, protein-mediated processes. Passive Transport Is Facilitated by Membrane Proteins When two aqueous compartments containing unequal concentrations of a soluble compound or ion are sepa- rated by a permeable divider (membrane), the solute moves by simple diffusion from the region of higher concentration, through the membrane, to the region of lower concentration, until the two compartments have equal solute concentrations (Fig. 11–27a). When ions of opposite charge are separated by a permeable mem- brane, there is a transmembrane electrical gradient, a membrane potential, V m (expressed in volts or milli- volts). This membrane potential produces a force op- posing ion movements that increase V m and driving ion movements that reduce V m (Fig. 11–27b). Thus the di- rection in which a charged solute tends to move spon- taneously across a membrane depends on both the 11.3 Solute Transport across Membranes 389 8885d_c11_369-420 2/7/04 6:58 AM Page 389 mac76 mac76:385_reb: Chapter 11 Biological Membranes and Transport390 S in S in S out S out S in S in Ion Ion Ion Ion Ion Ion S out S out ATP Facilitated diffusion (down electrochemical gradient) Simple diffusion (nonpolar compounds only, down concentration gradient) Primary active transport (against electrochemical gradient) Secondary active transport (against electrochemical gradient, driven by ion moving down its gradient) Ion channel (down electrochemical gradient; may be gated by a ligand or ion) Ionophore- mediated ion transport (down electrochemical gradient) ADP + P i FIGURE 11–26 Summary of transport types. FIGURE 11–27 Movement of solutes across a permeable membrane. (a) Net movement of electrically neutral solutes is toward the side of lower solute concentration until equilibrium is achieved. The solute concentrations on the left and right sides of the membrane are desig- nated C 1 and C 2 . The rate of transmembrane movement (indicated by the large arrows) is proportional to the concentration gradient, C 1 /C 2 . (b) Net movement of electrically charged solutes is dictated by a com- bination of the electrical potential (V m ) and the chemical concentra- tion difference across the membrane; net ion movement continues un- til this electrochemical potential reaches zero. C 1 >> C 2 Before equilibrium Net flux C 1 C 2 C 1 = C 2 At equilibrium No net flux (a) C 1 C 2 V m > 0 Before equilibrium V m = 0 At equilibrium (b) 8885d_c11_369-420 2/7/04 6:58 AM Page 390 mac76 mac76:385_reb: chemical gradient (the difference in solute concentra- tion) and the electrical gradient (V m ) across the mem- brane. Together, these two factors are referred to as the electrochemical gradient or electrochemical po- tential. This behavior of solutes is in accord with the second law of thermodynamics: molecules tend to spon- taneously assume the distribution of greatest random- ness and lowest energy. To pass through a lipid bilayer, a polar or charged solute must first give up its interactions with the water molecules in its hydration shell, then diffuse about 3 nm (30 ?) through a solvent (lipid) in which it is poorly soluble (Fig. 11–28). The energy used to strip away the hydration shell and to move the polar compound from water into and through lipid is regained as the com- pound leaves the membrane on the other side and is re- hydrated. However, the intermediate stage of trans- membrane passage is a high-energy state comparable to the transition state in an enzyme-catalyzed chemical re- action. In both cases, an activation barrier must be over- come to reach the intermediate stage (Fig. 11–28; com- pare with Fig. 6–3). The energy of activation (H9004G ? ) for translocation of a polar solute across the bilayer is so large that pure lipid bilayers are virtually impermeable to polar and charged species over periods of time rele- vant to cell growth and division. Membrane proteins lower the activation energy for transport of polar compounds and ions by providing an alternative path through the bilayer for specific solutes. Proteins that bring about this facilitated diffusion, or passive transport, are not enzymes in the usual sense; their “substrates” are moved from one compartment to another, but are not chemically altered. Membrane pro- teins that speed the movement of a solute across a mem- brane by facilitating diffusion are called transporters or permeases. Like enzymes, transporters bind their substrates with stereochemical specificity through multiple weak, noncovalent interactions. The negative free-energy change associated with these weak interactions, H9004G binding , counterbalances the positive free-energy change that accompanies loss of the water of hydration from the substrate, H9004G dehydration , thereby lowering H9004G ? for transmembrane passage (Fig. 11–28). Transporters span the lipid bilayer several times, forming a trans- membrane channel lined with hydrophilic amino acid side chains. The channel provides an alternative path for a specific substrate to move across the lipid bilayer without its having to dissolve in the bilayer, further low- ering H9004G ? for transmembrane diffusion. The result is an increase of several orders of magnitude in the rate of transmembrane passage of the substrate. Transporters Can Be Grouped into Superfamilies Based on Their Structures We know from genomic studies that transporters con- stitute a significant fraction of all proteins encoded in the genomes of both simple and complex organisms. There are probably a thousand or more different trans- porters in the human genome. A few hundred trans- porters from various species have been studied with bio- chemical, genetic, and electrophysiological tools, but investigators have determined the three-dimensional structures for only a handful of these. Examination of the many transporter genes reveals obvious sequence similarities among subsets of transporters. And as ex- perience has shown, similar amino acid sequences in proteins generally reflect similar three-dimensional structures and, often, similar mechanisms of action. It is reasonable to hope that by determining the structure and mechanism of action of at least one member of each transporter family, we can learn much about the other members of the family—about their structures, sub- strate specificities, transport rates, and mechanisms of energy coupling. A phylogenetic tree in which proteins are grouped together based on sequence homologies has the potential to tell us much about the transport prop- erties of individual proteins on that tree. When this 11.3 Solute Transport across Membranes 391 Free energy, G ?G simple diffusion ?G ? transport Simple diffusion without transporter Diffusion with transporter Transporter Hydrated solute (b) (a) ? FIGURE 11–28 Energy changes accompanying passage of a hydro- philic solute through the lipid bilayer of a biological membrane. (a) In simple diffusion, removal of the hydration shell is highly endergonic, and the energy of activation (H9004G ? ) for diffusion through the bilayer is very high. (b) A transporter protein reduces the H9004G ? for transmem- brane diffusion of the solute. It does this by forming noncovalent in- teractions with the dehydrated solute to replace the hydrogen bond- ing with water and by providing a hydrophilic transmembrane passageway. 8885d_c11_369-420 2/7/04 6:58 AM Page 391 mac76 mac76:385_reb: phylogeny is combined with knowledge of structure, specificity, or mechanism, we have a very useful and relatively simple representation of the huge group of transporters (Table 11–3). Transporters can usefully be classified into super- families, whose members have considerable similarity of sequence and might therefore be expected to share structural and functional properties. There are two very broad categories of transporters: carriers and channels (Fig. 11–29). Carriers bind their substrates with high stereospecificity, catalyze transport at rates well below the limits of free diffusion, and are saturable in the same sense as are enzymes: there is some substrate concen- tration above which further increases will not produce a greater rate of activity. Channels generally allow transmembrane movement at rates several orders of magnitude greater than those typical of carriers, rates approaching the limit of unhindered diffusion. Channels typically show less stereospecificity than carriers and are usually not saturable. Most channels are oligomeric complexes of several, often identical, subunits, whereas many carriers function as monomeric proteins. The clas- sification as carrier or channel is the broadest distinc- tion among transporters. Within each of these categories Chapter 11 Biological Membranes and Transport392 TABLE 11–3 The Transporter Classification (TC) System 1.A. H9251 Helix type channels 1.A.1. Voltage-gated ion channel VIC superfamily Voltage-gated K H11001 channel 1.A.3. Ryanodine/IP 3 receptor Ca 2H11001 channel 1.A.8. Major intrinsic protein family Aquaporins 1.A.9. Ligand-gated ion channel (LIC) of neurotransmitter receptors Acetylcholine receptor/channel 1.B. H9252 Barrel porins 1.B.1. General bacterial porin (GBP) family 1.C. Pore-forming toxins 1.C.7. Diphtheria toxin family 1.C.18. Mellitin family (bee venoms) 2.A. Porters: uniporters, symporters, and antiporters 2.A.1. Major facilitator superfamily (MFS) Lactose transporter/permease of E. coli 2.A.1.1. Sugar porter family GLUT1 glucose transporter of erythrocyte 2.A.1.9. P i -H H11001 symporter 2.A.12. ATP-ADP antiporter (AAA) family 2.A.13. C 4 -dicarboxylate uptake (Dcu) family 2.A.21. Solute-Na H11001 symporter (SSS) family Na H11001 -glucose symporter in epithelial cells 2.A.73. HCO 3 H11002 transporters HCO 3 H11002 -Cl H11002 antiporter 2.B. Nonribosomally synthesized porters 2.B.1. Valinomycin carrier family Valinomycin 3.A. Diphosphate bond hydrolysis–driven transporters (use PP i , not ATP) 3.A.1. ATP-binding cassette (ABC) superfamily CFTR Cl H11002 channel; multidrug transporter MDR1 3.A.2. H H11001 - or Na H11001 -translocating F-type, V-type, A-type ATPase superfamily F O F 1 ATPase proton pump; V O V 1 ATPase; A O A 1 ATPase 3.A.3. P-type ATPase superfamily Na H11001 K H11001 ATPase antiporter; SERCA Ca 2H11001 pump Note: The three broad groups correspond to groups 1, 2, and 3 in Figure 11-29. The individual transporters listed here (screened in yellow) are discussed in this chapter. 8885d_c11_369-420 2/7/04 6:58 AM Page 392 mac76 mac76:385_reb: are superfamilies of various types, defined not only by their primary sequences but by their secondary struc- tures. Some channels are constructed primarily of hel- ical transmembrane segments, others have H9252-barrel structures (Table 11–3). Among the carriers, some sim- ply facilitate diffusion down a concentration gradient; they are the uniporter superfamily. Others (active trans- porters) can drive substrates across the membrane against a concentration gradient, some using energy provided directly by a chemical reaction (primary ac- tive transporters) and some coupling uphill transport of one substrate with the downhill transport of another (secondary active transporters). We now consider some well-studied representatives of the main transporter su- perfamilies. You will encounter some of these trans- porters again in later chapters in the context of the metabolic pathways in which they participate. The Glucose Transporter of Erythrocytes Mediates Passive Transport Energy-yielding metabolism in erythrocytes depends on a constant supply of glucose from the blood plasma, where the glucose concentration is maintained at about 5 mM. Glucose enters the erythrocyte by facilitated dif- fusion via a specific glucose transporter, at a rate about 50,000 times greater than the uncatalyzed diffusion rate. The glucose transporter of erythrocytes (called GLUT1 to distinguish it from related glucose transporters in other tissues) is a type III integral protein (M r ~45,000) with 12 hydrophobic segments, each of which is believed to form a membrane-spanning helix. The detailed struc- ture of GLUT1 is not yet known, but one plausible model suggests that the side-by-side assembly of several he- lices produces a transmembrane channel lined with hy- drophilic residues that can hydrogen-bond with glucose as it moves through the channel (Fig. 11–30). The process of glucose transport can be described by analogy with an enzymatic reaction in which the “sub- strate” is glucose outside the cell (S out ), the “product” is glucose inside (S in ), and the “enzyme” is the trans- porter, T. When the rate of glucose uptake is measured 11.3 Solute Transport across Membranes 393 Transporters Carriers Channels Secondary active transporters Uniporters Primary active transporters 1 2 3 FIGURE 11–29 Classification of transporters. The numbers here cor- respond to the main subdivisions in Table 11–3. Outside Inside + NH 3 COO – Hydrophobic Polar Charged (a) 1 Ser Leu Val Thr Asn Ile Phe 2 3 4 5 6 7 (b) Ser Leu Val Thr Asn Phe Ile (c) Glc FIGURE 11–30 Proposed structure of GLUT1. (a) Transmembrane he- lices are represented as oblique (angled) rows of three or four amino acid residues, each row depicting one turn of the H9251 helix. Nine of the 12 helices contain three or more polar or charged amino acid residues, often separated by several hydrophobic residues. (b) A helical wheel diagram shows the distribution of polar and nonpolar residues on the surface of a helical segment. The helix is diagrammed as though ob- served along its axis from the amino terminus. Adjacent residues in the linear sequence are connected with arrows, and each residue is placed around the wheel in the position it occupies in the helix; re- call that 3.6 residues are required to make one complete turn of the H9251 helix. In this example, the polar residues (blue) are on one side of the helix and the hydrophobic residues (yellow) on the other. This is, by definition, an amphipathic helix. (c) Side-by-side association of five or six amphipathic helices, each with its polar face oriented toward the central cavity, can produce a transmembrane channel lined with polar and charged residues. This channel provides many opportuni- ties for hydrogen bonding with glucose as it moves through the trans- porter. The three-dimensional structure of GLUT1 has not yet been de- termined by x-ray crystallography, but researchers expect that the hydrophilic transmembrane channels of this and many other trans- porters and ion channels will resemble this model. 8885d_c11_369-420 2/7/04 6:58 AM Page 393 mac76 mac76:385_reb: as a function of external glucose concentration (Fig. 11–31), the resulting plot is hyperbolic; at high external glucose concentrations the rate of uptake ap- proaches V max . Formally, such a transport process can be described by the equations in which k 1 , k H110021 , and so forth, are the forward and re- verse rate constants for each step; T 2 is the transporter conformation that faces out, and T 2 the one that faces in. The steps are summarized in Figure 11–32. The rate equations for this process can be derived exactly as for enzyme-catalyzed reactions (Chapter 6), yielding an expression analogous to the Michaelis- Menten equation: V 0 H11005 V max [S] out H5007H5007 K t H11001 [S] out S out S in S out H11001 T 1 H11001 T 2 T 1 k 1 k H110021 ? S in T 2 ? k 3 k H110023 k 4 k H110024 k 2 k H110022 in which V 0 is the initial velocity of accumulation of glu- cose inside the cell when its concentration in the surrounding medium is [S] out , and K t (K transport ) is a constant analogous to the Michaelis constant, a combi- nation of rate constants that is characteristic of each transport system. This equation describes the initial velocity, the rate observed when [S] in H11005 0. As is the case for enzyme-catalyzed reactions, the slope-intercept form of the equation describes a linear plot of 1/V 0 against 1/[S] out , from which we can obtain values of K t and V max (Fig. 11–31b). When [S] H11005 K t , the rate of up- take is 1 ? 2 V max ; the transport process is half-saturated. The concentration of blood glucose, 4.5 to 5 mM, is about Chapter 11 Biological Membranes and Transport394 Extracellular glucose concentration, [S] out (mM) Initial velocity of glucose entry , V 0 ( m M /min) V max V max K t (a) 1 2 1 [S] out 1 mM K t 1 V max 1 ( ) H11002 1 H9262 M /min 1 V 0 ( ) (b) FIGURE 11–31 Kinetics of glucose transport into erythrocytes. (a) The initial rate of glucose entry into an erythrocyte, V 0 , depends upon the initial concentration of glucose on the outside, [S] out . (b) Double- reciprocal plot of the data in (a). The kinetics of facilitated diffusion is analogous to the kinetics of an enzyme-catalyzed reaction. Com- pare these plots with Figure 6–11, and Figure 1 in Box 6–1. Note that K t is analogous to K m , the Michaelis constant. D-Glucose Inside T 1 T 2 T 1 T 2 T 1 Outside 1 2 3 4 FIGURE 11–32 Model of glucose transport into erythrocytes by GLUT1. The transporter exists in two conformations: T 1 , with the glucose-binding site exposed on the outer surface of the plasma mem- brane, and T 2 , with the binding site exposed on the inner surface. Glu- cose transport occurs in four steps. 1 Glucose in blood plasma binds to a stereospecific site on T 1 ; this lowers the activation energy for 2 a conformational change from S out H11554 T 1 to S in H11554 T 2 , effecting the trans- membrane passage of the glucose. 3 Glucose is now released from T 2 into the cytoplasm, and 4 the transporter returns to the T 1 confor- mation, ready to transport another glucose molecule. 8885d_c11_394 2/11/04 12:13 PM Page 394 mac76 mac76:385_reb: three times K t , which ensures that GLUT1 is nearly sat- urated with substrate and operates near V max . Because no chemical bonds are made or broken in the conversion of S out to S in , neither “substrate” nor “product” is intrinsically more stable, and the process of entry is therefore fully reversible. As [S] in approaches [S] out , the rates of entry and exit become equal. Such a system is therefore incapable of accumulating the sub- strate (glucose) within a cell at concentrations above that in the surrounding medium; it simply achieves equi- libration of glucose on the two sides of the membrane much faster than would occur in the absence of a spe- cific transporter. GLUT1 is specific for D-glucose, hav- ing a measured K t of 1.5 mM. For the close analogs D- mannose and D-galactose, which differ only in the position of one hydroxyl group, the values of K t are 20 and 30 mM, respectively; and for L-glucose, K t exceeds 3,000 mM. Thus GLUT1 shows the three hallmarks of passive transport: high rates of diffusion down a concentration gradient, saturability, and specificity. Twelve glucose transporters are encoded in the hu- man genome, each with unique kinetic properties, pat- terns of tissue distribution, and function (Table 11–4). In liver, GLUT2 transports glucose out of hepatocytes when liver glycogen is broken down to replenish blood glucose. GLUT2 has a K t of about 66 mM and can there- fore respond to increased levels of intracellular glucose (produced by glycogen breakdown) by increasing out- ward transport. Skeletal muscle and adipose tissue have yet another glucose transporter, GLUT4 (K t H11005 5 mM), which is distinguished by its stimulation by insulin: its activity increases when release of insulin signals a high blood glucose concentration, thus increasing the rate of glucose uptake into muscle and adipose tissue (Box 11–2 describes some malfunctions of this transporter). The Chloride-Bicarbonate Exchanger Catalyzes Electroneutral Cotransport of Anions across the Plasma Membrane The erythrocyte contains another facilitated diffusion system, an anion exchanger that is essential in CO 2 transport to the lungs from tissues such as skeletal mus- cle and liver. Waste CO 2 released from respiring tissues into the blood plasma enters the erythrocyte, where it is converted to bicarbonate (HCO 3 H11002 ) by the enzyme car- bonic anhydrase. (Recall that HCO 3 H11002 is the primary buffer of blood pH; see Box 2–4). The HCO 3 H11002 reenters the blood plasma for transport to the lungs (Fig. 11–33). Because HCO 3 H11002 is much more soluble in blood plasma than is CO 2 , this roundabout route increases the ca- pacity of the blood to carry carbon dioxide from the tis- sues to the lungs. In the lungs, HCO 3 H11002 reenters the ery- throcyte and is converted to CO 2 , which is eventually released into the lung space and exhaled. To be effec- tive, this shuttle requires very rapid movement of HCO 3 H11002 across the erythrocyte membrane. The chloride-bicarbonate exchanger, also called the anion exchange (AE) protein, increases the permeability of the erythrocyte membrane to HCO 3 H11002 more than a millionfold. Like the glucose transporter, it is an integral protein that probably spans the membrane at least 12 times. This protein mediates the simultane- ous movement of two anions: for each HCO 3 H11002 ion that moves in one direction, one Cl H11002 ion moves in the op- posite direction (Fig. 11–33), with no net transfer of charge; the exchange is electroneutral. The coupling of Cl H11002 and HCO 3 H11002 movements is obligatory; in the ab- sence of chloride, bicarbonate transport stops. In this respect, the anion exchanger is typical of all systems, called cotransport systems, that simultaneously carry 11.3 Solute Transport across Membranes 395 TABLE 11–4 Glucose Transporters in the Human Genome Transporter Tissue(s) where expressed Gene Role * GLUT1 Ubiquitous SLC2A1 Basal glucose uptake GLUT2 Liver, pancreatic islets, intestine SLC2A2 In liver, removal of excess glucose from blood; in pancreas, regulation of insulin release GLUT3 Brain (neuronal) SLC2A3 Basal glucose uptake GLUT4 Muscle, fat, heart SLC2A4 Activity increased by insulin GLUT5 Intestine, testis, kidney, sperm SLC2A5 Primarily fructose transport GLUT6 Spleen, leukocytes, brain SLC2A6 Possibly no transporter function GLUT7 Liver microsomes SLC2A7 — GLUT8 Testis, blastocyst, brain SLC2A8 — GLUT9 Liver, kidney SLC2A9 — GLUT10 Liver, pancreas SLC2A10 — GLUT11 Heart, skeletal muscle SLC2A11 — GLUT12 Skeletal muscle, adipose, small intestine SLC2A12 — * Dash indicates role uncertain. 8885d_c11_369-420 2/7/04 6:58 AM Page 395 mac76 mac76:385_reb: BOX 11–2 BIOCHEMISTRY IN MEDICINE Defective Glucose and Water Transport in Two Forms of Diabetes When ingestion of a carbohydrate-rich meal causes blood glucose to exceed the usual concentration be- tween meals (about 5 mM), excess glucose is taken up by the myocytes of cardiac and skeletal muscle (which store it as glycogen) and by adipocytes (which convert it to triacylglycerols). Glucose uptake into myocytes and adipocytes is mediated by the glucose transporter GLUT4. Between meals, some GLUT4 is present in the plasma membrane, but most is sequestered in the membranes of small intracellular vesicles (Fig. 1). In- sulin released from the pancreas in response to high blood glucose triggers the movement of these intra- cellular vesicles to the plasma membrane, where they fuse, thus exposing GLUT4 molecules on the outer sur- face of the cell (see Fig. 12–8). With more GLUT4 mol- ecules in action, the rate of glucose uptake increases 15-fold or more. When blood glucose levels return to normal, insulin release slows and most GLUT4 mole- cules are removed from the plasma membrane and stored in vesicles. In type I (juvenile onset) diabetes mellitus, the in- ability to release insulin (and thus to mobilize glucose transporters) results in low rates of glucose uptake into muscle and adipose tissue. One consequence is a prolonged period of high blood glucose after a carbohydrate-rich meal. This condition is the basis for the glucose tolerance test used to diagnose diabetes (Chapter 23). The water permeability of epithelial cells lining the renal collecting duct in the kidney is due to the presence of an aquaporin (AQP-2) in their apical plasma membranes (facing the lumen of the duct). Antidiuretic hormone (ADH) regulates the retention of water by mobilizing AQP-2 molecules stored in vesicle membranes within the epithelial cells, much as insulin mobilizes GLUT4 in muscle and adipose tissue. When the vesicles fuse with the epithelial cell plasma membrane, water permeability greatly increases and more water is reabsorbed from the collecting duct and returned to the blood. When the ADH level drops, AQP-2 is resequestered within vesicles, reducing water retention. In the relatively rare human disease diabetes insipidus, a genetic defect in AQP-2 leads to impaired water reabsorption by the kidney. The result is excretion of copious volumes of very dilute urine. FIGURE 1 Regulation by insulin of glucose transport by GLUT4 into a myocyte. When insulin level drops, glucose transporters are removed from the plasma membrane by endocytosis, forming small vesicles. 2 3 1 5 4 Glucose transporters “stored” within cell in membrane vesicles. Patches of the endosome enriched with glucose transporters bud off to become small vesicles, ready to return to the surface when insulin levels rise again. The smaller vesicles fuse with larger endosome. Glucose transporter Plasma membrane Insulin receptor Insulin When insulin interacts with its receptor, vesicles move to surface and fuse with the plasma membrane, increasing the number of glucose transporters in the plasma membrane. 8885d_c11_396 2/11/04 12:14 PM Page 396 mac76 mac76:385_reb: two solutes across a membrane. When, as in this case, the two substrates move in opposite directions, the process is antiport. In symport, two substrates are moved simultaneously in the same direction. As we noted earlier, transporters that carry only one substrate, such as the erythrocyte glucose transporter, are uni- port systems (Fig. 11–34). The human genome has genes for three closely related chloride-bicarbonate exchangers, all with the same predicted transmembrane topology. Erythrocytes contain the AE1 transporter, AE2 is prominent in liver, and AE3 is present in plasma membranes of the brain, heart, and retina. Similar anion exchangers are also found in plants and microorganisms. Active Transport Results in Solute Movement against a Concentration or Electrochemical Gradient In passive transport, the transported species always moves down its electrochemical gradient and is not ac- cumulated above the equilibrium concentration. Active transport, by contrast, results in the accumulation of a solute above the equilibrium point. Active transport is thermodynamically unfavorable (endergonic) and takes place only when coupled (directly or indirectly) to an exergonic process such as the absorption of sunlight, an oxidation reaction, the breakdown of ATP, or the con- comitant flow of some other chemical species down its electrochemical gradient. In primary active trans- port, solute accumulation is coupled directly to an ex- ergonic chemical reaction, such as conversion of ATP to ADP H11001 P i (Fig. 11–35). Secondary active transport occurs when endergonic (uphill) transport of one solute is coupled to the exergonic (downhill) flow of a differ- ent solute that was originally pumped uphill by primary active transport. The amount of energy needed for the transport of a solute against a gradient can be calculated from the initial concentration gradient. The general equation for the free-energy change in the chemical process that con- verts S to P is H9004G H11005 H9004GH11032H11034 H11001 RT ln [P]/[S] (11–1) where R is the gas constant, 8.315 J/mol H11554 K, and T is the absolute temperature. When the “reaction” is simply 11.3 Solute Transport across Membranes 397 Carbon dioxide produced by catabolism enters erythrocyte. Bicarbonate dissolves in blood plasma. Carbon dioxide leaves erythrocyte and is exhaled. CO 2 H11001 H 2 O CO 2 H11001 H 2 O HH11001 Cl H11002 H11002H11001 HCO 3 HH11001 ClHCO 3 Bicarbonate enters erythrocyte from blood plasma. CO 2 H11002H11002 H11002H11002H11001 H11002H11002 CO 2 Chloride-bicarbonate exchange protein HCO 3 Cl HCO 3 Cl In respiring tissues In lungs carbonic anhydrase carbonic anhydrase S Uniport Symport Antiport Cotransport S 1 S 2 S 2 S 1 FIGURE 11–33 Chloride-bicarbonate exchanger of the erythrocyte membrane. This cotransport system allows the entry and exit of HCO 3 H11002 without changes in the transmembrane electrical potential. Its role is to increase the CO 2 -carrying capacity of the blood. FIGURE 11–34 Three general classes of transport systems. Trans- porters differ in the number of solutes (substrates) transported and the direction in which each is transported. Examples of all three types of transporters are discussed in the text. Note that this classification tells us nothing about whether these are energy-requiring (active transport) or energy-independent (passive transport) processes. FIGURE 11–35 Two types of active transport. (a) In primary active transport, the energy released by ATP hydrolysis drives solute move- ment against an electrochemical gradient. (b) In secondary active transport, a gradient of ion X (often Na H11001 ) has been established by primary active transport. Movement of X down its electrochemical gra- dient now provides the energy to drive cotransport of a second solute (S) against its electrochemical gradient. (a) Primary active transport (b) Secondary active transport ATP ADP H11001P i ATP ADP H11001P i X X X X X X X X X X X X X X XX X X X X X X X S S S S S S S X S X X 8885d_c11_369-420 2/7/04 6:58 AM Page 397 mac76 mac76:385_reb: transport of a solute from a region where its concen- tration is C 1 to a region where its concentration is C 2 , no bonds are made or broken and the standard free-en- ergy change, H9004GH11032H11034, is zero. The free-energy change for transport, H9004G t , is then H9004G t H11005 RT ln H5007 C C 2 1 H5007 (11–2) If there is a tenfold difference in concentration between two compartments, the cost of moving 1 mol of an un- charged solute at 25 H11034C across a membrane separating the compartments is therefore H9004G t H11005 (8.315 J/mol H11554 K)(298 K)(ln 10/1) H11005 5,700 J/mol H11005 5.7 kJ/mol Equation 11–2 holds for all uncharged solutes. When the solute is an ion, its movement without an accompanying counterion results in the endergonic sep- aration of positive and negative charges, producing an electrical potential; such a transport process is said to be electrogenic. The energetic cost of moving an ion depends on the electrochemical potential (p. 391), the sum of the chemical and electrical gradients: H9004G t H11005 RT ln H20898 H5007 C C 2 1 H5007 H20899 H11001 Z H9004H9274 (11–3) where Z is the charge on the ion, is the Faraday con- stant (96,480 J/V H11554 mol), and H9004H9274 is the transmembrane electrical potential (in volts). Eukaryotic cells typically have electrical potentials across their plasma mem- branes of about 0.05 to 0.1 V (with the inside negative relative to the outside), so the second term of Equation 11–3 can make a significant contribution to the total free-energy change for transporting an ion. Most cells maintain more than tenfold differences in ion concen- trations across their plasma or intracellular membranes, and for many cells and tissues active transport is there- fore a major energy-consuming process. The mechanism of active transport is of fundamen- tal importance in biology. As we shall see in Chapter 19, the formation of ATP in mitochondria and chloroplasts occurs by a mechanism that is essentially ATP-driven ion transport operating in reverse. The energy made available by the spontaneous flow of protons across a membrane is calculable from Equation 11–3; remember that H9004G for flow down an electrochemical gradient has a negative value, and H9004G for transport of ions against an electrochemical gradient has a positive value. P-Type ATPases Undergo Phosphorylation during Their Catalytic Cycles The family of active transporters called P-type ATPases are ATP-driven cation transporters that are reversibly phosphorylated by ATP as part of the transport cycle; phosphorylation forces a conformational change that is central to moving the cation across the membrane. All P-type transport ATPases have similarities in amino acid sequence, especially near the Asp residue that under- goes phosphorylation, and all are sensitive to inhibition by the phosphate analog vanadate. Each P-type ATPase transporter is an integral protein with ten predicted membrane-spanning regions in a sin- gle polypeptide; some also have a second subunit. The P-type transporters are very widely distributed. In ani- mal tissues, the Na H11001 K H11001 ATPase (an antiporter for Na H11001 and K H11001 ) and the Ca 2H11001 ATPase (a uniporter for Ca 2H11001 ) are ubiquitous P-type ATPases that maintain differences in the ionic composition of the cytosol and the extra- cellular medium. Parietal cells in the lining of the mam- malian stomach have a P-type ATPase that pumps H H11001 and K H11001 across the plasma membrane, thereby acidify- ing the stomach contents. In vascular plants, a P-type ATPase pumps protons out of the cell, establishing an electrochemical difference of as much as 2 pH units and 250 mV across the plasma membrane. A similar P-type ATPase in the bread mold Neurospora pumps protons out of cells to establish an inside-negative membrane potential, which is used to drive the uptake of substrates and ions from the surrounding medium by secondary active transport. Bacteria use P-type ATPases to pump out toxic heavy metal ions such as Cd 2H11001 and Cu 2H11001 . In virtually every animal cell type, the concentra- tion of Na H11001 is lower in the cell than in the surrounding medium, and the concentration of K H11001 is higher (Fig. 11–36). This imbalance is maintained by a primary ac- tive transport system in the plasma membrane. The en- zyme Na H11545 K H11545 ATPase, dis- covered by Jens Skou in 1957, couples breakdown of ATP to the simultaneous movement of both Na H11001 and K H11001 against their electrochemical gradi- ents. For each molecule of ATP converted to ADP and P i , the transporter moves two K H11001 ions inward and three Na H11001 ions outward across the plasma membrane. The Na H11001 K H11001 ATPase is an integral protein with two subunits (M r ~50,000 and ~110,000), both of which span the membrane. The detailed mechanism by which ATP hydrolysis is coupled to transport awaits determination of the pro- tein’s three-dimensional structure, but a current model (Fig. 11–37) proposes that the ATPase cycles between two forms, a phosphorylated form (designated P-Enz II ) with high affinity for K H11001 and low affinity for Na H11001 , and a dephosphorylated form (Enz I ) with high affinity for Na H11001 Vanadate V O H11002 OH O Phosphate P O H11002 OH OO H11002 O H11002 Chapter 11 Biological Membranes and Transport398 Jens Skou 8885d_c11_369-420 2/7/04 6:58 AM Page 398 mac76 mac76:385_reb: and low affinity for K H11001 . The conversion of ATP to ADP and P i takes place in two steps catalyzed by the enzyme, involving formation then hydrolysis of the phospho- enzyme: (1) ATP H11001 Enz I 88n ADP H11001 P-Enz II (2) P-Enz II H11001 H 2 O 88n Enz I H11001 P i Sum: ATP H11001 H 2 O 88n ADP H11001 P i Because three Na H11001 ions move outward for every two K H11001 ions that move inward, the process is electrogenic—it creates a net separation of charge across the membrane. The result is a transmembrane potential of H1100250 to H1100270 mV (inside negative relative to outside), which is char- acteristic of most animal cells and essential to the con- duction of action potentials in neurons. The central role of the Na H11001 K H11001 ATPase is reflected in the energy invested in this single reaction: about 25% of the total energy consumption of a human at rest! The steroid derivative ouabain (pronounced wah’-bane; from waa bayyo, Somali for “arrow poison”) is a potent and specific inhibitor of the Na H11001 K H11001 ATPase. Oubain binds preferentially to the form of the enzyme that is open to the extracellular side, locking in two Na H11001 ions and preventing the changes of conforma- tion necessary to ion transport. Another very potent toxin, palytoxin (produced by a coral on the Hawaiian shoreline), also targets the Na H11001 K H11001 ATPase, but it binds to the protein so as to lock it into a position in which the ion-binding sites are permanently accessible from both sides, converting the transporter into a nonspecific ion channel. This allows exit of K H11001 from cells and de- flates the (essential) ion gradient across the plasma membrane, which accounts for the high toxicity of this compound. OH CH 3 O H H H HH OH OH O Ouabain OH OH HO HO HC CH 2 OC O CH 2 H 3 C C OH 11.3 Solute Transport across Membranes 399 Membrane potential H11005 H1100250 to H1100270 mV H11001 H11001H11001H11001 H11001H11001H11001 H11002H11002H11002 H11002H11002H11002 H11001H11001H11001 H11001H11001H11001H11001 H11002H11002H11002 H11002H11002H11002H11002 H11001 H11001 H11001 H11001 H11002 H11002 H11002 H11002 H11002 H11002 H11002 H11002 H11002 H11002 H11001 H11001 H11001 H11001 H11001 Extracellular fluid or blood plasma 3 Na H11001 Na H11001 K H11001 ATPase 2 K H11001 Cytosol [K H11001 ] H11005 140 mM [Na H11001 ] H11005 12 mM [K H11001 ] H11005 4 mM [Na H11001 ] H11005 145 mM ATP ADP H11001 P i FIGURE 11–36 Na H11545 K H11545 ATPase. In animal cells, this active transport system is primarily responsible for setting and maintaining the intra- cellular concentrations of Na H11001 and K H11001 and for generating the trans- membrane electrical potential. It does this by moving three Na H11001 out of the cell for every two K H11001 it moves in. The electrical potential is cen- tral to electrical signaling in neurons, and the gradient of Na H11001 is used to drive the uphill cotransport of solutes in many cell types. FIGURE 11–37 Postulated mechanism of Na H11545 and K H11545 transport by the Na H11545 K H11545 ATPase. Transporter binds 3 Na H11001 from the inside of the cell. Enz I P–Enz II P–Enz II Enz I Phosphorylation favors P–Enz II . Transporter releases 3 Na H11001 to the outside and binds 2 K H11001 from the outside of the cell. Dephosphorylation favors Enz I . Transporter releases 2 K H11001 to the inside. OutsideInside P i 2 K H11001 2 K H11001 3 Na H11001 P P ATP ADP 3 Na H11001 8885d_c11_399 2/11/04 12:14 PM Page 399 mac76 mac76:385_reb: Ouabain and another steroid derivative, digitoxi- genin, are the active ingredients of digitalis, an extract of the leaves of the foxglove plant. (Ouabain is found in lower concentrations in a number of other plants, pre- sumably serving to discourage herbivores.) Digitalis has been used to treat congestive heart failure since its in- troduction for that purpose (treatment of “dropsy”) by the British physician William Withering in 1785. It strengthens heart muscle contractions without increas- ing the heart rate and thus increases the efficiency of the heart. Digitalis inhibits the efflux of Na H11001 , raising the intracellular [Na H11001 ] enough to activate a Na H11001 -Ca 2H11001 antiporter in cardiac muscle. The increased influx of Ca 2H11001 through this antiporter produces elevated cytoso- lic [Ca 2H11001 ], which strengthens the contractions of the heart. The potency of ouabain in animals led to the sug- gestion (50 years ago) that this plant product might act by mimicking a normal regulator of the Na H11001 K H11001 ATPase produced in animals, and it now appears that this may be so. Ouabain itself has been isolated from bovine ad- renal glands and has been detected in the blood plasma and hypothalamus of mammals. ■ P-Type Ca 2H11545 Pumps Maintain a Low Concentration of Calcium in the Cytosol The cytosolic concentration of free Ca 2H11001 is generally at or below 100 nM, far lower than that in the surrounding medium, whether pond water or blood plasma. The ubiq- uitous occurrence of inorganic phosphates (P i and PP i ) at millimolar concentrations in the cytosol necessitates a low cytosolic Ca 2H11001 concentration, because inorganic phosphate combines with calcium to form relatively in- soluble calcium phosphates. Calcium ions are pumped out of the cytosol by a P-type ATPase, the plasma mem- brane Ca 2H11545 pump. Another P-type Ca 2H11001 pump in the endoplasmic reticulum moves Ca 2H11001 into the ER lumen, a compartment separate from the cytosol. In myocytes, Ca 2H11001 is normally sequestered in a specialized form of endoplasmic reticulum called the sarcoplasmic reticu- lum. The sarcoplasmic and endoplasmic reticulum calcium (SERCA) pumps are closely related in struc- ture and mechanism, and both are inhibited by the tumor-promoting agent thapsigargin, which does not af- fect the plasma membrane Ca 2H11001 pump. The plasma membrane Ca 2H11001 pump and SERCA pumps are integral proteins that cycle between phos- phorylated and dephosphorylated conformations in a mechanism similar to that for Na H11001 K H11001 ATPase (Fig. 11–37). Phosphorylation favors a conformation with a high-affinity Ca 2H11001 -binding site exposed on the cyto- plasmic side, and dephosphorylation favors one with a low-affinity Ca 2H11001 -binding site on the lumenal side. By this mechanism, the energy released by hydrolysis of ATP during one phosphorylation-dephosphorylation cy- cle drives Ca 2H11001 across the membrane against a large electrochemical gradient. The Ca 2H11001 pump of the sarcoplasmic reticulum, which comprises 80% of the protein in that membrane, consists of a single polypeptide (M r ~100,000) that spans the membrane ten times and has three cytoplas- mic domains formed by loops that connect the trans- membrane helices (Fig. 11–38). The two Ca 2H11001 -binding sites are located near the middle of the membrane bi- Chapter 11 Biological Membranes and Transport400 ATP binding site N domain A domain Phosphorylation site (Asp 351 ) P domain Ca 2+ binding sites Ca 2+ Cytoplasm ER lumen 90° 20° FIGURE 11–38 Structure of the Ca 2H11545 pump of sarcoplasmic reticu- lum. (PDB ID 1EUL) Ten transmembrane helices surround the path for Ca 2H11001 movement through the membrane. Two of the helices are inter- rupted near the middle of the bilayer, and their nonhelical regions form the binding sites for two Ca 2H11001 ions (green). The carboxylate groups of an Asp residue in one helix and a Glu residue in another are central to the Ca 2H11001 -binding sites. Three globular domains extend from the cytoplasmic side: the N (nucleotide-binding) domain has the binding site for ATP; the P (phosphorylation) domain contains the Asp 351 residue (blue) that undergoes reversible phosphorylation, and the A (actuator) domain somehow mediates the structural changes that alter the Ca 2H11001 affinity of the Ca 2H11001 -binding site and its exposure to cytoplasm or lumen. Note the long distance between the phosphoryla- tion site and the Ca 2H11001 -binding site. There is strong evidence that dur- ing one transport cycle, the N domain tips about 20H11034 to the right, bring- ing the ATP site close to Asp 351 , and that during each catalytic cycle the A domain twists by about 90H11034 around the normal (perpendicular) to the membrane. These conformational changes must expose the Ca 2H11001 -binding site first on one side of the membrane, then on the other, changing the Ca 2H11001 affinity of the site from high on the cytoplasmic side to lower on the lumenal side. A complete understanding of the coupling between phosphorylation and Ca 2H11001 transport awaits deter- mination of all the conformations involved in the cycle. 8885d_c11_369-420 2/7/04 6:58 AM Page 400 mac76 mac76:385_reb: layer, 40 to 50 ? from the phosphorylated Asp residue characteristic of all P-type ATPases, so the effects of Asp phosphorylation are not direct. They must be me- diated by conformational changes that alter the affinity for Ca 2H11001 and open a path for Ca 2H11001 release on the lu- menal side of the membrane. The amino acid sequences of the SERCA pumps and the Na H11001 K H11001 ATPase share 30% identity and 65% se- quence similarity, and their topology relative to the membrane is also the same. Thus it seems likely that the Na H11001 K H11001 ATPase structure is similar to that of the SERCA pumps and that all P-type ATPase transporters share the same basic structure. F-Type ATPases Are Reversible, ATP-Driven Proton Pumps The F-type ATPase active transporters play a central role in energy-conserving reactions in mitochondria, bacteria, and chloroplasts; we discuss that role in detail in our description of oxidative phosphorylation and pho- tophosphorylation in Chapter 19. The F-type ATPases catalyze the uphill transmembrane passage of protons driven by ATP hydrolysis (“F-type” originated in the identification of these ATPases as energy-coupling fac- tors). The F o integral membrane protein complex (Fig. 11–39; subscript o denoting its inhibition by the drug oligomycin) provides a transmembrane pore for protons, and the peripheral protein F 1 (subscript 1 indicating that it was the first of several factors isolated from mi- tochondria) is a molecular machine that uses the energy of ATP to drive protons uphill (into a region of higher H H11001 concentration). The F o F 1 organization of proton- pumping transporters must have developed very early in evolution. Eubacteria such as E. coli use an F o F 1 ATPase complex in their plasma membrane to pump protons outward, and archaebacteria have a closely homologous proton pump, the A o A 1 ATPase. The reaction catalyzed by F-type ATPases is re- versible, so a proton gradient can supply the energy to drive the reverse reaction, ATP synthesis (Fig. 11–40). When functioning in this direction, the F-type ATPases are more appropriately named ATP synthases. ATP synthases are central to ATP production in mitochon- dria during oxidative phosphorylation and in chloro- plasts during photophosphorylation, as well as in eu- bacteria and archaebacteria. The proton gradient needed to drive ATP synthesis is produced by other types of proton pumps powered by substrate oxidation or sunlight. As noted above, we return to a detailed de- scription of these processes in Chapter 19. V-type ATPases, a class of proton-transporting ATPases structurally (and possibly mechanistically) re- lated to the F-type ATPases, are responsible for acidi- fying intracellular compartments in many organisms (thus V for vacuolar). Proton pumps of this type main- tain the vacuoles of fungi and higher plants at a pH be- tween 3 and 6, well below that of the surrounding cy- tosol (pH 7.5). V-type ATPases are also responsible for the acidification of lysosomes, endosomes, the Golgi complex, and secretory vesicles in animal cells. All V- type ATPases have a similar complex structure, with an integral (transmembrane) domain (V o ) that serves as a proton channel and a peripheral domain (V 1 ) that con- tains the ATP-binding site and the ATPase activity. The mechanism by which V-type ATPases couple ATP hy- drolysis to the uphill transport of protons is not under- stood in detail. 11.3 Solute Transport across Membranes 401 H9252 H9251 H9251 H9280 H9251 H9252 H9252 H9253 H9254 b 2 c 12 H + H + a ADP + P i ATP F 1 F o (f) FIGURE 11–39 Structure of the F o F 1 ATPase/ATP synthase. F-type ATPases have a peripheral domain, F 1 , consisting of three H9251 subunits, three H9252 subunits, one H9254 subunit (purple), and a central shaft (the H9253 sub- unit, green). The integral portion of F-type ATPases, F o (yellow), has multiple copies of c, one a, and two b subunits. F o provides a trans- membrane channel through which about four protons are pumped (red arrows) for each ATP hydrolyzed on the H9252 subunits of F 1 . The remarkable mechanism by which these two events are coupled is de- scribed in detail in Chapter 19. It involves rotation of F o relative to F 1 (black arrow). The structures of V o V 1 and A o A 1 are essentially similar to that of F o F 1 , and the mechanisms are probably similar, too. ATP ADP H11001P i H H11001 H H11001 ATP synthase Proton pump H H11001 FIGURE 11–40 Reversibility of F-type ATPases. An ATP-driven proton transporter also can catalyze ATP synthesis (red arrows) as protons flow down their electrochemical gradient. This is the central reaction in the processes of oxidative phosphorylation and photophosphorylation, both described in detail in Chapter 19. 8885d_c11_369-420 2/7/04 6:58 AM Page 401 mac76 mac76:385_reb: ABC Transporters Use ATP to Drive the Active Transport of a Wide Variety of Substrates ABC transporters (Fig. 11–41) constitute a large family of ATP-dependent transporters that pump amino acids, peptides, proteins, metal ions, various lipids, bile salts, and many hydrophobic compounds, including drugs, out of cells against a concentration gradient. One ABC transporter in humans, the multi- drug transporter (MDR1), is responsible for the striking resistance of certain tumors to some generally effective antitumor drugs. MDR1 has a broad substrate specificity for hydrophobic compounds, including, for example, the chemotherapeutic drugs adriamycin, doxorubicin, and vinblastine. By pumping these drugs out of the cell, the transporter prevents their accu- mulation within a tumor and thus blocks their thera- peutic effects. MDR1 is an integral membrane protein (M r 170,000) with 12 transmembrane segments and two ATP-binding domains (“cassettes”), which give the family its name: ATP-binding cassette transporters. All ABC transporters have two nucleotide-binding domains (NBDs) and two transmembrane domains (Fig. 11–41). In some cases, all these domains are in a single long polypeptide; other ABC transporters have two sub- units, each contributing an NBD and a domain with six (or in some cases ten) transmembrane helices. Although many of the ABC transporters are in the plasma mem- brane, some types are also found in the endoplasmic reticulum and in the membranes of mitochondria and lysosomes. Most ABC transporters act as pumps, but at least some members of the superfamily act as ion chan- nels that are opened and closed by ATP hydrolysis. The CFTR transporter (Box 11–3) is a Cl H11002 channel operated by ATP hydrolysis. The NBDs of all ABC proteins are similar in sequence and presumably in three-dimensional structure; they are the conserved molecular motor that can be coupled to a wide variety of pumps and channels. When coupled with a pump, the ATP-driven motor moves solutes against a concentration gradient; when coupled with an ion chan- nel, the motor opens and closes the channel using ATP as energy source. The stoichiometry of ABC pumps is about one ATP hydrolyzed per molecule of substrate transported, but neither the mechanism of coupling nor the site of substrate binding are known. Some ABC transporters have very high speci- ficity for a single substrate; others are more promiscuous. The human genome contains at least 48 genes that encode ABC transporters, many of which are involved in maintaining the lipid bilayer and in trans- porting sterols, sterol derivatives, and fatty acids throughout the body. The flippases that move mem- brane lipids from one leaflet of the bilayer to the other are ABC transporters, and the cellular machinery for ex- porting excess cholesterol includes an ABC transporter. Mutations in the genes that encode some of these pro- teins contribute to several genetic diseases, including cystic fibrosis (Box 11–3), Tangier disease (p. 827), reti- nal degeneration, anemia, and liver failure. ABC transporters are also present in simpler ani- mals and in plants and microorganisms. Yeast has 31 genes that encode ABC transporters, Drosophila has 56, and E. coli has 80, representing 2% of its entire genome. The presence of ABC transporters that confer antibiotic resistance in pathogenic microbes (Pseudomonas aeruginosa, Staphylococcus aureus, Candida albicans, Neisseria gonorrhoeae, and Plas- modium falciparum) is a serious public health con- cern and makes these transporters attractive targets for drug design. ■ Ion Gradients Provide the Energy for Secondary Active Transport The ion gradients formed by primary transport of Na H11001 or H H11001 can in turn provide the driving force for cotrans- port of other solutes. Many cell types contain transport Chapter 11 Biological Membranes and Transport402 FIGURE 11–41 Structures of two ABC trans- porters of E. coli. (a) The lipid A flippase MsbA (PDB ID 1JSQ) and (b) the vitamin B 12 importer BtuCD (PDB ID 1L7V). Both struc- tures are homodimers. The two nucleotide- binding domains (NBDs, in red) extend into the cytoplasm. In (b), residues involved in ATP binding and hydrolysis are shown as ball- and-stick structures. Each monomer of MsbA has six transmembrane helical segments (blue), and each monomer of BtuCD has ten. NBDs Cytoplasm Extracellular space (a) MsbA (b) BtuCD NBDs 8885d_c11_402 2/11/04 12:47 PM Page 402 mac76 mac76:385_reb: systems that couple the spontaneous, downhill flow of these ions to the simultaneous uphill pumping of an- other ion, sugar, or amino acid (Table 11–5). The lac- tose transporter (lactose permease) of E. coli is the well-studied prototype for proton-driven cotrans- porters. This protein consists of a single polypeptide chain (417 residues) that functions as a monomer to transport one proton and one lactose molecule into the 11.3 Solute Transport across Membranes 403 BOX 11–3 BIOCHEMISTRY IN MEDICINE A Defective Ion Channel in Cystic Fibrosis Cystic fibrosis (CF) is a serious and relatively com- mon hereditary disease of humans. About 5% of white Americans are carriers, having one defective and one normal copy of the gene. Only individuals with two de- fective copies show the severe symptoms of the dis- ease: obstruction of the gastrointestinal and respira- tory tracts, commonly leading to bacterial infection of the airways and death due to respiratory insufficiency before the age of 30. In CF, the thin layer of mucus that normally coats the internal surfaces of the lungs is abnormally thick, obstructing air flow and providing a haven for pathogenic bacteria, particularly Staphy- lococcus aureus and Pseudomonas aeruginosa. The defective gene in CF patients was discov- ered in 1989. It encodes a membrane protein called cystic fibrosis transmembrane conductance regula- tor, or CFTR. Hydropathy analysis predicted that CFTR has 12 transmembrane helices and is struc- turally related to the multidrug (MDR1) transporters of drug-resistant tumors (Fig. 1). The normal CFTR protein proved to be an ion channel specific for Cl H11002 ions. The Cl H11002 channel activity increases greatly when phosphoryl groups are transferred from ATP to several side chains of the protein, catalyzed by cAMP-dependent protein kinase (Chapter 12). The mutation responsible for CF in 70% of cases results in deletion of a Phe residue at position 508, with the effect that the mutant protein is not correctly folded and inserted in the plasma membrane. Other muta- tions yield a protein that is inserted properly but can- not be activated by phosphorylation. In each case, the fundamental problem is a nonfunctional Cl H11002 channel in the epithelial cells that line the airways (Fig. 2), the digestive tract, and exocrine glands (pancreas, sweat glands, bile ducts, and vas deferens). Normally, epithelial cells that line the inner sur- face of the lungs secrete a substance that traps and kills bacteria, and the cilia on the epithelial cells con- stantly sweep away the resulting debris. When CFTR is defective or missing, this process is less efficient, and frequent infections by bacteria such as S. aureus and P. aeruginosa progressively damage the lungs and reduce respiratory efficiency. FIGURE 1 Topology of the cystic fibrosis transmembrane conduc- tance regulator, CFTR. It has 12 transmembrane helices, and three functionally significant domains extend from the cytoplasmic sur- face: NBD 1 and NBD 2 are nucleotide-binding domains to which ATP binds, and a regulatory domain (R domain) is the site of phos- phorylation by cAMP-dependent protein kinase. Oligosaccharide chains are attached to several residues on the outer surface of the segment between helices 7 and 8. The most commonly occurring mutation leading to CF is the deletion of Phe 508 , in the NBD 1 do- main. The structure of CFTR is very similar to that of the multidrug transporter of tumors, described in the text. FIGURE 2 Mucus lining the surface of the lungs traps bacteria. In healthy lungs, these bacteria are killed and swept away by the ac- tion of cilia. In CF, the bactericidal activity is impaired, resulting in recurring infections and progressive damage to the lungs. Oligosaccharide chains of glycoprotein Outside Inside R domain Phe 508 COO – NH 3 NBD 1 NBD 2 + 8885d_c11_369-420 2/7/04 6:58 AM Page 403 mac76 mac76:385_reb: cell, with the net accumulation of lactose (Fig. 11–42). E. coli normally produces a gradient of protons and charge across its plasma membrane by oxidizing fuels and using the energy of oxidation to pump protons outward. (This mechanism is discussed in detail in Chapter 19.) The lipid bilayer is impermeable to pro- tons, but the lactose transporter provides a route for proton reentry, and lactose is simultaneously carried into the cell by symport. The endergonic accumulation of lactose is thereby coupled to the exergonic flow of protons into the cell, with a negative overall free-energy change. The lactose transporter is one member of the major facilitator superfamily (MFS) of trans- porters, which comprises 28 families. Almost all proteins in this superfamily have 12 transmembrane domains (the few exceptions have 14). The proteins share rela- tively little sequence homology, but the similarity of their secondary structures and topology suggests a common tertiary structure. The crystallographic solu- tion of the E. coli lactose transporter by Ron Kaback and So Iwata in 2003 may provide a glimpse of this gen- eral structure (Fig. 11–43a). The protein has 12 trans- membrane helices, and connecting loops that protrude into the cytoplasm or the periplasmic space. All six amino-terminal and six carboxyl-terminal helices form very similar domains, to produce a structure with a rough twofold symmetry. In the crystallized form of the protein, a large aqueous cavity is exposed on the cyto- plasmic side of the membrane. The substrate-binding site is in this cavity, more or less in the middle of the membrane. The side of the transporter facing outward (the periplasmic face) is closed tightly, with no channel big enough for lactose to enter. The proposed mecha- Chapter 11 Biological Membranes and Transport404 TABLE 11–5 Cotransport Systems Driven by Gradients of Na H11545 or H H11545 Transported solute Cotransported solute Organism/tissue/cell type (moving against its gradient) (moving down its gradient) Type of transport E. coli Lactose H H11001 Symport Proline H H11001 Symport Dicarboxylic acids H H11001 Symport Intestine, kidney (vertebrates) Glucose Na H11001 Symport Amino acids Na H11001 Symport Vertebrate cells (many types) Ca 2H11001 Na H11001 Antiport Higher plants K H11001 H H11001 Antiport Fungi (Neurospora)K H11001 H H11001 Antiport Lactose (outside) Lactose transporter Proton pump (inhibited by CN H11002 ) Lactose (inside) H H11001 H H11001 H H11001 H H11001 H H11001 H H11001 H H11001 H H11001 H H11001 H H11001 H H11001 (a) H11001H11001 H11001 H11001 H11001 H11001 H11002 H11002 H11002 H11002 H11002 H11002 Fuel CO 2 [Lactose] inside Time (b) [Lactose] medium Active transport +CN – , or mutation at Glu 325 or Arg 302 Efflux CN H11002 inhibition of fuel oxidation FIGURE 11–42 Lactose uptake in E. coli. (a) The primary transport of H H11001 out of the cell, driven by the oxidation of a variety of fuels, es- tablishes both a proton gradient and an electrical potential (inside neg- ative) across the membrane. Secondary active transport of lactose into the cell involves symport of H H11001 and lactose by the lactose transporter. The uptake of lactose against its concentration gradient is entirely de- pendent on this inflow of H H11001 , driven by the electrochemical gradient. (b) When the energy-yielding oxidation reactions of metabolism are blocked by cyanide (CN H11002 ), the lactose transporter allows equilibra- tion of lactose inside and outside the cell via passive transport. Mu- tations that affect Glu 325 or Arg 302 have the same effect as cyanide. The dashed line represents the concentration of lactose in the sur- rounding medium. 8885d_c11_369-420 2/7/04 6:58 AM Page 404 mac76 mac76:385_reb: nism for transmembrane passage of the substrate (Fig. 11–43b) involves a rocking motion between the two domains, driven by substrate binding and proton movement, alternately exposing the substrate-binding domain to the cytoplasm and to the periplasm. This so-called rocking banana model is similar to that shown in Figure 11–32 for GLUT1. How is proton movement into the cell coupled with lactose uptake? Extensive genetic studies of the lactose transporter have established that of the 417 residues in the protein, only 6 are absolutely essential for cotrans- port of H H11001 and lactose—some for lactose binding, oth- ers for proton transport. Mutation in either of two residues (Glu 325 and Arg 302 ; Fig. 11–43) results in a pro- tein still able to catalyze facilitated diffusion of lactose but incapable of coupling H H11001 flow to uphill lactose trans- port. A similar effect is seen in wild-type (unmutated) cells when their ability to generate a proton gradient is blocked with CN H11002 : the transporter carries out facilitated diffusion normally, but it cannot pump lactose against a concentration gradient (Fig. 11–42b). The balance be- tween the two conformations of the lactose transporter is affected by changes in charge pairing between side chains. In intestinal epithelial cells, glucose and certain amino acids are accumulated by symport with Na H11001 , down the Na H11001 gradient established by the Na H11001 K H11001 ATPase of the plasma membrane (Fig. 11–44). The api- cal surface of the intestinal epithelial cell is covered with microvilli, long thin projections of the plasma membrane 11.3 Solute Transport across Membranes 405 Cytoplasm Periplasmic space (a) (b) FIGURE 11–43 Structure of the lactose transporter (lactose perme- ase) of E. coli. (a) Ribbon representation viewed parallel to the plane of the membrane shows the 12 transmembrane helices arranged in two nearly symmetrical domains shown in different shades of blue. In the form of the protein for which the crystal structure was determined, the substrate sugar (red) is bound near the middle of the membrane where it is exposed to the cytoplasm (derived from PDB ID 1PV7). (b) The structural changes postulated to take place during one transport cycle. The two halves of the transporter undergo a large, reversible conformational change in which the two domains tilt relative to each other, exposing the substrate-binding site first to the periplasm (struc- ture on the right), where lactose is picked up, then to the cytoplasm (left), where the lactose is released. The interconversion of the two forms is driven by changes in the pairing of charged (protonatable) side chains such as those of Glu 325 and Arg 302 (green), which is af- fected by the transmembrane proton gradient. Apical surface Microvilli Intestinal lumen Blood Na H11001 K H11001 ATPase Basal surface Glucose Glucose Glucose uniporter GLUT2 (facilitates downhill efflux) Epithelial cell 2 Na H11001 Na H11001 - glucose symporter (driven by high extracellular [Na H11001 ]) 2 K H11001 3 Na H11001 FIGURE 11–44 Glucose transport in intestinal epithelial cells. Glucose is cotransported with Na H11001 across the apical plasma membrane into the epithelial cell. It moves through the cell to the basal surface, where it passes into the blood via GLUT2, a passive glucose transporter. The Na H11001 K H11001 ATPase continues to pump Na H11001 outward to maintain the Na H11001 gradient that drives glucose uptake. 8885d_c11_405 2/11/04 2:06 PM Page 405 mac76 mac76:385_reb: that greatly increase the surface area exposed to the intestinal contents. Na H11545 -glucose symporters in the apical plasma membrane take up glucose from the in- testine in a process driven by the downhill flow of Na H11001 : 2Na H11001 out H11001 glucose out 88n 2Na H11001 in H11001 glucose in The energy required for this process comes from two sources: the greater concentration of Na H11001 outside than inside (the chemical potential) and the transmembrane potential (the electrical potential), which is inside- negative and therefore draws Na H11001 inward. The electro- chemical potential of Na H11001 is where n H11005 2, the number of Na H11001 ions cotransported with each glucose molecule. Given the typical membrane potential of H1100250 mV, an intracellular [Na H11001 ] of 12 mM, and an extracellular [Na H11001 ] of 145 mM, the energy, H9004G, made available as two Na H11001 ions reenter the cell is 22.5 kJ, enough to pump glucose against a large concentration gradient: H9004G t H11005H1100222.5 kJ H11005 RT ln and thus H33360 9,000 That is, the cotransporter can pump glucose inward until its concentration within the epithelial cell is about 9,000 times that in the intestine. As glucose is pumped from the intestine into the epithelial cell at the apical surface, it is simultaneously moved from the cell into the blood by passive transport through a glucose trans- porter (GLUT2) in the basal surface (Fig. 11–44). The crucial role of Na H11001 in symport and antiport systems such as these requires the continued outward pumping of Na H11001 to maintain the transmembrane Na H11001 gradient. [Glucose] in H5007H5007 [Glucose] out [glucose] in H5007H5007 [glucose] out H9004G H11005 RT ln [NaH11001] in H11001 n H9004E [Na H11001 ] out Because of the essential role of ion gradients in active transport and energy conservation, compounds that collapse ion gradients across cellular membranes are effective poisons, and those that are specific for in- fectious microorganisms can serve as antibiotics. One such substance is valinomycin, a small cyclic peptide that neutralizes the K H11001 charge by surrounding it with six carbonyl oxygens (Fig. 11–45). The hydrophobic peptide then acts as a shuttle, carrying K H11001 across mem- branes down its concentration gradient and deflating that gradient. Compounds that shuttle ions across mem- branes in this way are called ionophores (“ion bear- ers”). Both valinomycin and monensin (a Na H11001 -carrying ionophore) are antibiotics; they kill microbial cells by disrupting secondary transport processes and energy- conserving reactions. Aquaporins Form Hydrophilic Transmembrane Channels for the Passage of Water A family of integral proteins discovered by Peter Agre, the aquaporins (AQPs), provide channels for rapid movement of water molecules across all plasma membranes (Table 11–6 lists a few exam- ples). Ten aquaporins are known in humans, each with its specialized role. Erythro- cytes, which swell or shrink rapidly in response to abrupt changes in extracellular os- molarity as blood travels through the renal medulla, have a high density of aqua- porin in their plasma mem- branes (2 H11003 10 5 copies of AQP-1 per cell). In the nephron (the functional unit of the kidney), the plasma membranes of proximal renal tubule cells have five different aquaporin types. Chapter 11 Biological Membranes and Transport406 FIGURE 11–45 Valinomycin, a peptide ionophore that binds K H11545 . In this image, the surface contours are shown as a transparent mesh, through which a stick structure of the peptide and a K H11001 atom (green) are visible. The oxygen atoms (red) that bind K H11001 are part of a central hydrophilic cavity. Hydrophobic amino acid side chains (yellow) coat the outside of the molecule. Because the exterior of the K H11001 - valinomycin complex is hydrophobic, the complex readily diffuses through membranes, carrying K H11001 down its concentration gradient. The resulting dissipation of the transmembrane ion gradient kills microbial cells, making valinomycin a potent antibiotic. Peter Agre 8885d_c11_406 2/11/04 12:47 PM Page 406 mac76 mac76:385_reb: These cells reabsorb water during urine formation, a process for which water movement across membranes is essential (Box 11–3). The plant Arabidopsis thaliana has 38 genes that encode various types of aquaporins, reflecting the critical roles of water move- ment in plant physiology. Changes in turgor pressure, for example, require rapid movement of water across a membrane. Water molecules flow through an AQP-1 channel at the rate of about 10 9 s H110021 . For comparison, the highest known turnover number for an enzyme is that for catalase, 4 H11003 10 7 s H110021 , and many enzymes have turnover numbers between 1 s H110021 and 10 4 s H110021 (see Table 6–7). The low activation energy for passage of water through aquaporin channels (H9004G ? H11021 15 kJ/mol) suggests that water moves through the channels in a continuous stream, in the direction dictated by the osmotic gradi- ent. (For a discussion of osmosis, see p. 57.) It is essential that aquaporins not allow passage of protons (hydronium ions, H 3 O H11001 ), which would collapse mem- brane electrochemical potentials. And they do not. What is the basis for this extraordinary selectivity? We find an answer in the structure of AQP-1, as de- termined by x-ray diffraction analysis (Fig. 11–46). AQP-1 has four monomers (each M r 28,000) associated in a tetramer, each monomer forming a transmembrane pore with a diameter (2 to 3 ?) sufficient to allow pas- sage of water molecules in single file. Each monomer consists of six transmembrane helical segments and two shorter helices, each of which contains the sequence Asn–Pro–Ala (NPA). The NPA-containing short helices extend toward the middle of the bilayer from opposite 11.3 Solute Transport across Membranes 407 (b) (a) (c) (d) FIGURE 11–46 Structure of an aquaporin, AQP-1. The protein is a tetramer of identical monomeric units, each of which forms a trans- membrane pore (derived from PBD ID 1J4N). (a) Surface model viewed perpendicular to the plane of the membrane. The protein contains four pores, one in each subunit. (The opening at the junction of the sub- units is not a pore.) (b) An AQP-1 tetramer, viewed in the plane of the membrane. The helices of each subunit cluster around a central trans- membrane pore. In each monomer, two short helical loops, one between helices 2 and 3 and the other between 5 and 6, contain the Asn–Pro–Ala (NPA) sequences found in all aquaporins, and form part of the water channel. (c) Surface representation of a single subunit, viewed in the plane of the membrane. The near side of the AQP-1 monomer has been cut away to reveal the channel running from top to bottom. The series of water molecules (orange spheres) shows the likely path of water molecules through the aquaporin channel, as pre- dicted by molecular dynamics simulations in which investigators use the properties of water and aquaporin to calculate the lowest energy states. Hydrophilic atoms that provide selective interactions with wa- ter in the channel are colored red. A Phe residue (Phe 58 ) at the con- striction is shown in blue. (d) A view down the channel, showing the constriction region of the specificity pore, which lets only a molecule as small as water pass. The side chains of Phe 58 , His 182 , Cys 191 , and Arg 197 create this constriction. 8885d_c11_407 2/11/04 12:48 PM Page 407 mac76 mac76:385_reb: sides, with their NPA regions overlapping in the middle of the membrane to form part of the specificity filter— the structure that allows only water to pass. The residues that line the channel of each AQP-1 monomer are generally nonpolar, but carbonyl oxygens in the peptide backbone, projecting into the narrow part of the channel at intervals, can form hydrogen bonds with individual water molecules as they pass through; the two Asn residues (Asn 76 and Asn 192 ) in the NPA loops also hydrogen-bond with the water. The structure does not admit closely spaced water molecules that might form a chain to allow proton hopping (see Fig. 2–14), which would effectively move protons across the membrane. Critical Arg and His residues and electric dipoles formed by the short helices of the NPA loops provide positive charges in positions that repel any pro- tons that might leak through the pore. Ion-Selective Channels Allow Rapid Movement of Ions across Membranes Ion-selective channels—first recognized in neurons and now known to be present in the plasma membranes of all cells, as well as in the intracellular membranes of eukaryotes—provide another mechanism for moving in- organic ions across membranes. Ion channels, together with ion pumps such as the Na H11001 K H11001 ATPase, determine a plasma membrane’s permeability to specific ions and regulate the cytosolic concentration of ions and the membrane potential. In neurons, very rapid changes in the activity of ion channels cause the changes in mem- brane potential (the action potentials) that carry signals from one end of a neuron to the other. In myocytes, rapid opening of Ca 2H11001 channels in the sarcoplasmic reticulum releases the Ca 2H11001 that triggers muscle con- traction. We discuss the signaling functions of ion chan- nels in Chapter 12. Here we describe the structural ba- sis for ion-channel function, using as examples a bacterial K H11001 channel, the neuronal Na H11001 channel, and the acetylcholine receptor ion channel. Ion channels are distinguished from ion trans- porters in at least three ways. First, the rate of flux through channels can be several orders of magnitude greater than the turnover number for a transporter— 10 7 to 10 8 ions/s for an ion channel, near the theoreti- cal maximum for unrestricted diffusion. Second, ion channels are not saturable: rates do not approach a max- imum at high substrate concentration. Third, they are “gated”—opened or closed in response to some cellular event. In ligand-gated channels (which are generally oligomeric), binding of an extracellular or intracellular small molecule forces an allosteric transition in the pro- tein, which opens or closes the channel. In voltage- gated ion channels, a change in transmembrane elec- trical potential (V m ) causes a charged protein domain to move relative to the membrane, opening or closing the ion channel. Both types of gating can be very fast. A channel typically opens in a fraction of a millisecond and may remain open for only milliseconds, making these molecular devices effective for very fast signal transmission in the nervous system. Ion-Channel Function Is Measured Electrically Because a single ion channel typically remains open for only a few milliseconds, monitoring this process is be- Chapter 11 Biological Membranes and Transport408 TABLE 11–6 Aquaporins Aquaporin Roles and/or location AQP-1 Fluid reabsorption in proximal renal tubule; secretion of aqueous humor in eye and cerebrospinal fluid in central nervous system; water homeostasis in lung AQP-2 Water permeability in renal collecting duct (mutations produce nephrogenic diabetes insipidus) AQP-3 Water retention in renal collecting duct AQP-4 Cerebrospinal fluid reabsorption in central nervous system; regulation of brain edema AQP-5 Fluid secretion in salivary glands, lachrymal glands, and alveolar epithelium of lung AQP-6 Kidney AQP-7 Renal proximal tubule, intestine AQP-8 Liver, pancreas, colon, placenta AQP-9 Liver, leukocytes TIP Regulation of turgor pressure in plant tonoplast PIP Plant plasma membrane AQY Yeast plasma membrane 8885d_c11_369-420 2/7/04 6:58 AM Page 408 mac76 mac76:385_reb: yond the limit of most biochemical measurements. Ion fluxes must therefore be measured electrically, either as changes in V m (in the millivolt range) or as electric cur- rents I (in the microampere or picoampere range), us- ing microelectrodes and appropriate amplifiers. In patch-clamping, a technique developed by Erwin Neher and Bert Sakmann in 1976, very small currents are measured through a tiny region of the membrane sur- face containing only one or a few ion-channel molecules (Fig. 11–47). The researcher can measure the size and duration of the current that flows during one opening of an ion channel and can determine how often a chan- nel opens and how that frequency is affected by trans- membrane potential, regulatory ligands, toxins, and other agents. Patch-clamp studies have revealed that as many as 10 4 ions can move through a single ion chan- nel in 1 ms. Such an ion flux represents a huge ampli- fication of the initial signal; for example, only two acetyl- choline molecules are needed to open an acetylcholine receptor channel (as described below). The Structure of a K H11545 Channel Reveals the Basis for Its Specificity The structure of a potassium channel from the bac- terium Streptomyces lividans, determined crystallo- graphically by Roderick MacKinnon in 1998, provides much insight into the way ion channels work. This bac- terial ion channel is related in sequence to all other known K H11001 channels and serves as the prototype for such channels, including the volt- age-gated K H11001 channel of neu- rons. Among the members of this protein family, the simi- larities in sequence are great- est in the “pore region,” which contains the ion selectivity fil- ter that allows K H11001 (radius 1.33 ?) to pass 10,000 times more readily than Na H11001 (radius 0.95 ?)—at a rate (about 10 8 ions/s) approaching the theo- retical limit for unrestricted diffusion. The K H11001 channel consists of four identical subunits that span the membrane and form a cone within a cone surrounding the ion channel, with the wide end of the double cone facing the extracellular space (Fig. 11–48). 11.3 Solute Transport across Membranes 409 Electronics to hold transmembrane potential (V m ) constant and measure current flowing across membrane Micropipette applied tightly to plasma membrane Patch of membrane pulled from cell Time Inw ard current 50ms 10pA Patch of membrane placed in aqueous solution Channel Electrodes Micropipette FIGURE 11–47 Electrical measurements of ion-channel function. The “activity” of an ion channel is estimated by measuring the flow of ions through it, using the patch-clamp technique. A finely drawn-out pipette (micropipette) is pressed against the cell surface, and negative pres- sure in the pipette forms a pressure seal between pipette and mem- brane. As the pipette is pulled away from the cell, it pulls off a tiny patch of membrane (which may contain one or a few ion channels). After placing the pipette and attached patch in an aqueous solution, the researcher can measure channel activity as the electric current that flows between the contents of the pipette and the aqueous solution. In practice, a circuit is set up that “clamps” the transmembrane po- tential at a given value and measures the current that must flow to maintain this voltage. With highly sensitive current detectors, re- searchers can measure the current flowing through a single ion chan- nel, typically a few picoamperes. The trace showing the current as a function of time (in milliseconds) reveals how fast the channel opens and closes, how frequently it opens, and how long it stays open. Clamping the V m at different values permits determination of the ef- fect of membrane potential on these parameters of channel function. Erwin Neher Bert Sakmann Roderick MacKinnon 8885d_c11_369-420 2/7/04 6:58 AM Page 409 mac76 mac76:385_reb: Each subunit has two transmembrane H9251 helices as well as a third, shorter helix that contributes to the pore re- gion. The outer cone is formed by one of the trans- membrane helices of each subunit. The inner cone, formed by the other four transmembrane helices, sur- rounds the ion channel and cradles the ion selectivity filter. Both the ion specificity and the high flux through the channel are understandable from what we know of the channel’s structure. At the inner and outer plasma membrane surfaces, the entryways to the channel have several negatively charged amino acid residues, which presumably increase the local concentration of cations such as K H11001 and Na H11001 . The ion path through the mem- brane begins (on the inner surface) as a wide, water- filled channel in which the ion can retain its hydration sphere. Further stabilization is provided by the short H9251 helices in the pore region of each subunit, with the par- tial negative charges of their electric dipoles pointed at K H11001 in the channel. About two-thirds of the way through the membrane, this channel narrows in the region of the selectivity filter, forcing the ion to give up its hydrating water molecules. Carbonyl oxygen atoms in the back- bone of the selectivity filter replace the water molecules in the hydration sphere, forming a series of perfect co- ordination shells through which the K H11001 moves. This fa- vorable interaction with the filter is not possible for Na H11001 , which is too small to make contact with all the poten- tial oxygen ligands. The preferential stabilization of K H11001 is the basis for the ion selectivity of the filter, and mu- tations that change residues in this part of the protein eliminate the channel’s ion selectivity. There are four potential K H11001 -binding sites along the selectivity filter, each composed of an oxygen “cage” that provides ligands for the K H11001 ions (Fig. 11–49). In the crystal structure, two K H11001 ions are visible within the selectivity filter, about 7.5 ? apart, and two water mol- ecules occupy the unfilled positions. K H11001 ions pass through the filter in single file; their mutual electrostatic repulsion most likely just balances the interaction of each ion with the selectivity filter and keeps them mov- ing. Movement of the two K H11001 ions is concerted: first they occupy positions 1 and 3, then they hop to positions 2 and 4 (Fig. 11–48c). The energetic difference between these two configurations (1, 3 and 2, 4) is very small; energetically, the selectivity pore is not a series of hills and valleys but a flat surface, which is ideal for rapid ion movement through the channel. The structure of the channel appears to have been optimized during evolu- tion to give maximal flow rates and high specificity. The Neuronal Na H11545 Channel Is a Voltage-Gated Ion Channel Sodium ion channels in the plasma membranes of neu- rons and of myocytes of heart and skeletal muscle sense Chapter 11 Biological Membranes and Transport410 (a) Outside Inside (b) Backbone carbonyl oxygens form cage that fits K + precisely, replacing waters of hydration sphere Alternating K + sites (blue or green) occupied Helix dipole stabilizes K + Large water-filled vestibule allows hydration of K + K + with hydrating water molecules Extracellular space Cytosol – – ++ FIGURE 11–48 Structure and function of the K H11545 channel of Strep- tomyces lividans. (PDB ID 1BL8) (a) Viewed in the plane of the mem- brane, the channel consists of eight transmembrane helices (two from each of the four identical subunits), forming a cone with its wide end toward the extracellular space. The inner helices of the cone (lighter colored) line the transmembrane channel, and the outer helices in- teract with the lipid bilayer. Short segments of each subunit converge in the open end of the cone to make a selectivity filter. (b) This view perpendicular to the plane of the membrane shows the four subunits arranged around a central channel just wide enough for a single K H11001 ion to pass. (c) Diagram of a K H11001 channel in cross section, showing the structural features critical to function. (See also Fig. 11–49.) (c) 8885d_c11_369-420 2/7/04 6:58 AM Page 410 mac76 mac76:385_reb: electrical gradients across the membrane and respond by opening or closing. These voltage-gated ion channels are typically very selective for Na H11001 over other monova- lent or divalent cations (by factors of 100 or more) and have a very high flux rate (H1102210 7 ions/s). Normally (in the resting state) in the closed conformation, Na H11001 chan- nels are opened—activated—by a reduction in the transmembrane electrical potential, then they undergo very rapid inactivation. Within milliseconds of the open- ing, the channel closes and remains inactive for many milliseconds. Activation followed by inactivation of Na H11001 channels is the basis for signaling by neurons (see Fig. 12–5). The essential component of a Na H11001 channel is a sin- gle, large polypeptide (1,840 amino acid residues) or- ganized into four domains clustered around a central channel (Fig. 11–50a, b), providing a path for Na H11001 through the membrane. The path is made Na H11001 -specific by a “pore region” composed of the segments between transmembrane helices 5 and 6 of each domain, which fold into the channel. Helix 4 of each domain has a high density of positively charged residues; this segment is believed to move within the membrane in response to changes in the transmembrane voltage, from the “rest- ing” potential of about H1100260 mV (inside negative) to about H1100130 mV. The movement of helix 4 triggers open- ing of the channel, and this is the basis for voltage gat- ing (Fig. 11–50c). Inactivation of the channel is thought to occur by a ball-and-chain mechanism. A protein domain on the cy- tosolic surface of the Na H11001 channel, the inactivation gate (the ball), is tethered to the channel by a short segment of the polypeptide (the chain) (Fig. 11–50b). This do- main is free to move about when the channel is closed, but when it opens, a site on the inner face of the chan- nel becomes available for the tethered ball to bind, blocking the channel. The length of the tether appears to determine how long an ion channel stays open; the longer the tether, the longer the open period. Inactiva- tion of other ion channels may proceed by a similar mechanism. The Acetylcholine Receptor Is a Ligand-Gated Ion Channel Another very well-studied ion channel is the nicotinic acetylcholine receptor, essential in the passage of an electrical signal from a motor neuron to a muscle fiber at the neuromuscular junction (signaling the muscle to contract). (Nicotinic receptors were originally distin- guished from muscarinic receptors by the sensitivity of the former to nicotine, the latter to the mushroom al- kaloid muscarine. They are structurally and functionally different.) Acetylcholine released by the motor neuron diffuses a few micrometers to the plasma membrane of a myocyte, where it binds to the acetylcholine receptor. This forces a conformational change in the receptor, causing its ion channel to open. The resulting inward movement of positive charges depolarizes the plasma membrane, triggering contraction. The acetylcholine re- ceptor allows Na H11001 , Ca 2H11001 , and K H11001 to pass through with equal ease, but other cations and all anions are unable to pass. Movement of Na H11001 through an acetylcholine re- ceptor ion channel is unsaturable (its rate is linear with respect to extracellular [Na H11001 ]) and very fast—about 2 H11003 10 7 ions/s under physiological conditions. This receptor channel is typical of many other ion channels that produce or respond to electrical signals: it has a “gate” that opens in response to stimulation by a signal molecule (in this case acetylcholine) and an in- trinsic timing mechanism that closes the gate after a split second. Thus the acetylcholine signal is transient— an essential feature of electrical signal conduction. We understand the structural changes underlying gating in the acetylcholine receptor, but not the exact mechanism of “desensitization”—of closing the gate even in the con- tinued presence of acetylcholine. CH 2 CH 3 H11001 N Acetylcholine O CH 2 C CH 3 CH 3 O CH 3 11.3 Solute Transport across Membranes 411 FIGURE 11–49 K H11545 binding sites in the selectivity pore of the K H11545 channel. (PDB ID 1J95) Carbonyl oxygens (red) of the peptide back- bone in the selectivity filter protrude into the channel, interacting with and stabilizing a K H11001 ion passing through. These ligands are perfectly positioned to interact with each of four K H11001 ions, but not with the smaller Na H11001 ions. This preferential interaction with K H11001 is the basis for the ion selectivity. The mutual repulsion between K H11001 ions results in occupation of only two of the four K H11001 sites at a time (both green or both blue) and counteracts the tendency for a lone K H11001 to stay bound in one site. The combined effect of K H11001 binding to carbonyl oxygens and repulsion between K H11001 ions ensures that an ion keeps moving, changing positions within 10 to 100 ns, and that there are no large energy barriers to ion flow along the path through the membrane. 8885d_c11_369-420 2/7/04 6:58 AM Page 411 mac76 mac76:385_reb: Chapter 11 Biological Membranes and Transport412 Inside Outside 1 2345 6 COO H11002 Domain Inactivation gate Voltage sensor Selectivity filter (pore region) Activation gate I II III IV NH 3 H11001 (a) Outside Inside H11001H11001H11001H11001H11001 H11002H11002H11002H11002H11002 Outside Inside Activation gate Membrane polarized, channel closed Aqueous ion channel Membrane depolarized, channel open Voltage sensor Na H11001 Na H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 H11001 (c) Selectivity filter (pore) II III IV Activation gate Voltage sensor Tether 1 2 3 4 56 Inactivation gate (open) I (b) FIGURE 11–50 Voltage-gated Na H11545 channels of neurons. Sodium channels of different tissues and organisms have a variety of subunits, but only the principal subunit (H9251) is essential. (a) The H9251 subunit is a large protein with four homologous domains (I to IV), each contain- ing six transmembrane helices (1 to 6). Helix 4 in each domain (blue) is the voltage sensor; helix 6 (orange) is thought to be the activation gate. The segments between helices 5 and 6, the pore region (red), form the selectivity filter, and the segment connecting domains III and IV (green) is the inactivation gate. (b) The four domains are wrapped about a central transmembrane channel lined with polar amino acid residues. The segments linking helices 5 and 6 (red) in each domain come together near the extracellular surface to form the selectivity fil- ter, which is conserved in all Na H11001 channels. The filter gives the chan- nel its ability to discriminate between Na H11001 and other ions of similar size. The inactivation gate (green) closes (dotted lines) soon after the activation gate opens. (c) The voltage-sensing mechanism involves movement of helix 4 (blue) perpendicular to the plane of the mem- brane in response to a change in transmembrane potential. As shown at the top, the strong positive charge on helix 4 allows it to be pulled inward in response to the inside-negative membrane potential (V m ). Depolarization lessens this pull, and helix 4 relaxes by moving out- ward (bottom). This movement is communicated to the activation gate (orange), inducing conformational changes that open the channel in response to depolarization. 8885d_c11_369-420 2/7/04 6:58 AM Page 412 mac76 mac76:385_reb: The nicotinic acetylcholine receptor has five sub- units: single copies of subunits H9252, H9253, and H9254, and two identical H9251 subunits each with an acetylcholine-binding site. All five subunits are related in sequence and terti- ary structure, each having four transmembrane helical segments (M1 to M4) (Fig. 11–51a). The five subunits surround a central pore, which is lined with their M2 helices. The pore is about 20 ? wide in the parts of the channel that protrude on the cytoplasmic and extra- cellular surfaces, but narrows as it passes through the 11.3 Solute Transport across Membranes 413 COO H11002 M1 M3 M4 M2 COO H11002 M1 M2 M3 M4 M1 M4 M2 M3 Inside Outside Acetylcholine binding sites M2 amphipathic helices surround channel (a) Subunit folds into four transmembrane a helices NH 3 H11001 H11001 a Subunit (b,g,d are homologous) bd g aa NH 3 Closed 2 Acetylcholine (b) Open Bulky hydrophobic Leu side chains of M2 helices close the channel. Binding of two acetylcholine molecules causes twisting of the M2 helices. M2 helices now have smaller, polar residues lining the channel. FIGURE 11–51 Structure of the acetylcholine receptor ion channel. (a) Each of the five subunits (H9251 2 H9252H9253H9254) has four transmembrane helices, M1 to M4. The M2 helices are amphipathic; the others have mainly hydrophobic residues. The five subunits are arranged around a central transmembrane channel, which is lined with the polar sides of the M2 helices. At the top and bottom of the channel are rings of negatively charged amino acid residues. (b) This top view of a cross section through the center of the M2 helices shows five Leu side chains (one from each M2 helix) protruding into the channel, constricting it to a diameter too small to allow passage of ions such as Ca 2H11001 , Na H11001 , and K H11001 . When both acetylcholine receptor sites (one on each H9251 subunit) are occupied, a conformational change occurs. As the M2 helices twist slightly, the five Leu residues (yellow) rotate away from the channel and are replaced by smaller, polar residues (blue). This gating mechanism opens the channel, allowing the passage of Ca 2H11001 , Na H11001 , or K H11001 . 8885d_c11_369-420 2/7/04 6:58 AM Page 413 mac76 mac76:385_reb: lipid bilayer. Near the center of the bilayer is a ring of bulky hydrophobic side chains of Leu residues in the M2 helices, positioned so close together that they prevent ions from passing through the channel. Allosteric con- formational changes induced by acetylcholine binding to the two H9251 subunits include a slight twisting of the M2 Chapter 11 Biological Membranes and Transport414 TABLE 11–7 Transport Systems Described Elsewhere in This Text Transport system and location Figure number Role Adenine nucleotide antiporter of mitochondrial 19–26 Imports substrate ADP for oxidative inner membrane phosphorylation, and exports product ATP Acyl-carnitine/carnitine transporter of mitochondrial 17–6 Imports fatty acids into matrix for H9252 oxidation inner membrane P i -H H11001 symporter of mitochondrial inner membrane 19–26 Supplies P i for oxidative phosphorylation Malate–H9251-ketoglutarate transporter of mitochondrial 19–27 Shuttles reducing equivalents (as malate) from inner membrane matrix to cytosol Glutamate-aspartate transporter of mitochondrial 19–27 Completes shuttling begun by inner membrane malate–H9251-ketoglutarate shuttle Citrate transporter of mitochondrial inner membrane 21–10 Provides cytosolic citrate as source of acetyl-CoA for lipid synthesis Pyruvate transporter of mitochondrial inner 21–10 Is part of mechanism for shuttling citrate from membrane matrix to cytosol Fatty acid transporter of myocyte plasma 17–3 Imports fatty acids for fuel membrane Complex I, III, and IV proton transporters of 19–15 Acts as energy-conserving mechanism in oxidative mitochondrial inner membrane phosphorylation, converting electron flow into proton gradient Thermogenin (uncoupler protein), a proton pore of 19–30, 23–22 Allows dissipation of proton gradient in mitochondrial inner membrane mitochondria as means of thermogenesis and/or disposal of excess fuel Cytochrome bf complex, a proton transporter of 19–50, 19–54 Acts as proton pump, driven by electron flow chloroplast thylakoid through the Z scheme; source of proton gradient for photosynthetic ATP synthesis Bacteriorhodopsin, a light-driven proton pump 19–59 Is light-driven source of proton gradient for ATP synthesis in halophilic bacterium F o F 1 ATPase/ATP synthase of mitochondrial inner 19–58 Interconverts energy of proton gradient and ATP membrane, chloroplast thylakoid, and bacterial during oxidative phosphorylation and plasma membrane photophosphorylation P i –triose phosphate antiporter of chloroplast inner 20–15, 20–16 Exports photosynthetic product from stroma; membrane imports P i for ATP synthesis Bacterial protein transporter 27–39 Exports secreted proteins through plasma membrane Protein translocase of ER 27–33 Transports into ER proteins destined for plasma membrane, secretion, or organelles Nuclear pore protein translocase 27–37 Shuttles proteins between nucleus and cytoplasm LDL receptor in animal cell plasma membrane 21–42 Imports, by receptor-mediated endocytosis, lipid carrying particles Glucose transporter of animal cell plasma 12–8 Increases capacity of muscle and adipose tissue to membrane; regulated by insulin take up excess glucose from blood IP 3 -gated Ca 2H11001 channel of endoplasmic reticulum 12–19 Allows signaling via changes of cytosolic Ca 2H11001 concentration cGMP-gated Ca 2H11001 channel of retinal rod and cone 12–32 Allows signaling via rhodopsin linked to cAMP cells phosphodiesterase in vertebrate eye Voltage-gated Na H11001 channel of neuron 12–5 Creates action potentials in neuronal signal transmission 8885d_c11_369-420 2/7/04 6:58 AM Page 414 mac76 mac76:385_reb: helices (Fig. 11–51b), which draws these hydrophobic side chains away from the center of the channel, open- ing it to the passage of ions. Based on similarities between the amino acid se- quences of other ligand-gated ion channels and the acetylcholine receptor, the receptor channels that re- spond to the extracellular signals H9253-aminobutyric acid (GABA), glycine, and serotonin have been classified in the acetylcholine receptor superfamily, and probably share three-dimensional structure and gating mecha- nisms. The GABA A and glycine receptors are anion channels specific for Cl H11002 or HCO 3 H11002 , whereas the serotonin receptor, like the acetylcholine receptor, is cation-specific. The subunits of each of these channels, like those of the acetylcholine receptor, have four transmembrane helical segments and form oligomeric channels. A second class of ligand-gated ion channels respond to intracellular ligands: 3H11032,5H11032-cyclic guanosine mono- nucleotide (cGMP) in the vertebrate eye, cGMP and cAMP in olfactory neurons, and ATP and inositol 1,4,5- trisphosphate (IP3) in many cell types. These channels are composed of multiple subunits, each with six trans- membrane helical domains. We discuss the signaling functions of these ion channels in Chapter 12. Table 11–7 shows a number of transporters not dis- cussed in this chapter but encountered later in the book in the context of the paths in which they act. Defective Ion Channels Can Have Adverse Physiological Consequences The importance of ion channels to physiological processes is clear from the effects of mutations in specific ion-channel proteins (Table 11–8). Genetic defects in the voltage-gated Na H11001 channel of the myocyte plasma membrane result in diseases in which muscles are periodically either paralyzed (as in hyperkalemic pe- riodic paralysis) or stiff (as in paramyotonia congenita). As noted earlier, cystic fibrosis is the result of a muta- tion that changes one amino acid in the protein CFTR, a Cl H11002 ion channel; the defective process here is not neu- rotransmission but secretion by various exocrine gland cells whose activities are tied to Cl H11002 ion fluxes. Many naturally occurring toxins act on ion channels, and the potency of these toxins further illustrates the importance of normal ion-channel function. Tetro- dotoxin (produced by the puffer fish, Sphaeroides rubripes) and saxitoxin (produced by the marine di- noflagellate Gonyaulax, which causes “red tides”) act by binding to the voltage-gated Na H11001 channels of neurons and preventing normal action potentials. Puffer fish is an ingredient of the Japanese delicacy fugu, which may be prepared only by chefs specially trained to separate 11.3 Solute Transport across Membranes 415 Ion channel Affected gene Disease Na H11001 (voltage-gated, skeletal muscle) SCN4A Hyperkalemic periodic paralysis (or paramyotonia congenita) Na H11001 (voltage-gated, neuronal ) SCN1A Generalized epilepsy with febrile seizures Na H11001 (voltage-gated, cardiac muscle) SCN5A Long QT syndrome 3 Ca 2H11001 (neuronal) CACNA1A Familial hemiplegic migraine Ca 2H11001 (voltage-gated, retina) CACNA1F Congenital stationary night blindness Ca 2H11001 (polycystin-1) PKD1 Polycystic kidney disease K H11001 (neuronal) KCNQ4 Dominant deafness K H11001 (voltage-gated, neuronal) KCNQ2 Benign familial neonatal convulsions Nonspecific cation (cGMP-gated, retinal) CNCG1 Retinitis pigmentosa Acetylcholine receptor (skeletal muscle) CHRNA1 Congenital myasthenic syndrome Cl H11002 CFTR Cystic fibrosis TABLE 11–8 Some Diseases Resulting from Ion Channel Defects H 2 N H 2 N HN O H HO OH N H N N H O Saxitoxin H11001 NH 2 H11001 H 2 N CH 2 OH H11001 N H H N O O OH HO HO OH O H11002 H H H H H H Tetrodotoxin 8885d_c11_369-420 2/7/04 6:58 AM Page 415 mac76 mac76:385_reb: succulent morsel from deadly poison. Eating shellfish that have fed on Gonyaulax can also be fatal; shellfish are not sensitive to saxitoxin, but they concentrate it in their muscles, which become highly poisonous to organ- isms higher up the food chain. The venom of the black mamba snake contains dendrotoxin, which interferes with voltage-gated K H11001 channels. Tubocurarine, the ac- tive component of curare (used as an arrow poison in the Amazon), and two other toxins from snake venoms, cobrotoxin and bungarotoxin, block the acetylcholine re- ceptor or prevent the opening of its ion channel. By blocking signals from nerves to muscles, all these toxins cause paralysis and possibly death. On the positive side, the extremely high affinity of bungarotoxin for the acetylcholine receptor (K d H11005 10 H1100215 M) has proved use- ful experimentally: the radiolabeled toxin was used to quantify the receptor during its purification. ■ SUMMARY 11.3 Solute Transport across Membranes ■ Movement of polar compounds and ions across biological membranes requires protein transporters. Some transporters simply facilitate passive diffusion across the membrane from the side with higher concentration to the side with lower. Others bring about active movement of solutes against an electrochemical gradient; such transport must be coupled to a source of metabolic energy. ■ Carriers, like enzymes, show saturation and stereospecificity for their substrates. Transport via these systems may be passive or active. Primary active transport is driven by ATP or electron-transfer reactions; secondary active transport, by coupled flow of two solutes, one of which (often H H11001 or Na H11001 ) flows down its electrochemical gradient as the other is pulled up its gradient. ■ The GLUT transporters, such as GLUT1 of erythrocytes, carry glucose into cells by facilitated diffusion. These transporters are uniporters, carrying only one substrate. Symporters permit simultaneous passage of two substances in the same direction; examples are the lactose transporter of E. coli, driven by the energy of a proton gradient (lactose-H H11001 symport), and the glucose transporter of intestinal epithelial cells, driven by a Na H11001 gradient (glucose-Na H11001 symport). Antiporters mediate simultaneous passage of two substances in opposite directions; examples are the chloride-bicarbonate exchanger of erythrocytes and the ubiquitous Na H11001 K H11001 ATPase. ■ In animal cells, Na H11001 K H11001 ATPase maintains the differences in cytosolic and extracellular concentrations of Na H11001 and K H11001 , and the resulting Na H11001 gradient is used as the energy source for a variety of secondary active transport processes. ■ The Na H11001 K H11001 ATPase of the plasma membrane and the Ca 2H11001 transporters of the sarcoplasmic and endoplasmic reticulums (the SERCA pumps) are examples of P-type ATPases; they undergo reversible phosphorylation during their catalytic cycle and are inhibited by the phosphate analog vanadate. F-type ATPase proton pumps (ATP synthases) are central to energy-conserving mechanisms in mitochondria and chloroplasts. V-type ATPases produce gradients of protons across some intracellular membranes, including plant vacuolar membranes. ■ ABC transporters carry a variety of substrates, including many drugs, out of cells, using ATP as energy source. ■ Ionophores are lipid-soluble molecules that bind specific ions and carry them passively across membranes, dissipating the energy of electrochemical ion gradients. ■ Water moves across membranes through aquaporins. ■ Ion channels provide hydrophilic pores through which select ions can diffuse, moving down their electrical or chemical concentration gradients; they are characteristically unsaturable and have very high flux rates. Many ion channels are highly specific for one ion, and most are gated by either voltage or a ligand. In bacterial K H11001 channels, a selectivity filter provides ligands with the right geometry to replace the water of hydration of a K H11001 ion as it crosses the membrane. Some K H11001 channels are voltage gated. The acetylcholine receptor/channel is gated by acetylcholine, which triggers subtle conformational changes that open and close the transmembrane path. Chapter 11 Biological Membranes and Transport416 O O H H H OH OH N CH 2 H 3 CO OCH 3 2Cl H11002 CH 2 H 3 CCH 3 CH 3 H11001 N H11001 D-Tubocurarine chloride 8885d_c11_369-420 2/7/04 6:58 AM Page 416 mac76 mac76:385_reb: Chapter 11 Further Reading 417 Key Terms fluid mosaic model 371 micelle 372 bilayer 373 integral proteins 373 peripheral proteins 373 hydropathy index 377 H9252 barrel 378 gel phase 380 liquid-disordered state 380 liquid-ordered state 380 flippases 382 FRAP 382 microdomains 383 rafts 384 caveolin 385 caveolae 385 fusion proteins 387 SNAREs 389 simple diffusion 389 membrane potential (V m ) 389 electrochemical gradient 391 electrochemical potential 391 facilitated diffusion 391 passive transport 391 transporters 391 carriers 392 channels 392 electroneutral 395 cotransport systems 395 antiport 397 symport 397 uniport 397 active transport 397 electrogenic 398 P-type ATPases 398 SERCA pump 400 F-type ATPases 401 ATP synthase 401 V-type ATPases 401 ABC transporters 402 multidrug transporter 402 ionophores 406 aquaporins (AQPs) 406 ion channel 408 Terms in bold are defined in the glossary. Further Reading Composition and Architecture of Membranes Boon, J.M. & Smith, B.D. (2002) Chemical control of phospho- lipid distribution across bilayer membranes. Med. Res. Rev. 22, 251–281. Intermediate-level review of phospholipid asymmetry and factors that influence it. Dowhan, W. (1997) Molecular basis for membrane phospholipids diversity: why are there so many lipids? Annu. Rev. Biochem. 66, 199–232. Ediden, M. (2002) Lipids on the frontier: a century of cell- membrane bilayers. Nat. Rev. Mol. Cell Biol. 4, 414–418. Short review of how the notion of a lipid bilayer membrane was developed and confirmed. Haltia, T. & Freire, E. (1995) Forces and factors that contribute to the structural stability of membrane proteins. Biochim. Biophys. Acta 1241, 295–322. Good discussion of the secondary and tertiary structures of membrane proteins and the factors that stabilize them. von Heijne, G. (1994) Membrane proteins: from sequence to structure. Annu. Rev. Biophys. Biomol. Struct. 23, 167–192. A review of the steps required to predict the structure of an integral protein from its sequence. White, S.H., Ladokhin, A.S., Jayasinghe, S., & Hristova, K. (2001) How membranes shape protein structure. J. Biol. Chem. 276, 32,395–32,398. Brief, intermediate-level review of the forces that shape transmembrane helices. Wimley, W.C. (2003) The versatile H9252 barrel membrane protein. Curr. Opin. Struct. Biol. 13, 1–8. Intermediate-level review. Membrane Dynamics Brown, D.A. & London, E. (1998) Functions of lipid rafts in biological membranes. Annu. Rev. Cell Dev. Biol. 14, 111–136. Chen, Y.A. & Scheller, R.H. (2001) SNARE-mediated membrane fusion. Nat. Rev. Mol. Cell Biol. 2, 98–106. Intermediate-level review. Edidin, M. (2003) The state of lipid rafts: from model membranes to cells. Annu. Rev. Biophys. Biomol. Struct. 32, 257–283. Advanced review. Frye, L.D. & Ediden, M. (1970) The rapid intermixing of cell- surface antigens after formation of mouse-human heterokaryons. J. Cell Sci. 7, 319–335. The classic demonstration of membrane protein mobility. Mayer, A. (2002) Membrane fusion in eukaryotic cells. Annu. Rev. Cell Dev. Biol. 18, 289–314. Advanced review of membrane fusion, with emphasis on the conserved general features. Parton, R.G. (2003) Caveolae—from ultrastructure to molecular mechanisms. Nat. Rev. Mol. Cell Biol. 4, 162–167. A concise historical review of caveolae, caveolin, and rafts. Sprong, H., van der Sluijs, P., & van Meer, G. (2001) How proteins move lipids and lipids move proteins. Nat. Rev. Mol. Cell Biol. 2, 504–513. Intermediate-level review. van Deurs, B., Roepstorff, K., Hommelgaard, A.M., & Sandvig, K. (2003) Caveolae: anchored, multifunctional platforms in the lipid ocean. Trends Cell Biol. 13, 92–100. Vereb, G., Sz?llósi, J., Matcó, J., Nagy, P., Farkas, T., Vigh, L., Mátyus, L., Waldmann, T.A., & Damjanovich, S. (2003) Dynamic, yet structured: the cell membrane three decades after the Singer-Nicolson model. Proc. Natl. Acad. Sci. USA 100, 8053–8058. Intermediate-level review of membrane structure and dynamics. Transporters Abramson, J., Smirnova, I., Kasho, V., Verner, G., Kaback, H.R., & Iwata, S. (2003) Structure and mechanism of the lactose permease of Escherichia coli. Science 301, 610–615. 8885d_c11_369-420 2/7/04 6:58 AM Page 417 mac76 mac76:385_reb: Chapter 11 Biological Membranes and Transport418 Fujiyoshi, Y., Mitsuoka, K., de Groot, B.L., Philippsen, A., Grubmüller, H., Agre, P., & Engel, A. (2002) Structure and function of water channels. Curr. Opin. Struct. Biol. 12, 509–515. Jorgensen, P.L., H?kansson, K.O., & Karlish, S.J.D. (2003) Structure and mechanism of Na,K-ATPase: functional sites and their interactions. Annu. Rev. Physiol. 65, 817–849. Kjellbom, P., Larsson, C., Johansson, I., Karlsson, M., & Johanson, U. (1999) Aquaporins and water homeostasis in plants. Trends Plant Sci. 4, 308–314. Intermediate-level review. Mueckler, M. (1994) Facilitative glucose transporters. Eur. J. Biochem. 219, 713–725. Saier, M.H., Jr. (2000) A functional-phylogenetic classification system for transmembrane solute transporters. Microbiol. Mol. Biol. Rev. 64, 354–411. Schmitt, L. & Tampé, R. (2002) Structure and mechanism of ABC transporters. Curr. Opin. Struct. Biol. 12, 754–760. Sheppard, D.N. & Welsh, M.J. (1999) Structure and function of the CFTR chloride channel. Physiol. Rev. 79, S23–S46. This issue of the journal has 11 reviews on the CFTR chloride channel, covering its structure, activity, regulation, biosynthesis, and pathophysiology. Stokes, D.L. & Green, N.M. (2003) Structure and function of the calcium pump. Annu. Rev. Biophys. Biomol. Struct. 32, 445–468. Advanced review. Sui, H., Han, B.-G., Lee, J.K., Walian, P., & Jap, B.K. (2001) Structural basis of water-specific transport through the AQP1 water channel. Nature 414, 872–878. High-resolution solution of the aquaporin structure by x-ray crystallography. Ion Channels Changeux, J.P. (1993) Chemical signaling in the brain. Sci. Am. 269 (November), 58–62. Discussion of structure and function of the acetylcholine receptor channel. Doyle, D.A., Cabral, K.M., Pfuetzner, R.A., Kuo, A., Gulbis, J.M., Cohen, S.L., Chait, B.T., & MacKinnon, R. (1998) The structure of the potassium channel: molecular basis of K H11001 conduction and selectivity. Science 280, 69–77. The first crystal structure of an ion channel is described. Edelstein, S.J. & Changeux, J.P. (1998) Allosteric transitions of the acetylcholine receptor. Adv. Prot. Chem. 51, 121–184. Advanced discussion of the conformational changes induced by acetylcholine. Hille, B. (2001) Ion Channels of Excitable Membranes, 3rd edn, Sinauer Associates, Sunderland, MA. Intermediate-level text emphasizing the function of ion channels. Jiang, Y., Lee, A., Chen, J., Ruta, V., Cadene, M., Chait, B.T., & MacKinnon, R. (2003) X-ray structure of a voltage-dependent K H11001 channel. Nature 423, 33–41. Lee, A.G. & East, J.M. (2001) What the structure of a calcium pump tells us about its mechanism. Biochem J. 356, 665–683. Miyazawa, A., Fujiyoshi, Y., & Unwin, N. (2003) Structure and gating mechanism of the acetylcholine receptor pore. Nature 423, 949–955. Intermediate-level review. Neher, E. & Sakmann, B. (1992) The patch clamp technique. Sci. Am. (March) 266, 44–51. Clear description of the electrophysiological methods used to measure the activity of single ion channels, by the Nobel Prize–winning developers of this technique. Yellen, G. (2002) The voltage-gated potassium channels and their relatives. Nature 419, 35–42. Zhou, Y., Morias-Cabral, J.H., Kaufman, A., & MacKinnon, R. (2001) Chemistry of ion coordination and hydration revealed by a K H11001 channel–Fab complex at 2.0 ? resolution. Nature 414, 43–48. 1. Determining the Cross-Sectional Area of a Lipid Molecule When phospholipids are layered gently onto the surface of water, they orient at the air-water interface with their head groups in the water and their hydrophobic tails in the air. An experimental apparatus (a) has been devised that reduces the surface area available to a layer of lipids. By meas- uring the force necessary to push the lipids together, it is pos- sible to determine when the molecules are packed tightly in a continuous monolayer; as that area is approached, the force needed to further reduce the surface area increases sharply (b). How would you use this apparatus to determine the av- erage area occupied by a single lipid molecule in the monolayer? 2. Evidence for a Lipid Bilayer In 1925, E. Gorter and F. Grendel used an apparatus like that described in Problem 1 to determine the surface area of a lipid monolayer formed by lipids extracted from erythrocytes of several animal species. They used a microscope to measure the dimensions of indi- vidual cells, from which they calculated the average surface area of one erythrocyte. They obtained the data shown in the Problems Force applied here to compress monolayer (a) Force (dyne/cm) Area (nm 2 /molecule) (b) 40 30 1.41.00.60.2 20 10 8885d_c11_369-420 2/7/04 6:58 AM Page 418 mac76 mac76:385_reb: Chapter 11 Problems 419 table. Were these investigators justified in concluding that “chromocytes [erythrocytes] are covered by a layer of fatty sub- stances that is two molecules thick” (i.e., a lipid bilayer)? Total surface Volume of Number area of lipid Total surface packed of cells monolayer area of one Animal cells (mL) (per mm 3 ) from cells (m 2 ) cell (H9262m 2 ) Dog 40 8,000,000 62 98 Sheep 10 9,900,000 6.0 29.8 Human 1 4,740,000 0.92 99.4 Source: Data from Gorter, E. & Grendel, F. (1925) On bimolecular layers of lipoids on the chromocytes of the blood. J. Exp. Med. 41, 439–443. 3. Number of Detergent Molecules per Micelle When a small amount of sodium dodecyl sulfate (SDS; Na H11001 CH 3 (CH 2 ) 11 OSO 3 H11002 ) is dissolved in water, the detergent ions enter the solution as monomeric species. As more de- tergent is added, a concentration is reached (the critical mi- celle concentration) at which the monomers associate to form micelles. The critical micelle concentration of SDS is 8.2 mM. The micelles have an average particle weight (the sum of the molecular weights of the constituent monomers) of 18,000. Calculate the number of detergent molecules in the average micelle. 4. Properties of Lipids and Lipid Bilayers Lipid bilay- ers formed between two aqueous phases have this important property: they form two-dimensional sheets, the edges of which close upon each other and undergo self-sealing to form liposomes. (a) What properties of lipids are responsible for this property of bilayers? Explain. (b) What are the consequences of this property for the structure of biological membranes? 5. Length of a Fatty Acid Molecule The carbon–carbon bond distance for single-bonded carbons such as those in a saturated fatty acyl chain is about 1.5 ?. Estimate the length of a single molecule of palmitate in its fully extended form. If two molecules of palmitate were placed end to end, how would their total length compare with the thickness of the lipid bilayer in a biological membrane? 6. Temperature Dependence of Lateral Diffusion The experiment described in Figure 11–17 was performed at 37 H11034C. If the experiment were carried out at 10 H11034C, what ef- fect would you expect on the rate of diffusion? Why? 7. Synthesis of Gastric Juice: Energetics Gastric juice (pH 1.5) is produced by pumping HCl from blood plasma (pH 7.4) into the stomach. Calculate the amount of free en- ergy required to concentrate the H H11001 in 1 L of gastric juice at 37 H11034C. Under cellular conditions, how many moles of ATP must be hydrolyzed to provide this amount of free energy? The free-energy change for ATP hydrolysis under cellular condi- tions is about H1100258 kJ/mol (as explained in Chapter 13). Ig- nore the effects of the transmembrane electrical potential. 8. Energetics of the Na H11545 K H11545 ATPase For a typical ver- tebrate cell with a transmembrane potential of H110020.070 V (in- side negative), what is the free-energy change for transport- ing 1 mol of Na H11001 out of the cell and into the blood at 37 H11034C? Assume the concentration of Na H11001 inside the cell is 12 mM, and that in blood plasma is 145 mM. 9. Action of Ouabain on Kidney Tissue Ouabain specif- ically inhibits the Na H11001 K H11001 ATPase activity of animal tissues but is not known to inhibit any other enzyme. When ouabain is added to thin slices of living kidney tissue, it inhibits oxygen consumption by 66%. Why? What does this observation tell us about the use of respiratory energy by kidney tissue? 10. Energetics of Symport Suppose that you determined experimentally that a cellular transport system for glucose, driven by symport of Na H11001 , could accumulate glucose to con- centrations 25 times greater than in the external medium, while the external [Na H11001 ] was only 10 times greater than the intracellular [Na H11001 ]. Would this violate the laws of thermody- namics? If not, how could you explain this observation? 11. Location of a Membrane Protein The following ob- servations are made on an unknown membrane protein, X. It can be extracted from disrupted erythrocyte membranes into a concentrated salt solution, and it can be cleaved into frag- ments by proteolytic enzymes. Treatment of erythrocytes with proteolytic enzymes followed by disruption and extrac- tion of membrane components yields intact X. However, treat- ment of erythrocyte “ghosts” (which consist of just plasma membranes, produced by disrupting the cells and washing out the hemoglobin) with proteolytic enzymes followed by disruption and extraction yields extensively fragmented X. What do these observations indicate about the location of X in the plasma membrane? Do the properties of X resemble those of an integral or peripheral membrane protein? 12. Membrane Self-sealing Cellular membranes are self- sealing—if they are punctured or disrupted mechanically, they quickly and automatically reseal. What properties of membranes are responsible for this important feature? 13. Lipid Melting Temperatures Membrane lipids in tis- sue samples obtained from different parts of the leg of a rein- deer have different fatty acid compositions. Membrane lipids from tissue near the hooves contain a larger proportion of un- saturated fatty acids than those from tissue in the upper leg. What is the significance of this observation? 14. Flip-Flop Diffusion The inner leaflet (monolayer) of the human erythrocyte membrane consists predominantly of phosphatidylethanolamine and phosphatidylserine. The outer leaflet consists predominantly of phosphatidylcholine and sphingomyelin. Although the phospholipid components of the membrane can diffuse in the fluid bilayer, this sidedness is preserved at all times. How? 15. Membrane Permeability At pH 7, tryptophan crosses a lipid bilayer at about one-thousandth the rate of the closely related substance indole: Suggest an explanation for this observation. N H 8885d_c11_369-420 2/7/04 6:58 AM Page 419 mac76 mac76:385_reb: Chapter 11 Biological Membranes and Transport420 16. Water Flow through an Aquaporin Each human erythrocyte has about 2 H11003 10 5 AQP-1 monomers. If water molecules flow through the plasma membrane at a rate of 5 H11003 10 8 per AQP-1 tetramer per second, and the volume of an erythrocyte is 5 H11003 10 H1100211 mL, how rapidly could an ery- throcyte halve its volume as it encounters the high osmolar- ity (1 M) in the interstitial fluid of the renal medulla? Assume that the erythrocyte consists entirely of water. 17. Labeling the Lactose Transporter A bacterial lac- tose transporter, which is highly specific for its substrate lac- tose, contains a Cys residue that is essential to its transport activity. Covalent reaction of N-ethylmaleimide (NEM) with this Cys residue irreversibly inactivates the transporter. A high concentration of lactose in the medium prevents inacti- vation by NEM, presumably by sterically protecting the Cys residue, which is in or near the lactose-binding site. You know nothing else about the transporter protein. Suggest an ex- periment that might allow you to determine the M r of the Cys- containing transporter polypeptide. 18. Predicting Membrane Protein Topology from Se- quence You have cloned the gene for a human erythrocyte protein, which you suspect is a membrane protein. From the nucleotide sequence of the gene, you know the amino acid sequence. From this sequence alone, how would you evalu- ate the possibility that the protein is an integral protein? Sup- pose the protein proves to be an integral protein, either type I or type II. Suggest biochemical or chemical experiments that might allow you to determine which type it is. 19. Intestinal Uptake of Leucine You are studying the uptake of L-leucine by epithelial cells of the mouse intestine. Measurements of the rate of uptake of L-leucine and several of its analogs, with and without Na H11001 in the assay buffer, yield the results given in the table. What can you conclude about the properties and mechanism of the leucine transporter? Would you expect L-leucine uptake to be inhibited by ouabain? Uptake in Uptake in presence of Na H11001 absence of Na H11001 Substrate V max K t (mM)V max K t (mM) L-Leucine 420 0.24 23 0.24 D-Leucine 310 4.7 5 4.7 L-Valine 225 0.31 19 0.31 20. Effect of an Ionophore on Active Transport Con- sider the leucine transporter described in Problem 19. Would V max and/or K t change if you added a Na H11001 ionophore to the assay solution containing Na H11001 ? Explain. 21. Surface Density of a Membrane Protein E. coli can be induced to make about 10,000 copies of the lactose trans- porter (M r 31,000) per cell. Assume that E. coli is a cylinder 1 H9262m in diameter and 2 H9262m long. What fraction of the plasma membrane surface is occupied by the lactose transporter mol- ecules? Explain how you arrived at this conclusion. Biochemistry on the Internet 22. Membrane Protein Topology The receptor for the hormone epinephrine in animal cells is an integral membrane protein (M r 64,000) that is believed to have seven membrane- spanning regions. (a) Show that a protein of this size is capable of span- ning the membrane seven times. (b) Given the amino acid sequence of this protein, how would you predict which regions of the protein form the membrane-spanning helices? (c) Go to the Protein Data Bank (www.rcsb.org/pdb). Use the PDB identifier 1DEP to retrieve the data page for a portion of the H9252-adrenergic receptor (one type of epineph- rine receptor) from a turkey. Using Chime to explore the structure, predict where this portion of the receptor is lo- cated: within the membrane or at the membrane surface. Explain. (d) Retrieve the data for a portion of another receptor, the acetylcholine receptor of neurons and myocytes, using the PDB identifier 1A11. As in (c), predict where this por- tion of the receptor is located and explain your answer. If you have not used the PDB or Chemscape Chime, you can find instructions at www.whfreeman.com/lehninger. 8885d_c11_420 2/11/04 12:48 PM Page 420 mac76 mac76:385_reb: chapter T he ability of cells to receive and act on signals from beyond the plasma membrane is fundamental to life. Bacterial cells receive constant input from membrane proteins that act as information receptors, sampling the surrounding medium for pH, osmotic strength, the avail- ability of food, oxygen, and light, and the presence of noxious chemicals, predators, or competitors for food. These signals elicit appropriate responses, such as mo- tion toward food or away from toxic substances or the formation of dormant spores in a nutrient-depleted medium. In multicellular organisms, cells with different functions exchange a wide variety of signals. Plant cells respond to growth hormones and to variations in sun- light. Animal cells exchange information about the con- centrations of ions and glucose in extracellular fluids, the interdependent metabolic activities taking place in different tissues, and, in an embryo, the correct place- ment of cells during development. In all these cases, the signal represents information that is detected by spe- cific receptors and converted to a cellular response, which always involves a chemical process. This con- version of information into a chemical change, signal transduction, is a universal property of living cells. The number of different biological signals is large (Table 12–1), as is the variety of biological responses to these signals, but organisms use just a few evolutionar- ily conserved mechanisms to detect extracellular signals and transduce them into intracellular changes. In this chapter we examine some examples of the major classes of signaling mechanisms, looking at how they are inte- grated in specific biological functions such as the trans- mission of nerve signals; responses to hormones and growth factors; the senses of sight, smell, and taste; and BIOSIGNALING 12.1 Molecular Mechanisms of Signal Transduction 422 12.2 Gated Ion Channels 425 12.3 Receptor Enzymes 429 12.4 G Protein–Coupled Receptors and Second Messengers 435 12.5 Multivalent Scaffold Proteins and Membrane Rafts 443 12.6 Signaling in Microorganisms and Plants 452 12.7 Sensory Transduction in Vision, Olfaction, and Gustation 456 12.8 Regulation of Transcription by Steroid Hormones 465 12.9 Regulation of the Cell Cycle by Protein Kinases 466 12.10 Oncogenes, Tumor Suppressor Genes, and Programmed Cell Death 471 When I first entered the study of hormone action, some 25 years ago, there was a widespread feeling among biologists that hormone action could not be studied meaningfully in the absence of organized cell structure. However, as I reflected on the history of biochemistry, it seemed to me there was a real possibility that hormones might act at the molecular level. —Earl W. Sutherland, Nobel Address, 1971 12 421 Antigens Cell surface glycoproteins/ oligosaccharides Developmental signals Extracellular matrix components Growth factors Hormones Light Mechanical touch Neurotransmitters Nutrients Odorants Pheromones Tastants Some Signals to Which Cells Respond TABLE 12–1 () 8885d_c12_421 2/23/04 9:11 AM Page 421 mac76 mac76: control of the cell cycle. Often, the end result of a sig- naling pathway is the phosphorylation of a few specific target-cell proteins, which changes their activities and thus the activities of the cell. Throughout our discus- sion we emphasize the conservation of fundamental mechanisms for the transduction of biological signals and the adaptation of these basic mechanisms to a wide range of signaling pathways. 12.1 Molecular Mechanisms of Signal Transduction Signal transductions are remarkably specific and exquisitely sensitive. Specificity is achieved by precise molecular complementarity between the signal and re- ceptor molecules (Fig. 12–1a), mediated by the same kinds of weak (noncovalent) forces that mediate enzyme-substrate and antigen-antibody interactions. Multicellular organisms have an additional level of speci- ficity, because the receptors for a given signal, or the intracellular targets of a given signal pathway, are pres- ent only in certain cell types. Thyrotropin-releasing hor- mone, for example, triggers responses in the cells of the anterior pituitary but not in hepatocytes, which lack re- ceptors for this hormone. Epinephrine alters glycogen metabolism in hepatocytes but not in erythrocytes; in this case, both cell types have receptors for the hor- mone, but whereas hepatocytes contain glycogen and the glycogen-metabolizing enzyme that is stimulated by epinephrine, erythrocytes contain neither. Three factors account for the extraordinary sensi- tivity of signal transducers: the high affinity of recep- tors for signal molecules, cooperativity (often but not always) in the ligand-receptor interaction, and amplifi- cation of the signal by enzyme cascades. The affinity between signal (ligand) and receptor can be expressed as the dissociation constant K d , usually 10 H1100210 M or less—meaning that the receptor detects picomolar concentrations of a signal molecule. Receptor-ligand in- teractions are quantified by Scatchard analysis, which yields a quantitative measure of affinity (K d ) and the number of ligand-binding sites in a receptor sam- ple (Box 12–1). Cooperativity in receptor-ligand interactions re- sults in large changes in receptor activation with small changes in ligand concentration (recall the effect of co- operativity on oxygen binding to hemoglobin; see Fig. 5–12). Amplification by enzyme cascades results when an enzyme associated with a signal receptor is ac- tivated and, in turn, catalyzes the activation of many molecules of a second enzyme, each of which activates many molecules of a third enzyme, and so on (Fig. 12–1b). Such cascades can produce amplifications of several orders of magnitude within milliseconds. The sensitivity of receptor systems is subject to modification. When a signal is present continuously, desensitization of the receptor system results (Fig. 12–1c); when the stimulus falls below a certain thresh- old, the system again becomes sensitive. Think of what happens to your visual transduction system when you walk from bright sunlight into a darkened room or from darkness into the light. A final noteworthy feature of signal-transducing systems is integration (Fig. 12–1d), the ability of the system to receive multiple signals and produce a uni- fied response appropriate to the needs of the cell or or- ganism. Different signaling pathways converse with Chapter 12 Biosignaling422 Receptor Response Signal Receptor 1 Signal 1 Receptor 2 Signal 2 (c) Desensitization/Adaptation Receptor activation triggers a feedback circuit that shuts off the receptor or removes it from the cell surface. (d) Integration When two signals have opposite effects on a metabolic characteristic such as the concentration of a second messenger X, or the membrane potential V m , the regulatory outcome results from the integrated input from both receptors. [X] or V m [X] or V m Response Net H9004[X] or V m Enzyme 3 33 333333 Enzyme 2 2 Enzyme 1 Signal 2 3 Effect S 1 S 2 (a) Specificity Signal molecule fits binding site on its complementary receptor; other signals do not fit. (b) Amplification When enzymes activate enzymes, the number of affected molecules increases geometrically in an enzyme cascade. Receptor FIGURE 12–1 Four features of signal-transducing systems. 8885d_c12_422 2/20/04 1:13 PM Page 422 mac76 mac76:385_reb: BOX 12–1 WORKING IN BIOCHEMISTRY Scatchard Analysis Quantifies the Receptor-Ligand Interaction The cellular actions of a hormone begin when the hor- mone (ligand, L) binds specifically and tightly to its protein receptor (R) on or in the target cell. Binding is mediated by noncovalent interactions (hydrogen- bonding, hydrophobic, and electrostatic) between the complementary surfaces of ligand and receptor. Receptor-ligand interaction brings about a conforma- tional change that alters the biological activity of the receptor, which may be an enzyme, an enzyme regu- lator, an ion channel, or a regulator of gene expression. Receptor-ligand binding is described by the equation R H11001 L 34 RL Receptor Ligand Receptor-ligand complex This binding, like that of an enzyme to its substrate, depends on the concentrations of the interacting com- ponents and can be described by an equilibrium con- stant: k H110011 R H11001 L 34 RL Receptor Ligand k H110021 Receptor-ligand complex K a H11005H11005H110051/K d where K a is the association constant and K d is the dis- sociation constant. Like enzyme-substrate binding, receptor-ligand binding is saturable. As more ligand is added to a fixed amount of receptor, an increasing fraction of receptor molecules is occupied by ligand (Fig. 1a). A rough measure of receptor-ligand affinity is given by the con- centration of ligand needed to give half-saturation of the receptor. Using Scatchard analysis of receptor- ligand binding, we can estimate both the dissociation constant K d and the number of receptor-binding sites in a given preparation. When binding has reached equilibrium, the total number of possible binding sites, B max , equals the number of unoccupied sites, repre- sented by [R], plus the number of occupied or ligand- bound sites, [RL]; that is, B max H11005 [R] H11001 [RL]. The num- ber of unbound sites can be expressed in terms of total sites minus occupied sites: [R] H11005 B max H11002 [RL]. The equilibrium expression can now be written K a H11005 Rearranging to obtain the ratio of receptor-bound lig- and to free (unbound) ligand, we get H11005H11005K a (B max H11002 [RL]) H11005 (B max H11002 [RL]) 1 H5007 K d [RL] H5007 [L] [Bound] H5007 [Free] [RL] H5007H5007H5007 [L](B max H11002 [RL]) k H110011 H5007 k H110021 [RL] H5007 [R][L] From this slope-intercept form of the equation, we can see that a plot of [bound ligand]/[free ligand] versus [bound ligand] should give a straight line with a slope of H11002K a (H110021/K d ) and an intercept on the abscissa of B max , the total number of binding sites (Fig. 1b). Hormone- ligand interactions typically have K d values of 10 H110029 to 10 H1100211 M, corresponding to very tight binding. Scatchard analysis is reliable for the simplest cases, but as with Lineweaver-Burk plots for enzymes, when the receptor is an allosteric protein, the plots deviate from linearity. Bound hormone , [ RL] Total hormone added, [L] H11001 [RL] Total binding Specific binding Nonspecific binding (a) Bound hormone F ree hormone , [ RL] [L ] Bound hormone, [RL](b) Slope H11005 H11002 1 K d B max FIGURE 1 Scatchard analysis of a receptor-ligand interaction. A radiolabeled ligand (L)—a hormone, for example—is added at sev- eral concentrations to a fixed amount of receptor (R), and the frac- tion of the hormone bound to receptor is determined by separating the receptor-hormone complex (RL) from free hormone. (a) A plot of [RL] versus [L] H11001 [RL] (total hormone added) is hyperbolic, rising toward a maximum for [RL] as the receptor sites become saturated. To control for nonsaturable, nonspecific binding sites (eicosanoid hormones bind nonspecifically to the lipid bilayer, for example), a separate series of binding experiments is also necessary. A large excess of unlabeled hormone is added along with the dilute so- lution of labeled hormone. The unlabeled molecules compete with the labeled molecules for specific binding to the saturable site on the receptor, but not for the nonspecific binding. The true value for specific binding is obtained by subtracting nonspecific binding from total binding. (b) A linear plot of [RL]/[L] versus [RL] gives K d and B max for the receptor-hormone complex. Compare these plots with those of V 0 versus [S] and 1/V 0 versus 1/[S] for an enzyme-substrate complex (see Fig. 6–12, Box 6–1). 8885d_c12_423 2/20/04 1:13 PM Page 423 mac76 mac76:385_reb: each other at several levels, generating a wealth of in- teractions that maintain homeostasis in the cell and the organism. We consider here the molecular details of several representative signal-transduction systems. The trigger for each system is different, but the general features of signal transduction are common to all: a signal interacts with a receptor; the activated receptor interacts with cellular machinery, producing a second signal or a change in the activity of a cellular protein; the meta- bolic activity (broadly defined to include metabolism of RNA, DNA, and protein) of the target cell undergoes a change; and finally, the transduction event ends and the cell returns to its prestimulus state. To illustrate these general features of signaling systems, we provide examples of each of six basic signaling mechanisms (Fig. 12–2). 1. Gated ion channels of the plasma membrane that open and close (hence the term “gating”) in response to the binding of chemical ligands or changes in transmembrane potential. These are the simplest signal transducers. The acetylcholine receptor ion channel is an example of this mechanism (Section 12.2). 2. Receptor enzymes, plasma membrane receptors that are also enzymes. When one of these receptors is activated by its extracellular ligand, it catalyzes the production of an intracellular second messenger. An example is the insulin receptor (Section 12.3). 3. Receptor proteins (serpentine receptors) that indirectly activate (through GTP-binding proteins, or G proteins) enzymes that generate intracellular second messengers. This is illustrated by the H9252-adrenergic receptor system that detects epinephrine (adrenaline) (Section 12.4). 4. Nuclear receptors (steroid receptors) that, when bound to their specific ligand (such as the hormone estrogen), alter the rate at which specific genes are transcribed and translated into cellular proteins. Because steroid hormones function through mechanisms intimately related to the regulation of gene expression, we consider them here only briefly (Section 12.8) and defer a detailed discussion of their action until Chapter 28. 5. Receptors that lack enzymatic activity but attract and activate cytoplasmic enzymes that act on downstream proteins, either by directly converting them to gene-regulating proteins or by activating a cascade of enzymes that finally activates a gene regulator. The JAK-STAT system exemplifies the first mechanism (Section 12.3); and the TLR4 (Toll) signaling system in humans, the second (Section 12.6). Chapter 12 Biosignaling424 FIGURE 12–2 Six general types of signal transducers. S SS S Ion S S S S Gated ion channel Opens or closes in response to concentration of signal ligand (S) or membrane potential. Receptor enzyme Ligand binding to extracellular domain stimulates enzyme activity in intracellular domain. Plasma membrane Nuclear envelope DNA Steroid receptor Steroid binding to a nuclear receptor protein allows the receptor to regulate the expression of specific genes. Serpentine receptor External ligand binding to receptor (R) activates an intracellular GTP-binding protein (G), which regulates an enzyme (Enz) that generates an intracellular second messenger, X. Receptor with no intrinsic enzyme activity Interacts with cytosolic protein kinase, which activates a gene-regulating protein (directly or through a cascade of protein kinases), changing gene expression. Kinase cascade R G Enz mRNA Protein mRNA DNA Protein X S S Adhesion receptor Binds molecules in extracellular matrix, changes conformation, thus altering its interaction with cytoskeleton. 8885d_c12_424 2/20/04 1:16 PM Page 424 mac76 mac76:385_reb: 6. Receptors (adhesion receptors) that interact with macromolecular components of the extracellular matrix (such as collagen) and convey to the cytoskeletal system instructions on cell migration or adherence to the matrix. Integrins (discussed in Chapter 10) illustrate this general type of transduction mechanism. As we shall see, transductions of all six types commonly require the activation of protein kinases, enzymes that transfer a phosphoryl group from ATP to a protein side chain. SUMMARY 12.1 Molecular Mechanisms of Signal Transduction ■ All cells have specific and highly sensitive signal-transducing mechanisms, which have been conserved during evolution. ■ A wide variety of stimuli, including hormones, neurotransmitters, and growth factors, act through specific protein receptors in the plasma membrane. ■ The receptors bind the signal molecule, amplify the signal, integrate it with input from other receptors, and transmit it into the cell. If the signal persists, receptor desensitization reduces or ends the response. ■ Eukaryotic cells have six general types of signaling mechanisms: gated ion channels; receptor enzymes; membrane proteins that act through G proteins; nuclear proteins that bind steroids and act as transcription factors; membrane proteins that attract and activate soluble protein kinases; and adhesion receptors that carry information between the extracellular matrix and the cytoskeleton. 12.2 Gated Ion Channels Ion Channels Underlie Electrical Signaling in Excitable Cells The excitability of sensory cells, neurons, and myocytes depends on ion channels, signal transducers that pro- vide a regulated path for the movement of inorganic ions such as Na H11001 , K H11001 , Ca 2H11001 , and Cl H11002 across the plasma mem- brane in response to various stimuli. Recall from Chap- ter 11 that these ion channels are “gated”; they may be open or closed, depending on whether the associated receptor has been activated by the binding of its spe- cific ligand (a neurotransmitter, for example) or by a change in the transmembrane electrical potential, V m . The Na H11001 K H11001 ATPase creates a charge imbalance across the plasma membrane by carrying 3 Na H11001 out of the cell for every 2 K H11001 carried in (Fig. 12–3a), making the in- side negative relative to the outside. The membrane is said to be polarized. By convention, V m is negative when the inside of the cell is negative relative to the outside. For a typical animal cell, V m H11005H1100260 to H1100270 mV. Because ion channels generally allow passage of ei- ther anions or cations but not both, ion flux through a channel causes a redistribution of charge on the two sides of the membrane, changing V m . Influx of a posi- tively charged ion such as Na H11001 , or efflux of a negatively charged ion such as Cl H11002 , depolarizes the membrane and brings V m closer to zero. Conversely, efflux of K H11001 hy- perpolarizes the membrane and V m becomes more neg- ative. These ion fluxes through channels are passive, in contrast to active transport by the Na H11001 K H11001 ATPase. The direction of spontaneous ion flow across a polarized membrane is dictated by the electrochemical 12.2 Gated Ion Channels 425 High High High Low Low Low Low High The electrogenic Na + K + ATPase establishes the membrane potential. Ions tend to move down their electrochemical gradient across the polarized membrane. ATP ADP H11545 P i Na H11545 K H11545 ATPase 2 K H11545 [Na H11545 ] [K H11545 ] [Ca 2H11545 ] [Cl H11546 ] 3 Na H11545 H11545H11545 H11546H11546 H11545 H11546 H11545H11545 H11546H11546 H11545 H11546 H11546 H11545 H11546 H11545 H11546 H11545 H11546 H11545 H11546 H11545 H11546 H11545 H11545H11546 H11545H11546 H11545H11546 H11546H11545 H11546 H11545 H11546 H11545 +H11546 +H11546 Plasma membrane (a) (b) FIGURE 12–3 Transmembrane electrical potential. (a) The electro- genic Na H11001 K H11001 ATPase produces a transmembrane electrical potential of H1100260 mV (inside negative). (b) Blue arrows show the direction in which ions tend to move spontaneously across the plasma membrane in an animal cell, driven by the combination of chemical and elec- trical gradients. The chemical gradient drives Na H11001 and Ca 2H11001 inward (producing depolarization) and K H11001 outward (producing hyperpolar- ization). The electrical gradient drives Cl H11002 outward, against its con- centration gradient (producing depolarization). 8885d_c12_425 2/20/04 1:16 PM Page 425 mac76 mac76:385_reb: potential of that ion across the membrane. The force (H9004G) that causes a cation (say, Na H11001 ) to pass sponta- neously inward through an ion channel is a function of the ratio of its concentrations on the two sides of the membrane (C in /C out ) and of the difference in electrical potential (H9004H9274 or V m ): H9004G H11005 RT ln H20898H20899 H11001 ZV m (12–1) where R is the gas constant, T the absolute tempera- ture, Z the charge on the ion, and the Faraday con- stant. In a typical neuron or myocyte, the concentra- tions of Na H11001 , K H11001 , Ca 2H11001 , and Cl H11002 in the cytosol are very different from those in the extracellular fluid (Table 12–2). Given these concentration differences, the rest- ing V m of H1100260 mV, and the relationship shown in Equa- tion 12–1, opening of a Na H11001 or Ca 2H11001 channel will result in a spontaneous inward flow of Na H11001 or Ca 2H11001 (and depolarization), whereas opening of a K H11001 channel will result in a spontaneous outward flux of K H11001 (and hyperpolarization) (Fig. 12–3b). A given ionic species continues to flow through a channel only as long as the combination of concentra- tion gradient and electrical potential provides a driving force, according to Equation 12–1. For example, as Na H11001 flows down its concentration gradient it depolarizes the membrane. When the membrane potential reaches H1100170 mV, the effect of this membrane potential (to resist further entry of Na H11001 ) exactly equals the effect of the Na H11001 concentration gradient (to cause more Na H11001 to flow inward). At this equilibrium potential (E), the driving force (H9004G) tending to move an ion is zero. The equilib- rium potential is different for each ionic species because the concentration gradients differ for each ion. The number of ions that must flow to change the membrane potential significantly is negligible relative to the concentrations of Na H11001 , K H11001 , and Cl H11002 in cells and ex- tracellular fluid, so the ion fluxes that occur during sig- naling in excitable cells have essentially no effect on the concentrations of those ions. However, because the in- tracellular concentration of Ca 2H11001 is generally very low (~10 H110027 M), inward flow of Ca 2H11001 can significantly alter the cytosolic [Ca 2H11001 ]. The membrane potential of a cell at a given time is the result of the types and numbers of ion channels open at that instant. In most cells at rest, more K H11001 channels C in H5007 C out than Na H11001 , Cl H11002 , or Ca 2H11001 channels are open and thus the resting potential is closer to the E for K H11001 (H1100298 mV) than that for any other ion. When channels for Na H11001 , Ca 2H11001 , or Cl H11002 open, the membrane potential moves to- ward the E for that ion. The precisely timed opening and closing of ion channels and the resulting transient changes in membrane potential underlie the electrical signaling by which the nervous system stimulates the skeletal muscles to contract, the heart to beat, or se- cretory cells to release their contents. Moreover, many hormones exert their effects by altering the membrane potentials of their target cells. These mechanisms are not limited to complex animals; ion channels play im- portant roles in the responses of bacteria, protists, and plants to environmental signals. To illustrate the action of ion channels in cell-to-cell signaling, we describe the mechanisms by which a neu- ron passes a signal along its length and across a synapse to the next neuron (or to a myocyte) in a cellular cir- cuit, using acetylcholine as the neurotransmitter. The Nicotinic Acetylcholine Receptor Is a Ligand-Gated Ion Channel One of the best-understood examples of a ligand-gated receptor channel is the nicotinic acetylcholine re- ceptor (see Fig. 11–51). The receptor channel opens in response to the neurotransmitter acetylcholine (and to nicotine, hence the name). This receptor is found in the postsynaptic membrane of neurons at certain synapses and in muscle fibers (myocytes) at neuro- muscular junctions. Acetylcholine released by an excited neuron dif- fuses a few micrometers across the synaptic cleft or neu- romuscular junction to the postsynaptic neuron or my- ocyte, where it interacts with the acetylcholine receptor and triggers electrical excitation (depolarization) of the receiving cell. The acetylcholine receptor is an allosteric protein with two high-affinity binding sites for acetyl- choline, about 3.0 nm from the ion gate, on the two H9251 Acetylcholine (Ach) CH 3 CH 2 CH 2 ON CH 3 CH 3 O H11001 C CH 3 Chapter 12 Biosignaling426 TABLE 12–2 Ion Concentrations in Cells and Extracellular Fluids (mM) K H11001 Na H11001 Ca 2H11001 Cl H11002 Cell type In Out In Out In Out In Out Squid axon 400 20 50 440 H113490.4 10 40–150 560 Frog muscle 124 2.3 10.4 109 H110210.1 2.1 1.5 78 8885d_c12_426 2/20/04 1:16 PM Page 426 mac76 mac76:385_reb: subunits. The binding of acetylcholine causes a change from the closed to the open conformation. The process is positively cooperative: binding of acetylcholine to the first site increases the acetylcholine-binding affinity of the second site. When the presynaptic cell releases a brief pulse of acetylcholine, both sites on the postsy- naptic cell receptor are occupied briefly and the chan- nel opens (Fig. 12–4). Either Na H11001 or Ca 2H11001 can now pass, and the inward flux of these ions depolarizes the plasma membrane, initiating subsequent events that vary with the type of tissue. In a postsynaptic neuron, depolar- ization initiates an action potential (see below); at a neu- romuscular junction, depolarization of the muscle fiber triggers muscle contraction. Normally, the acetylcholine concentration in the synaptic cleft is quickly lowered by the enzyme acetyl- cholinesterase, present in the cleft. When acetylcholine levels remain high for more than a few milliseconds, the receptor is desensitized (Fig. 12–1c). The receptor channel is converted to a third conformation (Fig. 12–4c) in which the channel is closed and the acetyl- choline is very tightly bound. The slow release (in tens of milliseconds) of acetylcholine from its binding sites eventually allows the receptor to return to its resting state—closed and resensitized to acetylcholine levels. Voltage-Gated Ion Channels Produce Neuronal Action Potentials Signaling in the nervous system is accomplished by net- works of neurons, specialized cells that carry an elec- trical impulse (action potential) from one end of the cell (the cell body) through an elongated cytoplasmic ex- tension (the axon). The electrical signal triggers release of neurotransmitter molecules at the synapse, carrying the signal to the next cell in the circuit. Three types of voltage-gated ion channels are essential to this signaling mechanism. Along the entire length of the axon are voltage-gated Na H11545 channels (Fig. 12–5; see also Fig. 11–50), which are closed when the membrane is at rest (V m H11005H1100260 mV) but open briefly when the membrane is depolarized locally in response to acetyl- choline (or some other neurotransmitter). The depo- larization induced by the opening of Na H11001 channels causes voltage-gated K H11545 channels to open, and the resulting efflux of K H11001 repolarizes the membrane locally. A brief pulse of depolarization traverses the axon as lo- cal depolarization triggers the brief opening of neigh- boring Na H11001 channels, then K H11001 channels. After each opening of a Na H11001 channel, a short refractory period fol- lows during which that channel cannot open again, and thus a unidirectional wave of depolarization sweeps from the nerve cell body toward the end of the axon. The voltage sensitivity of ion channels is due to the pres- ence at critical positions in the channel protein of charged amino acid side chains that interact with the electric field across the membrane. Changes in trans- membrane potential produce subtle conformational changes in the channel protein (see Fig. 11–50). At the distal tip of the axon are voltage-gated Ca 2H11545 channels. When the wave of depolarization reaches these channels, they open, and Ca 2H11001 enters from the extracellular space. The rise in cytoplasmic [Ca 2H11001 ] then triggers release of acetylcholine by exocy- tosis into the synaptic cleft (step 3 in Fig. 12–5). Acetylcholine diffuses to the postsynaptic cell (another 12.2 Gated Ion Channels 427 ACh Continued excitation Na H11545 , Ca 2H11545 Acetylcholine binding sites Outside Inside (a) Resting (gate closed) (b) Excited (gate open) (c) Desensitized (gate closed) ACh FIGURE 12–4 Three states of the acetylcholine receptor. Brief ex- posure of (a) the resting (closed) ion channel to acetylcholine (ACh) produces (b) the excited (open) state. Longer exposure leads to (c) de- sensitization and channel closure. 8885d_c12_427 2/20/04 1:16 PM Page 427 mac76 mac76:385_reb: neuron or a myocyte), where it binds to acetylcholine receptors and triggers depolarization. Thus the message is passed to the next cell in the circuit. We see, then, that gated ion channels convey sig- nals in either of two ways: by changing the cytosolic con- centration of an ion (such as Ca 2H11001 ), which then serves as an intracellular second messenger (the hormone or neurotransmitter is the first messenger), or by chang- ing V m and affecting other membrane proteins that are sensitive to V m . The passage of an electrical signal through one neuron and on to the next illustrates both types of mechanism. Neurons Have Receptor Channels That Respond to Different Neurotransmitters Animal cells, especially those of the nervous system, con- tain a variety of ion channels gated by ligands, voltage, or both. The neurotransmitters 5-hydroxytryptamine (serotonin), glutamate, and glycine can all act through receptor channels that are structurally related to the acetylcholine receptor. Serotonin and glutamate trigger the opening of cation (K H11001 , Na H11001 , Ca 2H11001 ) channels, whereas glycine opens Cl H11002 -specific channels. Cation and anion channels are distinguished by subtle differences in the amino acid residues that line the hydrophilic channel. Cation channels have negatively charged Glu and Asp side chains at crucial positions. When a few of these acidic residues are experimentally replaced with basic residues, the cation channel is converted to an anion channel. Depending on which ion passes through a channel, the ligand (neurotransmitter) for that channel either de- polarizes or hyperpolarizes the target cell. A single neu- ron normally receives input from several (or many) other neurons, each releasing its own characteristic neurotransmitter with its characteristic depolarizing or hyperpolarizing effect. The target cell’s V m therefore reflects the integrated input (Fig. 12–1d) from multi- Chapter 12 Biosignaling428 Axon of presynaptic neuron Voltage- gated Na H11001 channel Voltage- gated K H11001 channel Action potential Secretory vesicles containing acetylcholine Synaptic cleft Acetylcholine receptor ion channels Action potential Cell body of postsynaptic neuron Volted- gated Ca 2H11001 channel Na H11001 Na H11001 Ca 2H11001 Na H11001 ,Ca 2H11001 Na H11001 Na H11001 K H11001 K H11001 K H11001 H11001 H11001 H11001H11002 H11001H11002 H11001 H11001 H11001 H11002 H11001 H11001 H11001 H11001 H11001 H11001 H11002 H11002 H11002 H11002H11001 H11001 H11001 H11002 H11001 H11001 H11001 H11001 1 2 3 5 4 FIGURE 12–5 Role of voltage-gated and ligand-gated ion channels in neural transmission. Initially, the plasma membrane of the pre- synaptic neuron is polarized (inside negative) through the action of the electrogenic Na H11001 K H11001 ATPase, which pumps 3 Na H11001 out for every 2 K H11001 pumped into the neuron (see Fig. 12–3). 1 A stimulus to this neu- ron causes an action potential to move along the axon (white arrow), away from the cell body. The opening of one voltage-gated Na H11001 chan- nel allows Na H11001 entry, and the resulting local depolarization causes the adjacent Na H11001 channel to open, and so on. The directionality of movement of the action potential is ensured by the brief refractory period that follows the opening of each voltage-gated Na H11001 channel. 2 When the wave of depolarization reaches the axon tip, voltage- gated Ca 2H11001 channels open, allowing Ca 2H11001 entry into the presynaptic neuron. 3 The resulting increase in internal [Ca 2H11001 ] triggers exocytic release of the neurotransmitter acetylcholine into the synaptic cleft. 4 Acetylcholine binds to a receptor on the postsynaptic neuron, caus- ing its ligand-gated ion channel to open. 5 Extracellular Na H11001 and Ca 2H11001 enter through this channel, depolarizing the postsynaptic cell. The electrical signal has thus passed to the cell body of the post- synaptic neuron and will move along its axon to a third neuron by this same sequence of events. Glutamate CH 2 COO H11002 H 3 N CH CH 2 COO H11002 H11001 Serotonin (5-hydroxytryptamine) HO N H CH 2 CH 2 NH 3 H11001 8885d_c12_428 2/20/04 1:16 PM Page 428 mac76 mac76:385_reb: ple neurons. The cell responds with an action potential only if the integrated input adds up to a net depolar- ization of sufficient size. The receptor channels for acetylcholine, glycine, glutamate, and H9253-aminobutyric acid (GABA) are gated by extracellular ligands. Intracellular second messen- gers—such as cAMP, cGMP (3H11032,5H11032-cyclic GMP, a close analog of cAMP), IP 3 (inositol 1,4,5-trisphosphate), Ca 2H11001 , and ATP—regulate ion channels of another class, which, as we shall see in Section 12.7, participate in the sensory transductions of vision, olfaction, and gustation. SUMMARY 12.2 Gated Ion Channels ■ Ion channels gated by ligands or membrane potential are central to signaling in neurons and other cells. ■ The acetylcholine receptor of neurons and myocytes is a ligand-gated ion channel. ■ The voltage-gated Na H11001 and K H11001 channels of neuronal membranes carry the action potential along the axon as a wave of depolarization (Na H11001 influx) followed by repolarization (K H11001 efflux). ■ The arrival of an action potential triggers neurotransmitter release from the presynaptic cell. The neurotransmitter (acetylcholine, for example) diffuses to the postsynaptic cell, binds to specific receptors in the plasma membrane, and triggers a change in V m . 12.3 Receptor Enzymes A fundamentally different mechanism of signal trans- duction is carried out by the receptor enzymes. These proteins have a ligand-binding domain on the extracel- lular surface of the plasma membrane and an enzyme active site on the cytosolic side, with the two domains connected by a single transmembrane segment. Com- monly, the receptor enzyme is a protein kinase that phosphorylates Tyr residues in specific target proteins; the insulin receptor is the prototype for this group. In plants, the protein kinase of receptors is specific for Ser or Thr residues. Other receptor enzymes synthesize the intracellular second messenger cGMP in response to ex- tracellular signals. The receptor for atrial natriuretic fac- tor is typical of this type. The Insulin Receptor Is a Tyrosine-Specific Protein Kinase Insulin regulates both metabolism and gene expression: the insulin signal passes from the plasma membrane re- ceptor to insulin-sensitive metabolic enzymes and to the nucleus, where it stimulates the transcription of specific genes. The active insulin receptor consists of two iden- tical H9251 chains protruding from the outer face of the plasma membrane and two transmembrane H9252 subunits with their carboxyl termini protruding into the cytosol (Fig. 12–6, step 1 ). The H9251 chains contain the insulin- binding domain, and the intracellular domains of the H9252 chains contain the protein kinase activity that transfers a phosphoryl group from ATP to the hydroxyl group of Tyr residues in specific target proteins. Signaling through the insulin receptor begins (step 1 ) when binding of insulin to the H9251 chains activates the Tyr ki- nase activity of the H9252 chains, and each H9251H9252 monomer phosphorylates three critical Tyr residues near the car- boxyl terminus of the H9252 chain of its partner in the dimer. This autophosphorylation opens up the active site so that the enzyme can phosphorylate Tyr residues of other target proteins (Fig. 12–7). One of these target proteins (Fig. 12–6, step 2 ) is insulin receptor substrate-1 (IRS-1). Once phosphory- lated on its Tyr residues, IRS-1 becomes the point of nu- cleation for a complex of proteins (step 3 ) that carry the message from the insulin receptor to end targets in the cytosol and nucleus, through a long series of inter- mediate proteins. First, a P –Tyr residue in IRS-1 is bound by the SH2 domain of the protein Grb2. (SH2 is an abbreviation of Src homology 2; the sequences of SH2 domains are similar to a domain in another protein Tyr kinase, Src (pronounced sark).) A number of sig- naling proteins contain SH2 domains, all of which bind P –Tyr residues in a protein partner. Grb2 also contains a second protein-binding domain, SH3, that binds to re- gions rich in Pro residues. Grb2 binds to a proline-rich region of Sos, recruiting Sos to the growing receptor complex. When bound to Grb2, Sos catalyzes the re- placement of bound GDP by GTP on Ras, one of a family of guanosine nucleotide–binding proteins (G proteins) that mediate a wide variety of signal transductions (Sec- tion 12.4). When GTP is bound, Ras can activate a pro- tein kinase, Raf-1 (step 4 ), the first of three protein kinases—Raf-1, MEK, and ERK—that form a cascade in which each kinase activates the next by phosphoryla- tion (step 5 ). The protein kinase ERK is activated by phosphorylation of both a Thr and a Tyr residue. When activated, it mediates some of the biological effects of insulin by entering the nucleus and phosphorylating pro- teins such as Elk1, which modulates the transcription of about 100 insulin-regulated genes (step 6 ). The proteins Raf-1, MEK, and ERK are members of three larger families, for which several nomenclatures are employed. ERK is a member of the MAPK family (mitogen-activated protein kinases; mitogens are sig- nals that act from outside the cell to induce mitosis and cell division). Soon after discovery of the first MAPK, that enzyme was found to be activated by another protein kinase, which came to be called MAP kinase kinase (MEK 12.3 Receptor Enzymes 429 8885d_c12_429 2/20/04 1:17 PM Page 429 mac76 mac76:385_reb: belongs to this family); and when a third kinase that ac- tivated MAP kinase kinase was discovered, it was given the slightly ludicrous family name MAP kinase kinase kinase (Raf-1 is a member of this family; Fig. 12–6). Slightly less cumbersome are the acronyms for these three families, MAPK, MAPKK, and MAPKKK. Kinases in the MAPK and MAPKKK families are specific for Ser or Thr residues, but MAPKKs (here, MEK) phosphory- late both a Ser and a Tyr residue in their substrate, a MAPK (here, ERK). Biochemists now recognize the insulin pathway as but one instance of a more general theme in which hor- mone signals, via pathways similar to that shown in Fig- ure 12–6, result in phosphorylation of target enzymes by protein kinases. The target of phosphorylation is of- ten another protein kinase, which then phosphorylates a third protein kinase, and so on. The result is a cas- cade of reactions that amplifies the initial signal by many orders of magnitude (see Fig. 12–1b). Cascades such as that shown in Figure 12–6 are called MAPK cascades. Chapter 12 Biosignaling430 New proteins DNA Insulin P P P P P PP P P P P P P Grb2 Sos Ras GTP GDP Raf-1 MEK ERKERK P ERK MEK P P IRS-1 IRS-1 Elk1SRF Elk1SRF Insulin receptor phosphorylates IRS-1 on its Tyr residues. 2 Activated Ras binds and activates Raf-1. 4 ERK moves into the nucleus and phosphorylates nuclear transcription factors such as Elk1, activating them. 6 Phosphorylated Elk1 joins SRF to stimulate the transcription and translation of a set of genes needed for cell division. 7 Cytosol Nucleus SH2 domain of Grb2 binds to P –Tyr of IRS-1. Sos binds to Grb2, then to Ras, causing GDP release and GTP binding to Ras. 3 Raf-1 phosphorylates MEK on two Ser residues, activating it. MEK phosphorylates ERK on a Thr and a Tyr residue, activating it. 5 Insulin receptor binds insulin and undergoes autophosphorylation on its carboxyl-terminal Tyr residues. 1 aa bb FIGURE 12–6 Regulation of gene expression by insulin. The insulin receptor consists of two H9251 chains on the outer face of the plasma mem- brane and two H9252 chains that traverse the membrane and protrude from the cytoplasmic face. Binding of insulin to the H9251 chains triggers a con- formational change that allows the autophosphorylation of Tyr residues in the carboxyl-terminal domain of the H9252 subunits. Autophosphoryla- tion further activates the Tyr kinase domain, which then catalyzes phos- phorylation of other target proteins. The signaling pathway by which insulin regulates the expression of specific genes consists of a cascade of protein kinases, each of which activates the next. The insulin re- ceptor is a Tyr-specific kinase; the other kinases (all shown in blue) phosphorylate Ser or Thr residues. MEK is a dual-specificity kinase, which phosphorylates both a Thr and a Tyr residue in ERK (extracel- lular regulated kinase); MEK is mitogen-activated, ERK-activating ki- nase; SRF is serum response factor. Abbreviations for other compo- nents are explained in the text. 8885d_c12_430 2/20/04 1:17 PM Page 430 mac76 mac76:385_reb: Inactive (unphosphorylated) Tyr kinase domain (a) Activation loop blocks substrate- binding site Tyr 1158 Tyr 1162 Tyr 1163 Asp 1132 Tyr 1158 Tyr 1162 Tyr 1163 Target protein in substrate- binding site Active (triply phosphorylated) Tyr kinase domain (b) Grb2 is not the only protein that associates with phosphorylated IRS-1. The enzyme phosphoinositide 3- kinase (PI-3K) binds IRS-1 through the former’s SH2 domain (Fig. 12–8). Thus activated, PI-3K converts the membrane lipid phosphatidylinositol 4,5-bisphosphate (see Fig. 10–15), also called PIP 2 , to phosphatidylinos- itol 3,4,5-trisphosphate (PIP 3 ). When bound to PIP 3 , protein kinase B (PKB) is phosphorylated and activated by yet another protein kinase, PDK1. The activated PKB then phosphorylates Ser or Thr residues on its target proteins, one of which is glycogen synthase kinase 3 (GSK3). In its active, nonphosphorylated form, GSK3 phosphorylates glycogen synthase, inactivating it and thereby contributing to the slowing of glycogen synthe- sis. (This mechanism is believed to be only part of the explanation for the effects of insulin on glycogen me- tabolism.) When phosphorylated by PKB, GSK3 is inac- tivated. By thus preventing inactivation of glycogen synthase in liver and muscle, the cascade of protein phosphorylations initiated by insulin stimulates glyco- gen synthesis (Fig. 12–8). In muscle, PKB triggers the movement of glucose transporters (GLUT4) from inter- nal vesicles to the plasma membrane, stimulating glu- cose uptake from the blood (Fig. 12–8; see also Box 11–2). PKB also functions in several other signaling pathways, including that triggered by H9004 9 -tetrahydro- cannabinol (THC), the active ingredient of marijuana 12.3 Receptor Enzymes 431 FIGURE 12–7 Activation of the insulin-receptor Tyr kinase by au- tophosphorylation. (a) In the inactive form of the Tyr kinase domain (PDB ID 1IRK), the activation loop (blue) sits in the active site, and none of the critical Tyr residues (black and red ball-and-stick struc- tures) are phosphorylated. This conformation is stabilized by hydro- gen bonding between Tyr 1162 and Asp 1132 . (b) When insulin binds to the H9251 chains of insulin receptors, the Tyr kinase of each H9252 subunit of the dimer phosphorylates three Tyr residues (Tyr 1158 , Tyr 1162 , and Tyr 1163 ) on the other H9252 subunit (shown here; PDB ID 1IR3). (Phos- phoryl groups are depicted here as an orange space-filling phospho- rus atom and red ball-and-stick oxygen atoms.) The effect of intro- ducing three highly charged P –Tyr residues is to force a 30 ? change in the position of the activation loop, away from the substrate-binding site, which becomes available to bind to and phosphorylate a target protein, shown here as a red arrow. O H CH 3 (CH 2 ) 3 H CH 3 CH 3 HO CH 3 H9004 9 -Tetrahydrocannabinol (THC) 8885d_c12_431 2/20/04 1:17 PM Page 431 mac76 mac76:385_reb: P P PKB PI-3K PIP 2 PIP 3 P GS (inactive) GSK3 (active) GSK3 (inactive) GS (active) IRS-1 IRS-1, phosphorylated by the insulin receptor, activates PI-3K by binding to its SH2 domain. PI-3K converts PIP 2 to PIP 3 . Glucose GLUT4 1 GSK3, inactivated by phosphorylation, cannot convert glycogen synthase (GS) to its inactive form by phosphorylation, so GS remains active. 3 Synthesis of glycogen from glucose is accelerated. 4 PKB stimulates movement of glucose transporter GLUT4 from internal membrane vesicles to the plasma membrane, increasing the uptake of glucose. 5 Glycogen 2 P P P P PKB bound to PIP 3 is phosphorylated by PDK1 (not shown). Thus activated, PKB phosphorylates GSK3 on a Ser residue, inactivating it. and hashish. THC activates the CB 1 receptor in the plasma membrane of neurons in the brain, triggering a signaling cascade that involves MAPKs. One conse- quence of CB 1 activation is the stimulation of appetite, one of the well-established effects of marijuana use. The normal ligands for the CB 1 receptor are endocannabi- noids such as anandamide, which serve to protect the brain from the toxicity of excessive neuronal activity— as in an epileptic seizure, for example. Hashish has for centuries been used in the treatment of epilepsy. As in all signaling pathways, there is a mecha- nism for terminating signaling through the PI- 3K–PKB pathway. A PIP 3 -specific phosphatase (PTEN in humans) removes the phosphate at the 3 position of PIP 3 to produce PIP 2 , which no longer serves as a bind- ing site for PKB, and the signaling chain is broken. In various types of advanced cancer, tumor cells often have a defect in the PTEN gene and thus have abnormally high levels of PIP 3 and of PKB activity. The result seems to be a continuing signal for cell division and thus tu- mor growth. ■ O N H OH Anandamide (arachidonylethanolamide, an endogenous cannabinoid) What spurred the evolution of such complicated regulatory machinery? This system allows one activated receptor to activate several IRS-1 molecules, amplifying the insulin signal, and it provides for the integration of signals from several receptors, each of which can phos- phorylate IRS-1. Furthermore, because IRS-1 can acti- vate any of several proteins that contain SH2 domains, a single receptor acting through IRS-1 can trigger two or more signaling pathways; insulin affects gene ex- pression through the Grb2-Sos-Ras-MAPK pathway and glycogen metabolism through the PI-3K–PKB pathway. The insulin receptor is the prototype for a number of receptor enzymes with a similar structure and recep- tor Tyr kinase activity. The receptors for epidermal growth factor and platelet-derived growth factor, for ex- ample, have structural and sequence similarities to the insulin receptor, and both have a protein Tyr kinase ac- tivity that phosphorylates IRS-1. Many of these recep- tors dimerize after binding ligand; the insulin receptor is already a dimer before insulin binds. The binding of adaptor proteins such as Grb2 to P –Tyr residues is a common mechanism for promoting protein-protein in- teractions, a subject to which we return in Section 12.5. In addition to the many receptors that act as protein Tyr kinases, a number of receptorlike plasma membrane proteins have protein Tyr phosphatase activity. Based on the structures of these proteins, we can surmise that their ligands are components of the extracellular matrix or the Chapter 12 Biosignaling432 FIGURE 12–8 Activation of glycogen synthase by insulin. Transmission of the signal is mediated by PI-3 kinase (PI-3K) and protein kinase B (PKB). 8885d_c12_432 2/20/04 1:17 PM Page 432 mac76 mac76:385_reb: surfaces of other cells. Although their signaling roles are not yet as well understood as those of the receptor Tyr kinases, they clearly have the potential to reverse the ac- tions of signals that stimulate these kinases. A variation on the basic theme of receptor Tyr ki- nases is seen in receptors that have no intrinsic protein kinase activity but, when occupied by their ligand, bind a soluble Tyr kinase. One example is the system that regulates the formation of erythrocytes in mammals. The cytokine (developmental signal) for this system is erythropoietin (EPO), a 165 amino acid protein pro- duced in the kidneys. When EPO binds to its plasma membrane receptor (Fig. 12–9), the receptor dimerizes and can now bind the soluble protein kinase JAK (Janus kinase). This binding activates JAK, which phosphory- lates several Tyr residues in the cytoplasmic domain of the EPO receptor. A family of transcription factors, col- lectively called STATs (signal transducers and activa- tors of transcription), are also targets of the JAK kinase activity. An SH2 domain in STAT5 binds P –Tyr residues in the EPO receptor, positioning it for this phosphory- lation by JAK. When STAT5 is phosphorylated in re- sponse to EPO, it forms dimers, exposing a signal for its transport into the nucleus. There, STAT5 causes the ex- pression (transcription) of specific genes essential for erythrocyte maturation. This JAK-STAT system oper- ates in a number of other signaling pathways, including that for the hormone leptin, described in detail in Chap- ter 23 (see Fig. 23–34). Activated JAK can also trigger, through Grb2, the MAPK cascade (Fig. 12–6), which leads to altered expression of specific genes. Src is another soluble protein Tyr kinase that asso- ciates with certain receptors when they bind their lig- ands. Src was the first protein found to have the char- acteristic P –Tyr-binding domain that was subsequently named the Src homology (SH2) domain. Yet another ex- ample of a receptor’s association with a soluble protein kinase is the Toll-like receptor (TLR4) system through which mammals detect the bacterial lipopolysaccharide (LPS), a potent toxin. We return to the Toll-like recep- tor system in Section 12.6, in the context of the evolu- tion of signaling proteins. Receptor Guanylyl Cyclases Generate the Second Messenger cGMP Guanylyl cyclases (Fig. 12–10) are another type of re- ceptor enzyme. When activated, a guanylyl cyclase pro- duces guanosine 3H11541,5H11541-cyclic monophosphate (cyclic GMP, cGMP) from GTP: 12.3 Receptor Enzymes 433 SH2 domain dimerization NLS Affects gene expression in nucleus MAPK cascade MAPK Erythropoietin EPO receptor P P P P P P P P JAKJAK P STAT P STAT P STAT STAT Grb2 FIGURE 12–9 The JAK-STAT transduction mechanism for the ery- thropoietin receptor. Binding of erythropoietin (EPO) causes dimer- ization of the EPO receptor, which allows the soluble Tyr kinase JAK to bind to the internal domain of the receptor and phosphorylate it on several Tyr residues. The STAT protein STAT5 contains an SH2 domain and binds to the P –Tyr residues on the receptor, bringing the recep- tor into proximity with JAK. Phosphorylation of STAT5 by JAK allows two STAT molecules to dimerize, each binding the other’s P –Tyr residue. Dimerization of STAT5 exposes a nuclear localization se- quence (NLS) that targets STAT5 for transport into the nucleus. In the nucleus, STAT causes the expression of genes controlled by EPO. A second signaling pathway is also triggered by autophosphorylation of JAK that is associated with EPO binding to its receptor. The adaptor protein Grb2 binds P –Tyr in JAK and triggers the MAPK cascade, as in the insulin system (see Fig. 12–6). H11002 O H P O H11002 O P O P H H H N O O PP i O O O O H11002 O H11002 OO H N N HN CH 2 5H11032 Guanosine 3H11032,5H11032-cyclic monophosphate (cGMP) H P O H11002 O OO H H H N O OH O O H N N HN CH NH 2 NH 2 2 GTP 3H11032 8885d_c12_433 2/20/04 1:17 PM Page 433 mac76 mac76:385_reb: Cyclic GMP is a second messenger that carries different messages in different tissues. In the kidney and intes- tine it triggers changes in ion transport and water re- tention; in cardiac muscle (a type of smooth muscle) it signals relaxation; in the brain it may be involved both in development and in adult brain function. Guanylyl cy- clase in the kidney is activated by the hormone atrial natriuretic factor (ANF), which is released by cells in the atrium of the heart when the heart is stretched by increased blood volume. Carried in the blood to the kidney, ANF activates guanylyl cyclase in cells of the collecting ducts (Fig. 12–10a). The resulting rise in [cGMP] triggers increased renal excretion of Na H11001 and, consequently, of water, driven by the change in osmotic pressure. Water loss reduces the blood volume, coun- tering the stimulus that initially led to ANF secretion. Vascular smooth muscle also has an ANF receptor— guanylyl cyclase; on binding to this receptor, ANF causes relaxation (vasodilation) of the blood vessel, which in- creases blood flow while decreasing blood pressure. A similar receptor guanylyl cyclase in the plasma membrane of intestinal epithelial cells is activated by an intestinal peptide, guanylin, which regulates Cl H11002 secre- tion in the intestine. This receptor is also the target of a heat-stable peptide endotoxin produced by Escherichia coli and other gram-negative bacteria. The elevation in [cGMP] caused by the endotoxin increases Cl H11002 secretion and consequently decreases reabsorption of water by the intestinal epithelium, producing diarrhea. A distinctly different type of guanylyl cyclase is a cytosolic protein with a tightly associated heme group (Fig. 12–10b), an enzyme activated by nitric oxide (NO). Nitric oxide is produced from arginine by Ca 2H11001 - dependent NO synthase, present in many mammalian tissues, and diffuses from its cell of origin into nearby cells. NO is sufficiently nonpolar to cross plasma mem- branes without a carrier. In the target cell, it binds to the heme group of guanylyl cyclase and activates cGMP production. In the heart, cGMP reduces the forcefulness of contractions by stimulating the ion pump(s) that ex- pel Ca 2H11001 from the cytosol. This NO-induced relaxation of cardiac muscle is the same response brought about by nitroglyc- erin tablets and other nitrovasodilators taken to relieve angina, the pain caused by contraction of a heart de- prived of O 2 because of blocked coronary arteries. Ni- tric oxide is unstable and its action is brief; within sec- onds of its formation, it undergoes oxidation to nitrite or nitrate. Nitrovasodilators produce long-lasting relax- ation of cardiac muscle because they break down over several hours, yielding a steady stream of NO. The value of nitroglycerin as a treatment for angina was discov- ered serendipitously in factories producing nitroglycerin as an explosive in the 1860s. Workers with angina re- ported that their condition was much improved during the work week but returned on weekends. The physi- cians treating these workers heard this story so often that they made the connection, and a drug was born. The effects of increased cGMP synthesis diminish after the stimulus ceases, because a specific phospho- diesterase (cGMP PDE) converts cGMP to the inactive 5H11032-GMP. Humans have several isoforms of cGMP PDE, with different tissue distributions. The isoform in the blood vessels of the penis is inhibited by the drug sildenafil (Viagra), which therefore causes cGMP levels to remain elevated once raised by an appropriate stim- ulus, accounting for the usefulness of this drug in the treatment of erectile dysfunction. ■ Most of the actions of cGMP in animals are believed to be mediated by cGMP-dependent protein kinase, also called protein kinase G or PKG, which, when ac- Citrulline NH 2 NH 3 O NH (CH 2 ) 3 CH COO H11002 H11001 NADP H11001 NADPH O NO synthase 2 Ca 2H11001 Arginine NH 2 NH 3 C NH (CH 2 ) 3 CH COO H11002 H11001 NH 2 H11001 C H11001 NO Chapter 12 Biosignaling434 (a) (b) Extracellular ligand- binding (receptor) domains Guanylin and endotoxin receptors Intracellular catalytic (cGMP- forming) domains Membrane-spanning guanylyl cyclases Soluble NO- activated guanylyl cyclase NH 3 H11001 H 3 N H11001 ANF receptor COO H5008 COO H5008 Heme Fe FIGURE 12–10 Two types (isozymes) of guanylyl cyclase that par- ticipate in signal transduction. (a) One isozyme exists in two similar membrane-spanning forms that are activated by their extracellular lig- ands: atrial natriuretic factor, ANF (receptors in cells of the renal col- lecting ducts and the smooth muscle of blood vessels), and guanylin (receptors in intestinal epithelial cells). The guanylin receptor is also the target of a type of bacterial endotoxin that triggers severe diarrhea. (b) The other isozyme is a soluble enzyme that is activated by intra- cellular nitric oxide (NO); this form is found in many tissues, includ- ing smooth muscle of the heart and blood vessels. 8885d_c12_434 2/20/04 1:19 PM Page 434 mac76 mac76:385_reb: tivated by cGMP, phosphorylates Ser and Thr residues in target proteins. The catalytic and regulatory domains of this enzyme are in a single polypeptide (M r ~80,000). Part of the regulatory domain fits snugly in the substrate- binding site. Binding of cGMP forces this part of the regulatory domain out of the binding site, activating the catalytic domain. Cyclic GMP has a second mode of action in the ver- tebrate eye: it causes ion-specific channels to open in the retinal rod and cone cells. We return to this role of cGMP in the discussion of vision in Section 12.7. SUMMARY 12.3 Receptor Enzymes ■ The insulin receptor is the prototype of receptor enzymes with Tyr kinase activity. When insulin binds to its receptor, each H9251H9252 monomer of the receptor phosphorylates the H9252 chain of its partner, activating the receptor’s Tyr kinase activity. The kinase catalyzes the phosphorylation of Tyr residues on other proteins such as IRS-1. ■ P –Tyr residues in IRS-1 serve as binding sites for proteins with SH2 domains. Some of these proteins, such as Grb2, have two or more protein-binding domains and can serve as adaptors that bring two proteins into proximity. ■ Further protein-protein interactions result in GTP binding to and activation of the Ras protein, which in turn activates a protein kinase cascade that ends with the phosphorylation of target proteins in the cytosol and nucleus. The result is specific metabolic changes and altered gene expression. ■ Several signals, including atrial natriuretic factor and the intestinal peptide guanylin, act through receptor enzymes with guanylyl cyclase activity. The cGMP produced acts as a second messenger, activating cGMP-dependent protein kinase (PKG). This enzyme alters metabolism by phosphorylating specific enzyme targets. ■ Nitric oxide (NO) is a short-lived messenger that acts by stimulating a soluble guanylyl cyclase, raising [cGMP] and stimulating PKG. 12.4 G Protein–Coupled Receptors and Second Messengers A third mechanism of signal transduction, distinct from gated ion channels and receptor enzymes, is defined by three essential components: a plasma membrane receptor with seven transmembrane helical segments, an enzyme in the plasma membrane that generates an intracellular second messenger, and a guanosine nucleotide–binding protein (G protein). The G pro- tein, stimulated by the activated receptor, exchanges bound GDP for GTP; the GTP-protein dissociates from the occupied receptor and binds to a nearby enzyme, altering its activity. The human genome encodes more than 1,000 members of this family of receptors, spe- cialized for transducing messages as diverse as light, smells, tastes, and hormones. The H9252-adrenergic recep- tor, which mediates the effects of epinephrine on many tissues, is the prototype for this type of transducing system. The H9252-Adrenergic Receptor System Acts through the Second Messenger cAMP Epinephrine action begins when the hormone binds to a protein receptor in the plasma membrane of a hormone- sensitive cell. Adrenergic receptors (“adrenergic” re- flects the alternative name for epinephrine, adrenaline) are of four general types, H9251 1 , H9251 2 , H9252 1 , and H9252 2 , defined by subtle differences in their affinities and responses to a group of agonists and antagonists. Agonists are struc- tural analogs that bind to a receptor and mimic the ef- fects of its natural ligand; antagonists are analogs that bind without triggering the normal effect and thereby block the effects of agonists. In some cases, the affinity of the synthetic agonist or antagonist for the receptor is greater than that of the natural agonist (Fig. 12–11). The four types of adrenergic receptors are found in dif- ferent target tissues and mediate different responses to epinephrine. Here we focus on the H9252-adrenergic re- ceptors of muscle, liver, and adipose tissue. These receptors mediate changes in fuel metabolism, as de- scribed in Chapter 23, including the increased break- down of glycogen and fat. Adrenergic receptors of the H9252 1 and H9252 2 subtypes act through the same mechanism, so in our discussion, “H9252-adrenergic” applies to both types. The H9252-adrenergic receptor is an integral protein with seven hydrophobic regions of 20 to 28 amino acid residues that “snake” back and forth across the plasma membrane seven times. This protein is a mem- ber of a very large family of receptors, all with seven transmembrane helices, that are commonly called ser- pentine receptors, G protein–coupled receptors (GPCR), or 7 transmembrane segment (7tm) re- ceptors. The binding of epinephrine to a site on the 12.4 G Protein–Coupled Receptors and Second Messengers 435 S O O O N N Sildenafil (Viagra) N N N HN O 8885d_c12_435 2/20/04 1:19 PM Page 435 mac76 mac76:385_reb: receptor deep within the membrane (Fig. 12–12, step 1 ) promotes a conformational change in the receptor’s intracellular domain that affects its interaction with the second protein in the signal-transduction pathway, a heterotrimeric GTP-binding stimulatory G protein, or G S , on the cytosolic side of the plasma membrane. Alfred G. Gilman and Martin Rodbell discovered that when GTP is bound to G s , G s stimulates the production of cAMP by adenylyl cyclase (see below) in the plasma membrane. The function of G s as a molecular switch re- sembles that of another class of G proteins typified by Ras, discussed in Section 12.3 in the context of the in- sulin receptor. Structurally, G s and Ras are quite distinct; G proteins of the Ras type are monomers (M r ~20,000), whereas the G proteins that interact with serpentine Chapter 12 Biosignaling436 FIGURE 12–11 Epinephrine and its synthetic analogs. Epinephrine, also called adrenaline, is released from the adrenal gland and regulates energy-yielding metabolism in muscle, liver, and adipose tissue. It also serves as a neurotransmitter in adrenergic neurons. Its affinity for its re- ceptor is expressed as a dissociation constant for the receptor-ligand complex. Isoproterenol and propranolol are synthetic analogs, one an agonist with an affinity for the receptor that is higher than that of epi- nephrine, and the other an antagonist with extremely high affinity. HO OH CH CH 2 CH 3 NH HO HO OH CH CH 2 CH 3 CH 3 NH CH OH CHCH 2 CH 3 CH 3 NH CHCH 2 HO Epinephrine Isoproterenol (agonist) Propranolol (antagonist) k d (H9262M) 5 0.4 0.0046O 2 The occupied receptor causes replacement of the GDP bound to G s by GTP, activating G s . 1 Epinephrine binds to its specific receptor. ATP cAMP 5H11032-AMP cyclic nucleotide phosphodiesterase GTP GDP 5 cAMP activates PKA. 6 Phosphorylation of cellular proteins by PKA causes the cellular response to epinephrine. 7 cAMP is degraded, reversing the activation of PKA. E NH 3 H11002 OOC H11001 Rec H9253 H9251 H9252 G s G s GDP GTP AC H9251 4 Adenylyl cyclase catalyzes the formation of cAMP. 3 G s ( subunit) moves to adenylyl cyclase and activates it. H9251 FIGURE 12–12 Transduction of the epinephrine signal: the H9252- adrenergic pathway. The seven steps of the mechanism that couples binding of epinephrine (E) to its receptor (Rec) with activation of adenyl- yl cyclase (AC) are discussed further in the text. The same adenylyl cyclase molecule in the plasma membrane may be regulated by a stimulatory G protein (G s ), as shown, or an inhibitory G protein (G i , not shown). G s and G i are under the influence of different hormones. Hormones that induce GTP binding to G i cause inhibition of adenyl- yl cyclase, resulting in lower cellular [cAMP]. Alfred G. Gilman Martin Rodbell, 1925–1998 8885d_c12_436 2/20/04 1:19 PM Page 436 mac76 mac76:385_reb: G s GTP AC H9251 receptors are trimers of three different subunits, H9251 (M r 43,000), H9252 (M r 37,000), and H9253 (M r 7,500 to 10,000). When the nucleotide-binding site of G s (on the H9251 subunit) is occupied by GTP, G s is active and can acti- vate adenylyl cyclase (AC in Fig. 12–12); with GDP bound to the site, G s is inactive. Binding of epinephrine enables the receptor to catalyze displacement of bound GDP by GTP, converting G s to its active form (step 2 ). As this occurs, the H9252 and H9253 subunits of G s dissoci- ate from the H9251 subunit, and G sH9251 , with its bound GTP, moves in the plane of the membrane from the receptor to a nearby molecule of adenylyl cyclase (step 3 ). The G sH9251 is held to the membrane by a covalently attached palmitoyl group (see Fig. 11–14). Adenylyl cyclase (Fig. 12–13) is an integral protein of the plasma membrane, with its active site on the cyto- solic face. It catalyzes the synthesis of cAMP from ATP: The association of active G sH9251 with adenylyl cyclase stim- ulates the cyclase to catalyze cAMP synthesis (Fig. 12–12, step 4 ), raising the cytosolic [cAMP]. This stim- ulation by G sH9251 is self-limiting; G sH9251 is a GTPase that turns itself off by converting its bound GTP to GDP (Fig. 12–14). The now inactive G sH9251 dissociates from adenylyl cyclase, rendering the cyclase inactive. After G sH9251 reas- sociates with the H9252 and H9253 subunits (G sH9252H9253 ), G s is again available to interact with a hormone-bound receptor. Trimeric G Proteins: Molecular On/Off Switches H11002 O H P O H11002 O P O P H H H N O O PP i O O O H11002 O H11002 OO H N N N CH 2 5H11032 Adenosine 3H11032,5H11032-cyclic monophosphate (cAMP) H P O H11002 O OO H H H N O OH O H N N N CH NH 2 NH 2 2 ATP 3H11032 12.4 G Protein–Coupled Receptors and Second Messengers 437 FIGURE 12–13 Interaction of G sH9251 with adenylyl cyclase. (PDB ID 1AZS) The soluble catalytic core of the adenylyl cyclase (AC, blue), severed from its membrane anchor, was cocrystallized with G sH9251 (green) to give this crystal structure. The plant terpene forskolin (yellow) is a drug that strongly stimulates the enzyme, and GTP (red) bound to G sH9251 triggers interaction of G sH9251 with adenylyl cyclase. FIGURE 12–14 Self-inactivation of G s . The steps are further described in the text. The protein’s intrinsic GTPase activity, in many cases stim- ulated by RGS proteins (regulators of G protein signaling), determines how quickly bound GTP is hydrolyzed to GDP and thus how long the G protein remains active. H9253H9252 H9251 H9253 H9252 GDP GTP G s G s GTP GDP P i GDP G s 1 G s with GDP bound is turned off; it cannot activate adenylyl cyclase. 2 Contact of G s with hormone-receptor complex causes dis- placement of bound GDP by GTP. 3 G s with GTP bound dissociates into a and bg subunits. G sa -GTP is turned on; it can activate adenylyl cyclase. 4 GTP bound to G sa is hydrolyzed by the protein’s intrinsic GTPase; G sa thereby turns itself off. The inactive a subunit reassociates with the bg subunit. H9251 H9251 8885d_c12_437 2/20/04 1:19 PM Page 437 mac76 mac76:385_reb: One downstream effect of epinephrine is to activate glycogen phosphorylase b. This conversion is promoted by the enzyme phosphorylase b kinase, which catalyzes the phosphorylation of two specific Ser residues in phos- phorylase b, converting it to phosphorylase a (see Fig. 6–31). Cyclic AMP does not affect phosphorylase b ki- nase directly. Rather, cAMP-dependent protein ki- nase, also called protein kinase A or PKA, which is allosterically activated by cAMP (Fig. 12–12, step 5 ), catalyzes the phosphorylation of inactive phosphorylase b kinase to yield the active form. The inactive form of PKA contains two catalytic sub- units (C) and two regulatory subunits (R) (Fig. 12–15a), which are similar in sequence to the catalytic and reg- ulatory domains of PKG (cGMP-dependent protein ki- nase). The tetrameric R 2 C 2 complex is catalytically in- active, because an autoinhibitory domain of each R subunit occupies the substrate-binding site of each C subunit. When cAMP binds to two sites on each R sub- unit, the R subunits undergo a conformational change and the R 2 C 2 complex dissociates to yield two free, catalytically active C subunits. This same basic mecha- nism—displacement of an autoinhibitory domain— mediates the allosteric activation of many types of pro- tein kinases by their second messengers (as in Figs 12–7 and 12–23, for example). As indicated in Figure 12–12 (step 6 ), PKA regu- lates a number of enzymes (Table 12–3). Although the proteins regulated by cAMP-dependent phosphorylation have diverse functions, they share a region of sequence similarity around the Ser or Thr residue that undergoes phosphorylation, a sequence that marks them for regu- lation by PKA. The catalytic site of PKA (Fig. 12–15b) interacts with several residues near the Thr or Ser residue in the target protein, and these interactions de- fine the substrate specificity. Comparison of the se- quences of a number of protein substrates for PKA has yielded the consensus sequence—the specific neigh- boring residues needed to mark a Ser or Thr residue for phosphorylation (see Table 12–3). Signal transduction by adenylyl cyclase entails sev- eral steps that amplify the original hormone signal (Fig. Chapter 12 Biosignaling438 (b) R C R RR C C C 4 cAMP4 cAMP (a) Inactive PKA Regulatory subunits: empty cAMP sites Regulatory subunits: autoinhibitory domains buried Catalytic subunits: substrate-binding sites blocked by autoinhibitory domains of R subunits Active PKA Catalytic subunits: open substrate- binding sites + FIGURE 12–15 Activation of cAMP-dependent protein kinase, PKA. (a) A schematic representation of the inactive R 2 C 2 tetramer, in which the autoinhibitory domain of a regulatory (R) subunit occupies the substrate-binding site, inhibiting the activity of the catalytic (C) sub- unit. Cyclic AMP activates PKA by causing dissociation of the C sub- units from the inhibitory R subunits. Activated PKA can phosphorylate a variety of protein substrates (Table 12–3) that contain the PKA con- sensus sequence (X–Arg–(Arg/Lys)–X–(Ser/Thr)–B, where X is any residue and B is any hydrophobic residue), including phosphorylase b kinase. (b) The substrate-binding region of a catalytic subunit re- vealed by x-ray crystallography (derived from PDB ID 1JBP). Enzyme side chains known to be critical in substrate binding and specificity are in blue. The peptide substrate (red) lies in a groove in the enzyme surface, with its Ser residue (yellow) positioned in the catalytic site. In the inactive R 2 C 2 tetramer, the autoinhibitory domain of R lies in this groove, blocking access to the substrate. 8885d_c12_438 2/20/04 1:20 PM Page 438 mac76 mac76:385_reb: CH 2 N O OH P O H11002 H11002 O H N N N H HH NH 2 3H11032 5H11032 O O OH Cyclic AMP Adenosine 5H11032-monophosphate (AMP) H 2 O CH 2 N O OHP O H11002 H N N N H HH 3H11032 5H11032 O OO NH 2 12–16). First, the binding of one hormone molecule to one receptor catalytically activates several G s molecules. Next, by activating a molecule of adenylyl cyclase, each active G sH9251 molecule stimulates the catalytic synthesis of many molecules of cAMP. The second messenger cAMP now activates PKA, each molecule of which catalyzes the phosphorylation of many molecules of the target protein—phosphorylase b kinase in Figure 12–16. This The intracellular signal therefore persists only as long as the hormone receptor remains occupied by epineph- rine. Methyl xanthines such as caffeine and theophylline (a component of tea) inhibit the phosphodiesterase, in- creasing the half-life of cAMP and thereby potentiating agents that act by stimulating adenylyl cyclase. The H9252-Adrenergic Receptor Is Desensitized by Phosphorylation As noted earlier, signal-transducing systems undergo desensitization when the signal persists. Desensitization of the H9252-adrenergic receptor is mediated by a protein kinase that phosphorylates the receptor on the intra- cellular domain that normally interacts with G s (Fig. 12–17). When the receptor is occupied by epinephrine, 12.4 G Protein–Coupled Receptors and Second Messengers 439 x molecules Epinephrine-receptor complex x molecules Active PKAInactive PKA 10x molecules Active phosphorylase b kinase 100x molecules Inactive phosphorylase b kinase Active glycogen phosphorylase a 1,000x molecules Inactive glycogen phosphorylase b Glycogen Glucose 1-phosphate Hepatocyte ATP Cyclic AMP adenylyl cyclase 20x molecules Blood glucose 10,000x molecules Glucose 10,000x molecules Epinephrine G Sa many steps FIGURE 12–16 Epinephrine cascade. Epinephrine triggers a series of reactions in hepatocytes in which catalysts activate catalysts, resulting in great amplification of the signal. Binding of a small number of mol- ecules of epinephrine to specific H9252-adrenergic receptors on the cell surface activates adenylyl cyclase. To illustrate amplification, we show 20 molecules of cAMP produced by each molecule of adenylyl cyclase, the 20 cAMP molecules activating 10 molecules of PKA, each PKA molecule activating 10 molecules of the next enzyme (a total of 100), and so forth. These amplifications are probably gross underestimates. kinase activates glycogen phosphorylase b, which leads to the rapid mobilization of glucose from glycogen. The net effect of the cascade is amplification of the hormonal signal by several orders of magnitude, which accounts for the very low concentration of epinephrine (or any other hormone) required for hormone activity. Cyclic AMP, the intracellular second messenger in this system, is short-lived. It is quickly degraded by cyclic nucleotide phosphodiesterase to 5H11032-AMP (Fig. 12–12, step 7 ), which is not active as a second messenger: 8885d_c12_439 2/20/04 1:20 PM Page 439 mac76 mac76:385_reb: Chapter 12 Biosignaling440 TABLE 12–3 Some Enzymes and Other Proteins Regulated by cAMP-Dependent Phosphorylation (by PKA) Enzyme/protein Sequence phosphorylated * Pathway/process regulated Glycogen synthase RASCTSSS Glycogen synthesis Phosphorylase b kinase H9251 subunit VEFRRLSI Glycogen breakdown H9252 subunit RTKRSGSV Pyruvate kinase (rat liver) GVLRRASVAZL Glycolysis Pyruvate dehydrogenase complex (type L) GYLRRASV Pyruvate to acetyl-CoA Hormone-sensitive lipase PMRRSV Triacylglycerol mobilization and fatty acid oxidation Phosphofructokinase-2/fructose 2,6-bisphosphatase LQRRRGSSIPQ Glycolysis/gluconeogenesis Tyrosine hydroxylase FIGRRQSL Synthesis of L-DOPA, dopamine, norepinephrine, and epinephrine Histone H1 AKRKASGPPVS DNA condensation Histone H2B KKAKASRKESYSVYVYK DNA condensation Cardiac phospholamban (cardiac pump regulator) AIRRAST Intracellular [Ca 2H11001 ] Protein phosphatase-1 inhibitor-1 IRRRRPTP Protein dephosphorylation PKA consensus sequence ? XR(R/K)X(S/T)B Many *The phosphorylated S or T residue is shown in red. All residues are given as their one-letter abbreviations (see Table 3–1). ? X is any amino acid; B is any hydrophobic amino acid. } P P Binding of epinephrine (E) to b-adrenergic receptor triggers dissociation of G sbg from G sa (not shown). 1 G sbg recruits bARK to the membrane, where it phosphorylates Ser residues at the carboxyl terminus of the receptor. 2 barr binds to the phosphorylated carboxyl-terminal domain of the receptor. 3 EE E bARK b arr G sbg G sbg P P P P P P In endocytic vesicle, arrestin dissociates; receptor is dephosphorylated and returned to cell surface. 5 Receptor-arrestin complex enters the cell by endocytosis. 4 FIGURE 12–17 Desensitization of the H9252-adrenergic receptor in the continued presence of epinephrine. This process is mediated by two proteins: H9252-adrenergic protein kinase (H9252ARK) and H9252-arrestin (H9252arr; arrestin 2). 8885d_c12_440 2/20/04 1:20 PM Page 440 mac76 mac76:385_reb: H9252-adrenergic receptor kinase (H9252ARK) phosphory- lates Ser residues near the carboxyl terminus of the re- ceptor. Normally located in the cytosol, H9252ARK is drawn to the plasma membrane by its association with th G sH9252H9253 subunits and is thus positioned to phosphorylate the re- ceptor. The phosphorylation creates a binding site for the protein H9252-arrestin (H9252arr), also called arrestin 2, and binding of H9252-arrestin effectively prevents interac- tion between the receptor and the G protein. The bind- ing of H9252-arrestin also facilitates receptor sequestration, the removal of receptors from the plasma membrane by endocytosis into small intracellular vesicles. Receptors in the endocytic vesicles are dephosphorylated, then re- turned to the plasma membrane, completing the circuit and resensitizing the system to epinephrine. H9252-Adrenergic receptor kinase is a member of a family of G protein– coupled receptor kinases (GRKs), all of which phos- phorylate serpentine receptors on their carboxyl-terminal cytosolic domains and play roles similar to that of H9252ARK in desensitization and resensitization of their receptors. At least five different GRKs and four different arrestins are encoded in the human genome; each GRK is capable of desensitizing a subset of the serpentine receptors, and each arrestin can interact with many different types of phosphorylated receptors. While preventing the signal from a serpentine re- ceptor from reaching its associated G protein, arrestins can also initiate a second signaling cascade, by acting as scaffold proteins that bring together several pro- tein kinases that function in a cascade. For example, the H9252-arrestin associated with the serpentine receptor for angiotensin, a potent regulator of blood pressure, binds the three protein kinases Raf-1, MEK1, and ERK (Fig. 12–18), serving as a scaffold that facilitates any signal- ing process, such as insulin signaling (Fig. 12–6), that requires these three protein kinases to interact. This is one of many known examples of cross-talk between sys- tems triggered by different ligands (angiotensin and in- sulin, in this case). Cyclic AMP Acts as a Second Messenger for a Number of Regulatory Molecules Epinephrine is only one of many hormones, growth fac- tors, and other regulatory molecules that act by chang- ing the intracellular [cAMP] and thus the activity of PKA (Table 12–4). For example, glucagon binds to its re- ceptors in the plasma membrane of adipocytes, activat- ing (via a G s protein) adenylyl cyclase. PKA, stimulated by the resulting rise in [cAMP], phosphorylates and ac- tivates two proteins critical to the conversion of stored fat to fatty acids (perilipin and hormone-sensitive tri- acylglycerol lipase; see Fig. 17–3), leading to the mobi- lization of fatty acids. Similarly, the peptide hormone ACTH (adrenocorticotropic hormone, also called corti- cotropin), produced by the anterior pituitary, binds to specific receptors in the adrenal cortex, activating adenylyl cyclase and raising the intracellular [cAMP]. PKA then phosphorylates and activates several of the enzymes required for the synthesis of cortisol and other steroid hormones. The catalytic subunit of PKA can also move into the nucleus, where it phosphorylates a pro- tein that alters the expression of specific genes. Some hormones act by inhibiting adenylyl cyclase, lowering cAMP levels, and suppressing protein phos- phorylation. For example, the binding of somatostatin to its receptor leads to activation of an inhibitory G pro- tein, or G i , structurally homologous to G s , that inhibits adenylyl cyclase and lowers [cAMP]. Somatostatin there- fore counterbalances the effects of glucagon. In adipose tissue, prostaglandin E 1 (PGE 1 ; see Fig. 10–18b) inhibits adenylyl cyclase, thus lowering [cAMP] and slowing the 12.4 G Protein–Coupled Receptors and Second Messengers 441 FIGURE 12–18 H9252-Arrestin uncouples the serpentine receptor from its G protein and brings together the three enzymes of the MAPK cas- cade. The effect is that one stimulus triggers two distinct response path- ways: the path activated by the G protein and the MAPK cascade. MEK1 MAPKK ERK1/2 MAPK E barr P P Raf-1 MAPKKK Corticotropin (ACTH) Corticotropin-releasing hormone (CRH) Dopamine [D 1 ,D 2 ] * Epinephrine (H9252-adrenergic) Follicle-stimulating hormone (FSH) Glucagon Histamine [H 2 ] * Luteinizing hormone (LH) Melanocyte-stimulating hormone (MSH) Odorants (many) Parathyroid hormone Prostaglandins E 1 ,E 2 (PGE 1 , PGE 2 ) Serotonin [5-HT-1a, 5-HT-2] * Somatostatin Tastants (sweet, bitter) Thyroid-stimulating hormone (TSH) Some Signals That Use cAMP as Second Messenger TABLE 12–4 * Receptor subtypes in square brackets. Subtypes may have different transduction mechanisms. For example, serotonin is detected in some tissues by receptor subtypes 5-HT-1a and 5-HT-1b, which act through adenylyl cyclase and cAMP, and in other tissues by receptor subtype 5-HT-1c, acting through the phospholipase C–IP 3 mechanism (see Table 12–5). 8885d_c12_441 2/20/04 1:20 PM Page 441 mac76 mac76:385_reb: mobilization of lipid reserves triggered by epinephrine and glucagon. In certain other tissues PGE 1 stimulates cAMP synthesis, because its receptors are coupled to adenylyl cyclase through a stimulatory G protein, G s . In tissues with H9251 2 -adrenergic receptors, epinephrine low- ers [cAMP], because the H9251 2 receptors are coupled to adenylyl cyclase through an inhibitory G protein, G i . In short, an extracellular signal such as epinephrine or PGE 1 can have quite different effects on different tis- sues or cell types, depending on three factors: the type of receptor in each tissue, the type of G protein (G s or G i ) with which the receptor is coupled, and the set of PKA target enzymes in the cells. A fourth factor that explains how so many signals can be mediated by a single second messenger (cAMP) is the confinement of the signaling process to a specific region of the cell by scaffold proteins. AKAPs (Akinase anchoring proteins) are bivalent; one part binds to the R subunit of PKA, and another to a specific structure within the cell, confining the PKA to the vicinity of that structure. For example, specific AKAPs bind PKA to microtubules, actin filaments, Ca 2H11001 channels, mito- chondria, and the nucleus. Different types of cells have different AKAPs, so cAMP might stimulate phosphory- lation of mitochondrial proteins in one cell and phos- phorylation of actin filaments in another. In studies of the intracellular localization of biochemical changes, biochemistry meets cell biology, and techniques that cross this boundary become invaluable (Box 12–2). Two Second Messengers Are Derived from Phosphatidylinositols A second class of serpentine receptors are coupled through a G protein to a plasma membrane phospholi- pase C (PLC) that is specific for the plasma membrane lipid phosphatidylinositol 4,5-bisphosphate (see Fig. 10–15). This hormone-sensitive enzyme catalyzes the formation of two potent second messengers: diacyl- glycerol and inositol 1,4,5-trisphosphate, or IP 3 (not to be confused with PIP 3 , p. 431). When a hormone of this class (Table 12–5) binds its specific receptor in the plasma membrane (Fig. 12–19, step 1 ), the receptor-hormone complex catalyzes GTP-GDP exchange on an associated G protein, G q H P O O O H H HH H HH 4 32 1 6 Inositol 1,4,5-trisphosphate (IP 3 ) O O H11002 H11002 O H11002 O P O H11002 OPO O H11002 O OH O 5 O H11002 (step 2 ), activating it exactly as the H9252-adrenergic re- ceptor activates G s (Fig. 12–12). The activated G q in turn activates a specific membrane-bound PLC (step 3 ), which catalyzes the production of the two second mes- sengers diacylglycerol and IP 3 by hydrolysis of phos- phatidylinositol 4,5-bisphosphate in the plasma mem- brane (step 4 ). Inositol trisphosphate, a water-soluble compound, diffuses from the plasma membrane to the endoplasmic reticulum, where it binds to specific IP 3 receptors and causes Ca 2H11001 channels within the ER to open. Seques- tered Ca 2H11001 is thus released into the cytosol (step 5 ), and the cytosolic [Ca 2H11001 ] rises sharply to about 10 H110026 M. One effect of elevated [Ca 2H11001 ] is the activation of pro- tein kinase C (PKC). Diacylglycerol cooperates with Ca 2H11001 in activating PKC, thus also acting as a second messenger (step 6 ). PKC phosphorylates Ser or Thr residues of specific target proteins, changing their cat- alytic activities (step 7 ). There are a number of isozymes of PKC, each with a characteristic tissue dis- tribution, target protein specificity, and role. The action of a group of compounds known as tumor promoters is attributable to their effects on PKC. The best understood of these are the phorbol esters, synthetic compounds that are potent activators of PKC. They apparently mimic cellular diacylglycerol as second messengers, but unlike naturally occurring di- acylglycerols they are not rapidly metabolized. By con- tinuously activating PKC, these synthetic tumor pro- moters interfere with the normal regulation of cell growth and division (discussed in Section 12.10). ■ Calcium Is a Second Messenger in Many Signal Transductions In many cells that respond to extracellular signals, Ca 2H11001 serves as a second messenger that triggers intracellu- lar responses, such as exocytosis in neurons and en- docrine cells, contraction in muscle, and cytoskeletal rearrangement during amoeboid movement. Normally, cytosolic [Ca 2H11001 ] is kept very low (H1102110 H110027 M) by the ac- tion of Ca 2H11001 pumps in the ER, mitochondria, and plasma membrane. Hormonal, neural, or other stimuli cause either an influx of Ca 2H11001 into the cell through specific CH 2 CO HO OH CH 3 CH 3 O CH 3 (CH 2 ) 12 CO O HO Myristoylphorbol acetate (a phorbol ester) CH 3 O CH 3 CH 3 Chapter 12 Biosignaling442 8885d_c12_442 2/20/04 1:21 PM Page 442 mac76 mac76:385_reb: 12.4 G Protein–Coupled Receptors and Second Messengers 443 Phospholipase C (PLC) Hormone (H) binds to a specific receptor. The occupied receptor causes GDP-GTP exchange on G q . G q , with bound GTP, moves to PLC and activates it. Active PLC cleaves phosphatidyl- inositol 4,5-bisphosphate to inositol trisphosphate (IP 3 ) and diacylglycerol. Endoplasmic reticulum IP 3 binds to a specific receptor on the endoplasmic reticulum, releasing sequestered Ca 2H11001 . Diacylglycerol and Ca 2H11001 activate protein kinase C at the surface of the plasma membrane. Protein kinase C IP 3 Ca 2H11001 Ca 2H11001 channel Diacylglycerol Plasma membrane Extracellular space Cytosol Phosphorylation of cellular proteins by protein kinase C produces some of the cellular responses to the hormone. Receptor GTP G q G q GTP H GDP GDP 1 2 3 4 5 6 7 FIGURE 12–19 Hormone-activated phospholipase C and IP 3 . Two in- tracellular second messengers are produced in the hormone-sensitive phosphatidylinositol system: inositol 1,4,5-trisphosphate (IP 3 ) and diacylglycerol. Both contribute to the activation of protein kinase C. By raising cytosolic [Ca 2H11001 ], IP 3 also activates other Ca 2H11001 -dependent enzymes; thus Ca 2H11001 also acts as a second messenger. Acetylcholine [muscarinic M 1 ] H9251 1 -Adrenergic agonists Angiogenin Angiotensin II ATP [P 2x and P 2y ] * Auxin Gastrin-releasing peptide Glutamate Gonadotropin-releasing hormone (GRH) Histamine [H 1 ] * Light (Drosophila) Oxytocin Platelet-derived growth factor (PDGF) Serotonin [5-HT-1c] * Thyrotropin-releasing hormone (TRH) Vasopressin TABLE 12–5 Some Signals That Act through Phospholipase C and IP 3 * Receptor subtypes are in square brackets; see footnote to Table 12–4. 8885d_c12_443 2/20/04 1:21 PM Page 443 mac76 mac76:385_reb: Ca 2H11001 channels in the plasma membrane or the release of sequestered Ca 2H11001 from the ER or mitochondria, in either case raising the cytosolic [Ca 2H11001 ] and triggering a cellular response. Very commonly, [Ca 2H11001 ] does not simply rise and then decrease, but rather oscillates with a period of a few seconds (Fig. 12–20), even when the extracellular concentration of hormone remains constant. The mech- anism underlying [Ca 2H11001 ] oscillations presumably entails feedback regulation by Ca 2H11001 of either the phospholipase Chapter 12 Biosignaling444 Cytosolic [Ca 2 H11001 ] (n M ) 0 (b) 100 100 200 300 400 600 200 300 400 500 0 0.5 1.0 [Ca 2H11001 ] (mM) (a) FIGURE 12–20 Triggering of oscillations in intracellular [Ca 2H11545 ] by extracellular signals. (a) A dye (fura) that undergoes fluorescence changes when it binds Ca 2H11001 is allowed to diffuse into cells, and its instantaneous light output is measured by fluorescence microscopy. Fluorescence intensity is represented by color; the color scale relates intensity of color to [Ca 2H11001 ], allowing determination of the absolute [Ca 2H11001 ]. In this case, thymocytes (cells of the thymus) have been stim- ulated with extracellular ATP, which raises their internal [Ca 2H11001 ]. The cells are heterogeneous in their responses; some have high intracel- lular [Ca 2H11001 ] (red), others much lower (blue). (b) When such a probe is used to measure [Ca 2H11001 ] in a single hepatocyte, we observe that the agonist norepinephrine (added at the arrow) causes oscillations of [Ca 2H11001 ] from 200 to 500 nM. Similar oscillations are induced in other cell types by other extracellular signals. Adenylyl cyclase (brain) Ca 2H11001 /calmodulin-dependent protein kinases (CaM kinases I to IV) Ca 2H11001 -dependent Na H11001 channel (Paramecium) Ca 2H11001 -release channel of sarcoplasmic reticulum Calcineurin (phosphoprotein phosphatase 2B) cAMP phosphodiesterase cAMP-gated olfactory channel cGMP-gated Na H11001 ,Ca 2H11001 channels (rod and cone cells) Glutamate decarboxylase Myosin light chain kinases NAD H11001 kinase Nitric oxide synthase Phosphoinositide 3-kinase Plasma membrane Ca 2H11001 ATPase (Ca 2H11001 pump) RNA helicase (p68) Some Proteins Regulated by Ca 2H11545 and Calmodulin TABLE 12–6 that generates IP 3 or the ion channel that regulates Ca 2H11001 release from the ER, or both. Whatever the mechanism, the effect is that one kind of signal (hormone concen- tration, for example) is converted into another (fre- quency and amplitude of intracellular [Ca 2H11001 ] “spikes”). Changes in intracellular [Ca 2H11001 ] are detected by Ca 2+ -binding proteins that regulate a variety of Ca 2H11001 - dependent enzymes. Calmodulin (CaM) (M r 17,000) is an acidic protein with four high-affinity Ca 2H11001 -binding sites. When intracellular [Ca 2H11001 ] rises to about 10 H110026 M (1 H9262M), the binding of Ca 2H11001 to calmodulin drives a con- formational change in the protein (Fig. 12–21). Calmod- ulin associates with a variety of proteins and, in its Ca 2H11001 - bound state, modulates their activities. Calmodulin is a member of a family of Ca 2H11001 -binding proteins that also includes troponin (p. 185), which triggers skeletal mus- cle contraction in response to increased [Ca 2H11001 ]. This family shares a characteristic Ca 2H11001 -binding structure, the EF hand (Fig. 12–21c). Calmodulin is also an integral subunit of a family of enzymes, the Ca 2H11545 /calmodulin-dependent protein kinases (CaM kinases I–IV). When intracellular [Ca 2H11001 ] increases in response to some stimulus, calmod- ulin binds Ca 2H11001 , undergoes a change in conformation, and activates the CaM kinase. The kinase then phos- phorylates a number of target enzymes, regulating their activities. Calmodulin is also a regulatory subunit of phosphorylase b kinase of muscle, which is activated by Ca 2H11001 . Thus Ca 2H11001 triggers ATP-requiring muscle con- tractions while also activating glycogen breakdown, pro- viding fuel for ATP synthesis. Many other enzymes are also known to be modulated by Ca 2H11001 through calmod- ulin (Table 12–6). 8885d_c12_444 2/20/04 1:21 PM Page 444 mac76 mac76:385_reb: SUMMARY 12.4 G Protein–Coupled Receptors and Second Messengers ■ A large family of plasma membrane receptors with seven transmembrane segments act through heterotrimeric G proteins. On ligand binding, these receptors catalyze the exchange of GTP for GDP bound to an associated G protein, forcing dissociation of the H9251 subunit of the G protein. This subunit stimulates or inhibits the activity of a nearby membrane-bound enzyme, changing the level of its second messenger product. ■ The H9252-adrenergic receptor binds epinephrine, then through a stimulatory G protein, G s , activates adenylyl cyclase in the plasma membrane. The cAMP produced by adenylyl cyclase is an intracellular second messenger that stimulates cAMP-dependent protein kinase, which mediates the effects of epinephrine by phosphorylating key proteins, changing their enzymatic activities or structural features. ■ The cascade of events in which a single molecule of hormone activates a catalyst that in turn activates another catalyst, and so on, results in large signal amplification; this is characteristic of most hormone-activated systems. ■ Some receptors stimulate adenylyl cyclase through G s ; others inhibit it through G i . Thus cellular [cAMP] reflects the integrated input of two (or more) signals. ■ Cyclic AMP is eventually eliminated by cAMP phosphodiesterase, and G s turns itself off by hydrolysis of its bound GTP to GDP. When the epinephrine signal persists, H9252-adrenergic receptor–specific protein kinase and arrestin 2 temporarily desensitize the receptor and cause it to move into intracellular vesicles. In some cases, arrestin also acts as a scaffold protein, bringing together protein components of a signaling pathway such as the MAPK cascade. ■ Some serpentine receptors are coupled to a plasma membrane phospholipase C that cleaves PIP 2 to diacylglycerol and IP 3 . By opening Ca 2H11001 channels in the endoplasmic reticulum, IP 3 raises cytosolic [Ca 2H11001 ]. Diacylglycerol and Ca 2H11001 act together to activate protein kinase C, which phosphorylates and changes the activity of specific cellular proteins. Cellular [Ca 2H11001 ] also regulates a number of other enzymes, often through calmodulin. 12.4 G Protein–Coupled Receptors and Second Messengers 445 (b) (c) EF hand (a) Ca 2+ E helix F helix FIGURE 12–21 Calmodulin. This is the protein mediator of many Ca 2H11001 -stimu- lated enzymatic reactions. Calmodulin has four high-affinity Ca 2H11001 -binding sites (K d H11015 0.1 to 1 H9262M). (a) A ribbon model of the crystal structure of calmodulin (PDB ID 1CLL). The four Ca 2H11001 -binding sites are occupied by Ca 2H11001 (purple). The amino-terminal domain is on the left; the carboxyl-terminal domain on the right. (b) Calmodulin associated with a helical domain (red) of one of the many enzymes it regulates, calmodulin-dependent protein kinase II (PDB ID 1CDL). Notice that the long central H9251 helix visible in (a) has bent back on itself in binding to the helical substrate domain. The central helix is clearly more flexible in solution than in the crystal. (c) Each of the four Ca 2H11001 -binding sites occurs in a helix-loop-helix motif called the EF hand, also found in many other Ca 2H11001 -binding proteins. 8885d_c12_445 2/20/04 3:16 PM Page 445 mac76 mac76:385_reb: Chapter 12 Biosignaling446 BOX 12–2 WORKING IN BIOCHEMISTRY FRET: Biochemistry Visualized in a Living Cell Fluorescent probes are commonly used to detect rapid biochemical changes in single living cells. They can be designed to give an essentially instantaneous report (within nanoseconds) on the changes in intracellular concentration of a second messenger or in the activ- ity of a protein kinase. Furthermore, fluorescence mi- croscopy has sufficient resolution to reveal where in the cell such changes are occurring. In one widely used procedure, the fluorescent probes are derived from a naturally occurring fluorescent protein, the green fluorescent protein (GFP) of the jellyfish Ae- quorea victoria (Fig. 1). When excited by absorption of a photon of light, GFP emits a photon (that is, it fluoresces) in the green region of the spectrum. GFP is an 11-stranded H9252 bar- rel, and the light-absorbing/emitting center of the pro- tein (its chromophore) comprises the tripeptide Ser 65 –Tyr 66 –Gly 67 , located within the barrel (Fig. 2). Variants of this protein, with different fluorescence spectra, can be produced by genetic engineering of the GFP gene. For example, in the yellow fluorescent protein (YFP), Ala 206 in GFP is replaced by a Lys residue, changing the wavelength of light absorption and fluorescence. Other variants of GFP fluoresce blue (BFP) or cyan (CFP) light, and a related protein (mRFP1) fluoresces red light (Fig. 3). GFP and its variants are compact structures that retain their abil- ity to fold into their native H9252-barrel conformation even when fused with another protein. Investigators are us- ing these fluorescent hybrid proteins as spectroscopic rulers to measure distances between interacting com- ponents within a cell. FIGURE 1 Aequorea victoria, a jellyfish abundant in Puget Sound, Washington State. FIGURE 2 Green fluorescent protein (GFP), with the fluorescent chro- mophore shown in ball-and-stick form (derived from PDB ID 1GFL). Chromophore (Ser 65 Tyr 66 Gly 67 ) 100 BFP CFP GFP mRFP1 80 60 Relative fluorescence 40 20 0 400 500 600 Wavelength (nm) 700 YFP FIGURE 3 Emission spectra of GFP variants. 8885d_c12_446 2/20/04 1:21 PM Page 446 mac76 mac76:385_reb: 12.4 G Protein–Coupled Receptors and Second Messengers 447 An excited fluorescent molecule such as GFP or YFP can dispose of the energy from the absorbed pho- ton in either of two ways: (1) by fluorescence, emitting a photon of slightly longer wavelength (lower energy) than the exciting light, or (2) by nonradiative fluores- cence resonance energy transfer (FRET), in which the energy of the excited molecule (the donor) passes directly to a nearby molecule (the acceptor) without emission of a photon, exciting the acceptor (Fig. 4). The acceptor can now decay to its ground state by flu- orescence; the emitted photon has a longer wavelength (lower energy) than both the original exciting light and the fluorescence emission of the donor. This second mode of decay (FRET) is possible only when donor and acceptor are close to each other (within 1 to 50 ?); the efficiency of FRET is inversely proportional to the sixth power of the distance between donor and acceptor. Thus very small changes in the distance between donor and acceptor register as very large changes in FRET, measured as the fluorescence of the acceptor molecule when the donor is excited. With sufficiently sensitive light detectors, this fluorescence signal can be located to specific regions of a single, living cell. FRET has been used to measure [cAMP] in living cells. The gene for GFP is fused with that for the reg- ulatory subunit (R) of cAMP-dependent protein ki- nase, and the gene for BFP is fused with that for the catalytic subunit (C) (Fig. 5). When these two hybrid proteins are expressed in a cell, BFP (donor; excita- tion at 380 nm, emission at 460 nm) and GFP (ac- ceptor; excitation at 475 nm, emission at 545 nm) in the inactive PKA (R 2 C 2 tetramer) are close enough to undergo FRET. Wherever in the cell [cAMP] increases, the R 2 C 2 complex dissociates into R 2 and 2C and the FRET signal is lost, because donor and acceptor are now too far apart for efficient FRET. Viewed in the fluorescence microscope, the region of higher [cAMP] has a minimal GFP signal and higher BFP signal. Mea- suring the ratio of emission at 460 nm and 545 nm gives a sensitive measure of the change in [cAMP]. By determining this ratio for all regions of the cell, the investigator can generate a false color image of the 433 nm 433 nm 476 nm 527 nm CFP YFP protein– protein interaction Genetically engineered hybrid proteins FRET FIGURE 4 When the donor protein (CFP) is excited with mono- chromatic light of wavelength 433 nm, it emits fluorescent light at 476 nm (left). When the (red) protein fused with CFP interacts with the (purple) protein fused with YFP, that interaction brings CFP and YFP close enough to allow fluorescence resonance energy transfer (FRET) between them. Now, when CFP absorbs light of 433 nm, in- stead of fluorescing at 476 nm, it transfers energy directly to YFP, which then fluoresces at its characteristic emission wavelength, 527 nm. The ratio of light emission at 527 and 476 nm is therefore a measure of the interaction of the red and purple protein. 460 nm 380 nm 545 nm BFP 380 nm (inactive) (active) cAMP-dependent protein kinase (PKA) GFP R no emission at 545 nm R C R RR C C C cAMPcAMP + FRET 433 nm FIGURE 5 Measuring [cAMP] with FRET. Gene fusion creates hy- brid proteins that exhibit FRET when the PKA regulatory and cat- alytic subunits are associated (low [cAMP]). When [cAMP] rises, the subunits dissociate, and FRET ceases. The ratio of emission at 460 nm (dissociated) and 545 nm (complexed) thus offers a sensitive measure of [cAMP]. (continued on next page) 8885d_c12_447 2/20/04 1:22 PM Page 447 mac76 mac76:385_reb: 12.5 Multivalent Scaffold Proteins and Membrane Rafts About 10% of the 30,000 to 35,000 genes in the human genome encode signaling proteins—receptors, G pro- teins, enzymes that generate second messengers, pro- tein kinases (H11022500), proteins involved in desensitiza- tion, and ion channels. Not every signaling protein is expressed in a given cell type, but most cells doubtless contain many such proteins. How does one protein find another in a signaling pathway, and how are their inter- actions regulated? As is becoming clear, the reversible phosphorylation of Tyr, Ser, and Thr residues in signal- ing proteins creates docking sites for other proteins, and many signaling proteins are multivalent in that they can interact with several different proteins simul- taneously to form multiprotein signaling complexes. In this section we present a few examples to illustrate the general principles of protein interactions in signaling. Protein Modules Bind Phosphorylated Tyr, Ser, or Thr Residues in Partner Proteins We have seen that the protein Grb2 in the insulin sig- naling pathway (Fig. 12–6) binds through its SH2 do- main to other proteins that contain exposed P –Tyr residues. The human genome encodes at least 87 SH2- containing proteins, many already known to participate in signaling. The P –Tyr residue is bound in a deep pocket in an SH2 domain, with each of its phosphate oxy- gens participating in hydrogen-bonding or electrostatic interactions; the positive charges on two Arg residues figure prominently in the binding. Subtle differences in the structure of SH2 domains in different proteins account for the specificities of their interactions with various P –Tyr-containing proteins. The three to five residues on the carboxyl-terminal side of the P –Tyr residue are critical in determining the specificity of in- teractions with SH2 domains (Fig. 12–22). PTB domains (phosphotyrosine-binding domains) also bind P –Tyr in partner proteins, but their critical sequences and three-dimensional structures distinguish them from SH2 domains. The human genome encodes 24 proteins that contain PTB domains, including IRS-1, which we have already met in its role as a scaffold pro- tein in insulin-signal transduction (Fig. 12–6). Many of the signaling protein kinases, including PKA, PKC, PKG, and members of the MAPK cascade, phosphorylate Ser or Thr residues in their target pro- teins, which in some cases acquire the ability to inter- act with partner proteins through the phosphorylated residue, triggering a downstream process. An alphabet soup of domains that bind P –Ser or P –Thr residues has been identified, and more are sure to be found. Each domain favors a certain sequence around the phosphor- ylated residue, so the domains represent families of highly specific recognition sites, able to bind to a spe- cific subset of phosphorylated proteins. Chapter 12 Biosignaling448 BOX 12–2 WORKING IN BIOCHEMISTRY (continued from previous page) cell in which the ratio, or relative [cAMP], is repre- sented by the intensity of the color. Images recorded at timed intervals reveal changes in [cAMP] over time. A variation of this technology has been used to measure the activity of PKA in a living cell (Fig. 6). Researchers create a phosphorylation target for PKA by producing a hybrid protein containing four ele- ments: YFP (acceptor); a short peptide with a Ser residue surrounded by the consensus sequence for PKA; a P –Ser-binding domain (called 14-3-3); and CFP (donor). When the Ser residue is not phosphor- ylated, 14-3-3 has no affinity for the Ser residue and the hybrid protein exists in an extended form, with the donor and acceptor too far apart to generate a FRET signal. Wherever PKA is active in the cell, it phosphorylates the Ser residue of the hybrid protein, and 14-3-3 binds to the P –Ser. In doing so, it draws YFP and CFP together and a FRET signal is detected with the fluorescence microscope, revealing the pres- ence of active PKA. ATP PKA consensus sequence 14-3-3 (Phosphoserine- binding domain) ADP PKA YFP 433 nm 476 nm CFP Ser 433 nm FRET 527 nm P FIGURE 6 Measuring the activity of PKA with FRET. An engineered protein links YFP and CFP via a peptide that contains a Ser residue surrounded by the consensus sequence for phosphorylation by PKA, and the 14-3-3 phosphoserine binding domain. Active PKA phos- phorylates the Ser residue, which docks with the 14-3-3 binding do- main, bringing the fluorescence proteins close enough to allow FRET to occur, revealing the presence of active PKA. 8885d_c12_448 2/20/04 1:22 PM Page 448 mac76 mac76:385_reb: FIGURE 12–22 Structure of an SH2 domain and its interaction with a P –Tyr residue in a partner protein. (PDB ID 1SHC) The SH2 domain is shown as a gray surface contour representation. The phosphorus of the phosphate group in the interacting P –Tyr is visible as an orange sphere; most of the residue is obscured in this view. The next few residues toward the carboxyl end of the partner protein are shown in red. The SH2 domain interacts with P –Tyr (which, as the phosphorylated residue, is assigned the index position 0) and also with the next three residues toward the carboxyl terminus (designated H110011, H110012, H110013). The residues important in the P –Tyr residue are conserved in all SH2 domains. Some SH2 domains (Src, Fyn, Hck, Nck) favor negatively charged residues in the H110011 and H110012 positions; others (PLC-H92531, SHP-2) have a long hydrophobic groove that selects for aliphatic residues in positions H110011 to H110015. These differences define subclasses of SH2 domains that have different partner specificities. In some cases, the domain-binding partner is inter- nal. Phosphorylation of some protein kinases inhibits their activity by favoring the interaction of an SH2 do- main with a P –Tyr in another domain of the same en- zyme. For example, the soluble protein Tyr kinase Src, when phosphorylated on a critical Tyr residue, is ren- dered inactive as an SH2 domain needed to bind to the substrate protein instead binds to an internal P –Tyr (Fig. 12–23). Glycogen synthase kinase 3 (GSK3) is in- active when phosphorylated on a Ser residue in its auto- inhibitory domain (Fig. 12–23b). Dephosphorylation of that domain frees the enzyme to bind and phosphory- late its target proteins. Similarly, the polar head group of the phospholipid PIP 3 , protruding from the inner leaf- let of the plasma membrane, provides points of attach- ment for proteins that contain SH3 and other domains. 12.5 Multivalent Scaffold Proteins and Membrane Rafts 449 (a) Autoinhibited Src kinase SH3 SH3 SH2 SH2 P Tyr Tyr TyrHO Pro Pro Active site Active; substrate positioned for phosphorylation P Tyr Tyr Tyr Autoinhibited (b) Glycogen synthase Active site GSK3 Ser Ser HO Ser Ser Active; substrate positioned for phosphorylation P SerP GSK3 FIGURE 12–23 Mechanism of autoinhibition of Src Tyr kinase and GSK3. (a) In the active form of Src kinase, an SH2 domain binds a P –Tyr in the substrate, and an SH3 domain binds a proline-rich region of the substrate, lining up the active site of the kinase with several target Tyr residues in the substrate. When Src is phosphorylated on a specific Tyr residue, the SH2 domain binds to the internal P –Tyr instead of to the P –Tyr of the substrate, preventing productive binding of the kinase to its protein substrate; the enzyme is thus autoinhibited. (b) In the autoinhibited glycogen synthase kinase 3 (GSK3), an internal P –Ser residue is bound to an internal P –Ser-binding domain (top). Dephosphorylation of this internal Ser residue leaves the P –Ser-binding site of GSK3 avail- able to bind P –Ser in a protein substrate, and thus to position the kinase to phosphory- late neighboring Ser residues (bottom). 8885d_c12_449 2/20/04 1:22 PM Page 449 mac76 mac76:385_reb: Most of the proteins involved in signaling at the plasma membrane have one or more protein- or phos- pholipid-binding domains; many have three or more, and thus are multivalent in their interactions with other sig- naling proteins. Figure 12–24 shows a few of the many multivalent proteins known to participate in signaling. A remarkable picture of signaling pathways has emerged from studies of many signaling proteins and the multiple binding domains they contain (Fig. 12–25). An initial signal results in phosphorylation of the re- ceptor or a target protein, triggering the assembly of large multiprotein complexes, held together on scaffolds made from adaptor proteins with multivalent binding ca- pacities. Some of these complexes have several protein kinases that activate each other in turn, producing a cas- cade of phosphorylation and a great amplification of the initial signal. Animal cells also have phosphotyrosine phosphatases (PTPases), which remove the phosphate from P –Tyr residues, reversing the effect of phosphor- ylation. Some of these phosphatases are receptorlike membrane proteins, presumably controlled by extracel- lular factors not yet identified; other PTPases are solu- ble and contain SH2 domains. In addition, animal cells have protein phosphoserine and phosphothreonine phosphatases, which reverse the effects of Ser- and Thr- specific protein kinases. We can see, then, that signal- ing occurs in protein circuits, effectively hard-wired from signal receptor to response effector and able to be switched off instantly by the hydrolysis of a single phos- phate ester bond. The multivalency of signaling proteins allows for the assembly of many different combinations of signaling modules, each combination presumably suited to partic- ular signals, cell types, and metabolic circumstances. The large variety of protein kinases and of phosphoprotein- binding domains, each with its own specificity (the con- sensus sequence required in its substrate), provides for many permutations and combinations and many differ- ent signaling circuits of extraordinary complexity. And given the variety of specific phosphatases that reverse Chapter 12 Biosignaling450 SH2 Adaptor Grb2 Shc SH3 DNA STAT PTB Tyr kinase Src Scaffold Kinase Ras signaling Phosphatase Signal regulation Transcription Phospholipid second- messenger signaling SH2 SH3 SH2 SH2SH3 Tyr phosphatase Shp2SH2SH2 SH3 SH2SH2 C2PH RasGAPGTPase-activating SH2 SH3SH2 C2PH PH PH PLC PLCgPLC TA SH2 SOCSSOCS Binding domains proline-rich protein or membrane lipid PIP 3 Tyr– P Tyr– PIP 3 phospholipids (Ca 2+ -dependent) DNA transcriptional activation carboxyl-terminal domain marking protein for attachment of ubiquitin P FIGURE 12–24 Some binding modules of signaling proteins. Each protein is represented by a line (with the amino terminus to the left); symbols indicate the location of conserved binding domains (with specificities as listed in the key; PH denotes plextrin homology; other abbreviations explained in the text); green boxes indicate catalytic ac- tivities. The name of each protein is given at its carboxyl-terminal end. These signaling proteins interact with phosphorylated proteins or phospholipids in many permutations and combinations to form inte- grated signaling complexes. 8885d_c12_450 2/20/04 1:22 PM Page 450 mac76 mac76:385_reb: the action of protein kinases, some under specific types of external control, a cell can quickly “disconnect” the entire protein circuitry of a signaling pathway. Together, these mechanisms confer a huge capacity for cellular reg- ulation in response to signals of many types. Membrane Rafts and Caveolae May Segregate Signaling Proteins Membrane rafts are regions of the membrane bilayer en- riched in sphingolipids, sterols, and certain proteins, in- cluding many attached to the bilayer by GPI anchors (Chapter 11). Some receptor Tyr kinases, such as the receptors for epidermal growth factor (EGF) and platelet-derived growth factor (PDGF), appear to be lo- calized in rafts; other signaling proteins, such as the small G protein Ras (which is prenylated) and the hetero- trimeric G protein G s (also prenylated, on the H9251 and H9253 subunits), are not. Growing evidence suggests that this sequestration of signaling proteins is functionally signif- icant. When cholesterol is removed from rafts by treat- ment with cyclodextrin (which binds cholesterol and removes it from membranes), the rafts are disrupted and a number of signaling pathways become defective. How might localization in rafts influence signaling through a receptor? There are several possibilities. If a receptor Tyr kinase in a raft is phosphorylated, and the phosphotyrosine phosphatase that reverses this phos- phorylation is in another raft, then dephosphorylation of the Tyr kinase will be slowed or prevented. If two sig- naling proteins must interact during transduction of a signal, the probability of encounters between these pro- teins is greatly enhanced if both are in the same raft. Interactions between scaffold proteins might be strong enough to pull into a raft a signaling protein not nor- mally located there, or strong enough to pull receptors out of a raft. For example, the EGF receptor in isolated fibroblasts is normally concentrated in the specialized rafts called caveolae (see Fig. 11–21), but treatment with EGF causes the receptor to leave the raft. This mi- gration depends on the receptor’s protein kinase activ- ity; mutant receptors lacking this activity remain in the rafts during treatment with EGF. Caveolin, an integral membrane protein localized in caveolae, is phosphory- lated on Tyr residues in response to insulin, and phos- phorylation may allow the now-activated EGF receptor to draw its binding partners into the raft. Finally, an- other example of the clustering of signaling proteins in rafts is the H9252-adrenergic receptor. This receptor is seg- regated in membrane rafts that also contain the G pro- teins, adenylyl cyclase, PKA, and a specific protein phos- phatase, PP2, providing a highly integrated signaling unit. Spatial segregation of signaling proteins in rafts adds yet another dimension to the already complex processes initiated by extracellular signals. SUMMARY 12.5 Multivalent Scaffold Proteins and Membrane Rafts ■ Many signaling proteins have domains that bind phosphorylated Tyr, Ser, or Thr residues in other proteins; the binding specificity for each domain is determined by sequences that adjoin the phosphorylated residue. ■ SH2 and PTB domains bind to proteins containing P –Tyr residues; other domains bind P –Ser and P –Thr residues in various contexts. ■ Plextrin homology domains bind the membrane phospholipid PIP 3 . ■ Many signaling proteins are multivalent, with several different binding modules. By combining the substrate specificities of various protein kinases with the specificities of domains that bind phosphorylated Ser, Thr, or Tyr residues, and with phosphatases that can rapidly inactivate a pathway, cells create a large number of multiprotein signaling complexes. ■ Membrane rafts and caveolae sequester groups of signaling proteins in small regions of the plasma membrane, enhancing their interactions and making signaling more efficient. 12.5 Multivalent Scaffold Proteins and Membrane Rafts 451 14-3-3 MEK ERK MP1 IRS-1 Insulin receptor P P P P P P PKC PKB Raf-1 Grb2 Sos PI-3K Ras PIP 3 PIP 3 FIGURE 12–25 Insulin-induced formation of supramolecular signal- ing complexes. The binding of insulin to its receptor sets off a series of events that lead eventually to the formation of membrane-associated complexes involving the 12 signaling proteins shown here, as well as others. Phosphorylation of Tyr residues in the insulin receptor initiates complex formation, and dephosphorylation of any of the phospho- proteins breaks the circuit. Four general types of interaction hold the complex together: the binding of a protein to a second phosphopro- tein through SH2 or PTB domains in the first (red); the binding of SH3 domains in the first with proline-rich domains in the second (orange); the binding of PH domains in one protein to the phospholipid PIP 3 in the plasma membrane (blue); or the association of a protein (RAS) with the plasma membrane through a lipid covalently bound to the pro- tein (yellow). Two proteins shown here are not described in the text: 14-3-3, which binds a P –Ser in Raf and mediates its interaction with MEK; and MP1, a scaffold protein that cements the links between Raf, MEK, and ERK. 8885d_c12_451 2/20/04 1:22 PM Page 451 mac76 mac76:385_reb: 12.6 Signaling in Microorganisms and Plants Much of what we have said here about signaling relates to mammalian tissues or cultured cells from such tis- sues. Bacteria, eukaryotic microorganisms, and vascu- lar plants must also respond to a variety of external sig- nals, such as O 2 , nutrients, light, noxious chemicals, and so on. We turn here to a brief consideration of the kinds of signaling machinery used by microorganisms and plants. Bacterial Signaling Entails Phosphorylation in a Two-Component System E. coli responds to a number of nutrients in its envi- ronment, including sugars and amino acids, by swim- ming toward them, propelled by one or a few flagella. A family of membrane proteins have binding domains on the outside of the plasma membrane to which specific attractants (sugars or amino acids) bind (Fig. 12–26). Ligand binding causes another domain on the inside of the plasma membrane to phosphorylate itself on a His residue. This first component of the two-component system, the receptor His kinase, then catalyzes the transfer of the phosphoryl group from the His residue to an Asp residue on a second, soluble protein, the re- sponse regulator; this phosphoprotein moves to the base of the flagellum, carrying the signal from the mem- brane receptor. The flagellum is driven by a rotary mo- tor that can propel the cell through its medium or cause it to stall, depending on the direction of the motor’s ro- tation. Information from the receptor allows the cell to determine whether it is moving toward or away from the source of the attractant. If its motion is toward the at- tractant, the response regulator signals the cell to con- tinue in a straight line; if away from it, the cell tumbles momentarily, acquiring a new direction. Repetition of this behavior results in a random path, biased toward movement in the direction of increasing attractant concentration. E. coli detects not only sugars and amino acids but also O 2 , extremes of temperature, and other environ- mental factors, using this basic two-component system. Two-component systems have been detected in many other bacteria, including gram-positive and gram- negative eubacteria and archaebacteria, as well as in protists and fungi. Clearly this signaling mechanism de- veloped early in the course of cellular evolution and has been conserved. Various signaling systems used by animal cells also have analogs in the prokaryotes. As the full genomic se- quences of more, and more diverse, bacteria become known, researchers have discovered genes that encode proteins similar to protein Ser or Thr kinases, Ras-like proteins regulated by GTP binding, and proteins with SH3 domains. Receptor Tyr kinases have not been detected in bacteria, but P –Tyr residues do occur in some bacterial proteins, so there must be an enzyme that phosphorylates Tyr residues. Signaling Systems of Plants Have Some of the Same Components Used by Microbes and Mammals Like animals, vascular plants must have a means of com- munication between tissues to coordinate and direct growth and development; to adapt to conditions of O 2 , nutrients, light, and temperature; and to warn of the presence of noxious chemicals and damaging pathogens (Fig. 12–27). At least a billion years of evolution have passed since the plant and animal branches of the eu- karyotes diverged, which is reflected in the differences in signaling mechanisms: some plant mechanisms are conserved—that is, are similar to those in animals (pro- tein kinases, scaffold proteins, cyclic nucleotides, elec- trogenic ion pumps, and gated ion channels); some are similar to bacterial two-component systems; and some are unique to plants (light-sensing mechanisms, for ex- Chapter 12 Biosignaling452 AAttractant Receptor His kinase (component 1) A His A His His ATP ADP E. coli Response regulator (component 2) Plasma membrane Rotary motor (controls flagellum) Phosphorylated form of component 2 reverses direction of motor Asp Asp PP FIGURE 12–26 The two-component signaling mechanism in bacterial chemotaxis. When an attractant ligand (A) binds to the receptor domain of the membrane-bound receptor, a protein His kinase in the cytosolic domain (component 1) is activated and autophosphorylates on a His residue. This phosphoryl group is then trans- ferred to an Asp residue on component 2 (in some cases a separate protein; in others, another domain of the receptor protein). After phosphorylation on Asp, component 2 moves to the base of the flagellum, where it determines the direction of rotation of the flagellar motor. 8885d_c12_452 2/20/04 1:23 PM Page 452 mac76 mac76:385_reb: ample) (Table 12–7). The genome of the widely stud- ied plant Arabidopsis thaliana, for example, encodes about 1,000 protein Ser/Thr kinases, including about 60 MAPKs and nearly 400 membrane-associated receptor kinases that phosphorylate Ser or Thr residues; a vari- ety of protein phosphatases; scaffold proteins that bring other proteins together in signaling complexes; enzymes for the synthesis and degradation of cyclic nucleotides; and 100 or more ion channels, including about 20 gated by cyclic nucleotides. Inositol phospholipids are pres- ent, as are kinases that interconvert them by phospho- rylation of inositol head groups. However, some types of signaling proteins common in animal tissues are not present in plants, or are rep- resented by only a few genes. Cyclic nucleotide– dependent protein kinases (PKA and PKG) appear to be absent, for example. Heterotrimeric G proteins and protein Tyr kinase genes are much less prominent in the plant genome, and serpentine (G protein–coupled) receptors, the largest gene family in the human genome (H110221,000 genes), are very sparsely represented in the plant genome. DNA-binding nuclear steroid re- ceptors are certainly not prominent, and may be ab- sent from plants. Although plants lack the most widely conserved light-sensing mechanism present in animals (rhodopsin, with retinal as pigment), they have a rich collection of other light-detecting mechanisms not found in animal tissues—phytochromes and cryp- tochromes, for example (Chapter 19). The kinds of compounds that elicit signals in plants are similar to certain signaling molecules in mammals (Fig. 12–28). Instead of prostaglandins, plants have jas- monate; instead of steroid hormones, brassinosteroids. 12.6 Signaling in Microorganisms and Plants 453 Gravity Light Humidity Temperature Wind Herbivores Pathogens Pathogens Parasites Microorganisms O 2 Minerals Toxic molecules Water status CO 2 C 2 H 4 FIGURE 12–27 Some stimuli that produce responses in plants. TABLE 12–7 Signaling Components Present in Mammals, Plants, or Bacteria Signaling protein Mammals Plants Bacteria Ion channels H11001H11001H11001 Electrogenic ion pumps H11001H11001H11001 Two-component His kinases H11001H11001H11001 Adenylyl cyclase H11001H11001H11001 Guanylyl cyclase H11001H11001 ? Receptor protein kinases (Ser/Thr) H11001H11001 ? Ca 2H11001 as second messenger H11001H11001 ? Ca 2H11001 channels H11001H11001 ? Calmodulin, CaM-binding protein H11001H11001H11002 MAPK cascade H11001H11001H11002 Cyclic nucleotide–gated channels H11001H11001H11002 IP 3 -gated Ca 2H11001 channels H11001H11001H11002 Phosphatidylinositol kinases H11001H11001H11002 Serpentine receptors H11001H11001/H11002H11001 Trimeric G proteins H11001H11001/H11002H11002 PI-specific phospholipase C H11001 ? H11002 Tyrosine kinase receptors H11001 ? H11002 SH2 domains H11001 ?? Nuclear steroid receptors H11001H11002H11002 Protein kinase A H11001H11002H11002 Protein kinase G H11001H11002H11002 8885d_c12_453 2/20/04 1:23 PM Page 453 mac76 mac76:385_reb: About 100 different small peptides serve as plant signals, and both plants and animals use compounds derived from aromatic amino acids as signals. Plants Detect Ethylene through a Two-Component System and a MAPK Cascade The receptors for the plant hormone ethylene (CH 2 UCH 2 ) are related in primary sequence to the receptor His kinases of the bacterial two-component systems and probably evolved from them; the cyanobac- terial origin of chloroplasts (see Fig. 1–36) may have brought the bacterial signaling genes into the plant cell nucleus. In Arabidopsis, the two-component signaling system is contained within a single protein. The first downstream component affected by ethylene signaling is a protein Ser/Thr kinase (CTR-1; Fig. 12–29) with se- quence homology to Raf, the protein kinase that begins the MAPK cascade in the mammalian response to in- sulin (see the comparison in Fig. 12–30). In plants, in the absence of ethylene, the CTR-1 kinase is active and inhibits the MAPK cascade, preventing transcription of ethylene-responsive genes. Exposure to ethylene inac- tivates the CTR-1 kinase, thereby activating the MAPK cascade that leads to activation of the transcription fac- tor EIN3. Active EIN3 stimulates the synthesis of a sec- ond transcription factor (ERF1), which in turn activates transcription of a number of ethylene-responsive genes; the gene products affect processes ranging from seedling development to fruit ripening. Although apparently derived from the bacterial two- component signaling system, the ethylene system in Arabidopsis is different in that the His kinase activity that defines component 1 in bacteria is not essential to the transduction in Arabidopsis. The genome of the cyanobacterium Anabaena encodes proteins with both an ethylene-binding domain and an active His kinase do- main. It seems likely that in the course of evolution, the ethylene receptor of vascular plants was derived from that of a cyanobacterial endosymbiont, and that the bac- terial His kinase became a Ser/Thr kinase in the plant. Chapter 12 Biosignaling454 O COO H11002 Jasmonate Plants Animals O COO H11002 COO H11002 Prostaglandin E 1 OH HO 8 12 OH Estradiol H 3 HO C Brassinolide (a brassinosteroid) HO H O O HO OH OH Serotonin (5-hydroxytryptamine) HO N H Indole-3-acetate (an auxin) N H H11001 NH 3 Cytosol Nucleus MAPK cascade DNA mRNA DNA mRNA Ethylene- response proteins Ethylene Ethylene receptor Two-component system Plasma membrane CTR-1 MAPKKK EIN2 EIN3 11 22 ERF1 FIGURE 12–28 Structural similarities between plant and animal sig- nals. The plant signals jasmonate, indole-3-acetate, and brassinolide resemble the mammalian signals prostaglandin E 1 , serotonin, and estradiol. FIGURE 12–29 Transduction mechanism for detection of ethylene by plants. The ethylene receptor in the plasma membrane (red) is a two-component system contained within a single protein, which has both a receptor domain (component 1) and a response regulator do- main (component 2). The receptor controls (in ways we do not yet un- derstand) the activity of CTR1, a protein kinase similar to MAPKKKs and therefore presumed to be part of a MAPK cascade. CTR1 is a neg- ative regulator of the ethylene response; when CTR1 is inactive, the ethylene signal passes through the gene product EIN2 (thought to be a nuclear envelope protein), which somehow causes increased syn- thesis of ERF1, a transcription factor; ERF1 in turn stimulates expres- sion of proteins specific to the ethylene response. 8885d_c12_454 2/20/04 1:25 PM Page 454 mac76 mac76:385_reb: Receptorlike Protein Kinases Transduce Signals from Peptides and Brassinosteroids One common motif in plant signaling involves recep- torlike kinases (RLKs) with a single helical segment in the plasma membrane that connects a receptor do- main on the outside of the membrane with a protein Ser/Thr kinase on the cytoplasmic side. This type of re- ceptor participates in the defense mechanism triggered by infection with a bacterial pathogen (Fig. 12–30a). The signal to turn on the genes needed for defense against infection is a peptide (flg22) released by breakdown of flagellin, the major protein of the bacterial flagellum. Binding of flg22 to the FLS2 receptor of Arabidopsis induces receptor dimerization and autophosphorylation on Ser and Thr residues, and the downstream effect is activation of a MAPK cascade like that described above for insulin action (Fig. 12–6). The final kinase in this cascade activates a specific transcription factor, trig- gering synthesis of the proteins that defend against the bacterial infection. The steps between receptor phos- phorylation and the MAPK cascade are not yet known. A phosphoprotein phosphatase (KAPP) associates with the active receptor protein and inactivates it by de- phosphorylation to end the response. The MAPK cascade in the plant’s defense against bacterial pathogens is remarkably similar to the innate immune response triggered by bacterial lipopolysac- charide and mediated by the Toll-like receptors in mam- mals (Fig. 12–30b). Other membrane receptors use sim- ilar mechanisms to activate a MAPK cascade, ultimately activating transcription factors and turning on the genes essential to the defense response. Most of the several hundred RLKs in plants are presumed to act in similar ways: ligand binding in- duces dimerization and autophosphorylation, and the 12.6 Signaling in Microorganisms and Plants 455 (a) (b)Plant (Arabidopsis) Mammal Dimeric receptor flg22 Protein kinase domain Transcription factors WRKY22/29 Immune- response proteins Transcription factors Jun, Fos Transcription factors NFkB Immune- response proteins MAPK cascade Protein kinase IRAK LPS Flagellin Receptors Plasma membrane Ser, Thr MAPK cascade MAPK cascade P FIGURE 12–30 Similarities between the signaling pathways that trig- ger immune responses in plants and animals. (a) In the plant Ara- bidopsis thaliana, the peptide flg22, derived from the flagella of a bac- terial pathogen, binds to its receptor in the plasma membrane, causing the receptors to form dimers and triggering autophosphorylation of the cytosolic protein kinase domain on a Ser or Thr residue (not a Tyr). Autophosphorylation activates the receptor protein kinase, which then phosphorylates downstream proteins. The activated receptor also ac- tivates (by means unknown) a MAPKKK. The resulting kinase cascade leads to phosphorylation of a nuclear protein that normally inhibits the transcription factors WRKY22 and 29, triggering proteolytic degra- dation of the inhibitor and freeing the transcription factors to stimu- late gene expression related to the immune response. (b) In mammals, the toxic bacterial lipopolysaccharide (LPS; see Fig. 7–32) is detected by plasma membrane receptors that associate with and activate a sol- uble protein kinase (IRAK). The major flagellar protein of pathogenic bacteria acts through a similar receptor to activate IRAK. Then IRAK initiates two distinct MAPK cascades that end in the nucleus, causing the synthesis of proteins needed in the immune response. Jun, Fos, and NFH9260B are transcription factors. 8885d_c12_455 2/20/04 1:25 PM Page 455 mac76 mac76:385_reb: activated receptor kinase triggers downstream responses by phosphorylating key proteins at Ser or Thr residues. The ligands for these kinases have been identified in only a few cases: brassinosteroids, the peptide trigger for the self-incompatibility response that prevents self- pollination, and CLV1 peptide, a factor involved in reg- ulating the fate of stem cells (undifferentiated cells) in plant development. SUMMARY 12.6 Signaling in Microorganisms and Plants ■ Bacteria and unicellular eukaryotes have a variety of sensory systems that allow them to sample and respond to their environment. In the two-component system, a receptor His kinase senses the signal and autophosphory- lates a His residue, then phosphorylates the response regulator on an Asp residue. ■ Plants respond to many environmental stimuli, and employ hormones and growth factors to coordinate the development and metabolic activities of their tissues. Plant genomes encode hundreds of signaling proteins, including some very similar to those used in signal transductions in mammalian cells. ■ Two-component signaling mechanisms common in bacteria have been acquired in altered forms by plants. Cyanobacteria use typical two-component systems in the detection of chemical signals and light; plants use related proteins—which autophosphorylate on Ser/Thr, not His, residues—to detect ethylene. ■ Plant receptorlike kinases (RLKs), with an extracellular ligand-binding domain, a single transmembrane segment, and a cytosolic protein kinase domain, participate in detecting a wide variety of stimuli, including peptides that originate from pathogens, brassinosteroid hormones, self-incompatible pollen, and developmental signals. RLKs autophosphorylate Ser/Thr residues, then activate downstream proteins that in some cases are MAPK cascades. The end result of many such signals is increased transcription of specific genes. 12.7 Sensory Transduction in Vision, Olfaction, and Gustation The detection of light, smells, and tastes (vision, olfac- tion, and gustation, respectively) in animals is accom- plished by specialized sensory neurons that use signal- transduction mechanisms fundamentally similar to those that detect hormones, neurotransmitters, and growth factors. An initial sensory signal is amplified greatly by mechanisms that include gated ion channels and intra- cellular second messengers; the system adapts to con- tinued stimulation by changing its sensitivity to the stimulus (desensitization); and sensory input from sev- eral receptors is integrated before the final signal goes to the brain. Light Hyperpolarizes Rod and Cone Cells of the Vertebrate Eye In the vertebrate eye, light entering through the pupil is focused on a highly organized collection of light- sensitive neurons (Fig. 12–31). The light-sensing cells are of two types: rods (about 10 9 per retina), which sense low levels of light but cannot discriminate colors, and cones (about 3 H11003 10 6 per retina), which are less sensitive to light but can discriminate colors. Both cell types are long, narrow, specialized sensory neurons with two distinct cellular compartments: the outer segment contains dozens of membranous disks loaded with the membrane protein rhodopsin, and the inner segment contains the nucleus and many mitochondria, which produce the ATP essential to phototransduction. Chapter 12 Biosignaling456 Light To optic nerve Ganglion neurons Interconnecting neurons Rod Cone Light Lens Eye Retina Optic nerve FIGURE 12–31 Light reception in the vertebrate eye. The lens of the eye focuses light on the retina, which is composed of layers of neu- rons. The primary photosensory neurons are rod cells (yellow), which are responsible for high-resolution and night vision, and cone cells of three subtypes (pink), which initiate color vision. The rods and cones form synapses with several ranks of interconnecting neurons that con- vey and integrate the electrical signals. The signals eventually pass from ganglion neurons through the optic nerve to the brain. 8885d_c12_456 2/20/04 1:25 PM Page 456 mac76 mac76:385_reb: Like other neurons, rods and cones have a trans- membrane electrical potential (V m ), produced by the electrogenic pumping of the Na H11001 K H11001 ATPase in the plasma membrane of the inner segment (Fig. 12–32). Also con- tributing to the membrane potential is an ion channel in the outer segment that permits passage of either Na H11001 or Ca 2H11001 and is gated (opened) by cGMP. In the dark, rod cells contain enough cGMP to keep this channel open. The membrane potential is therefore determined by the net difference between the Na H11001 and K H11001 pumped by the inner segment (which polarizes the membrane) and the influx of Na H11001 through the ion channels of the outer seg- ment (which tends to depolarize the membrane). The essence of signaling in the retinal rod or cone cell is a light-induced decrease in the concentration of cGMP, which causes the cGMP-gated ion channel to close. The plasma membrane then becomes hyperpolar- ized by the Na H11001 K H11001 ATPase. Rod and cone cells synapse with interconnecting neurons (Fig. 12–31) that carry information about the electrical activity to the ganglion neurons near the inner surface of the retina. The gan- glion neurons integrate the output from many rod or cone cells and send the resulting signal through the optic nerve to the visual cortex of the brain. Light Triggers Conformational Changes in the Receptor Rhodopsin Visual transduction begins when light falls on rhodopsin, many thousands of molecules of which are present in each disk of the outer segments of rod and cone cells. Rhodopsin (M r 40,000) is an integral protein with seven membrane-spanning H9251 helices (Fig. 12–33), the charac- teristic serpentine architecture. The amino-terminal do- main projects into the disk, and the carboxyl-terminal domain faces the cytosol of the outer segment. The light-absorbing pigment (chromophore) 11-cis-retinal is covalently attached to opsin, the protein component of rhodopsin, through a Schiff base to a Lys residue. The retinal lies near the middle of the bilayer (Fig. 12–33), oriented with its long axis approximately in the plane of the membrane. When a photon is absorbed by the reti- nal component of rhodopsin, the energy causes a pho- tochemical change; 11-cis-retinal is converted to all- trans-retinal (see Figs 1–18b, 10–21). This change in the structure of the chromophore causes conformational changes in the rhodopsin molecule—the first stage in visual transduction. Excited Rhodopsin Acts through the G Protein Transducin to Reduce the cGMP Concentration In its excited conformation, rhodopsin interacts with a second protein, transducin, which hovers nearby on the cytoplasmic face of the disk membrane (Fig. 12–33). Transducin (T) belongs to the same family of hetero- trimeric GTP-binding proteins as G s and G i . Although 12.7 Sensory Transduction in Vision, Olfaction, and Gustation 457 Inner segment Outer segment Na H11545 K H11545 ATPase Ion channel open Ion channel closed Light Na H11545 Na H11545 Na H11545 Na H11545 Ca 2H11545 V m H11005H1100245 mV V m H11005H1100275 mV cGMP FIGURE 12–32 Light-induced hyperpolarization of rod cells. The rod cell consists of an outer segment that is filled with stacks of membra- nous disks (not shown) containing the photoreceptor rhodopsin and an inner segment that contains the nucleus and other organelles. Cones have a similar structure. ATP in the inner segment powers the Na H11001 K H11001 ATPase, which creates a transmembrane electrical potential by pump- ing 3 Na H11001 out for every 2 K H11001 pumped in. The membrane potential is reduced by the flow of Na H11001 and Ca 2H11001 into the cell through cGMP- gated cation channels in the plasma membrane of the outer segment. When rhodopsin absorbs light, it triggers degradation of cGMP (green dots) in the outer segment, causing closure of the cation channel. Without cation influx through this channel, the cell becomes hyper- polarized. This electrical signal is passed to the brain through the ranks of neurons shown in Figure 12–31. 8885d_c12_457 2/20/04 1:25 PM Page 457 mac76 mac76:385_reb: specialized for visual transduction, transducin shares many functional features with G s and G i . It can bind ei- ther GDP or GTP. In the dark, GDP is bound, all three subunits of the protein (T H9251 , T H9252 , and T H9253 ) remain together, and no signal is sent. When rhodopsin is excited by light, it interacts with transducin, catalyzing the replacement of bound GDP by GTP from the cytosol (Fig. 12–34, steps 1 and 2 ). Transducin then dissociates into T H9251 and T H9252H9253 , and the T H9251 -GTP carries the signal from the ex- cited receptor to the next element in the transduction pathway, cGMP phosphodiesterase (PDE); this enzyme converts cGMP to 5H11032-GMP (steps 3 and 4 ). Note that this is not the same cyclic nucleotide phosphodiesterase that hydrolyzes cAMP to terminate the H9252-adrenergic re- sponse. The cGMP-specific PDE is unique to the visual cells of the retina. PDE is an integral protein with its active site on the cytoplasmic side of the disk membrane. In the dark, a tightly bound inhibitory subunit very effectively sup- presses PDE activity. When T H9251 -GTP encounters PDE, the inhibitory subunit is released, and the enzyme’s ac- tivity immediately increases by several orders of mag- nitude. Each molecule of active PDE degrades many molecules of cGMP to the biologically inactive 5H11032-GMP, lowering [cGMP] in the outer segment within a fraction of a second. At the new, lower [cGMP], the cGMP-gated ion channels close, blocking reentry of Na H11001 and Ca 2H11001 into the outer segment and hyperpolarizing the mem- brane of the rod or cone cell (step 5 ). Through this process, the initial stimulus—a photon—changes the V m of the cell. Amplification of the Visual Signal Occurs in the Rod and Cone Cells Several steps in the visual-transduction process result in great amplification of the signal. Each excited rhodopsin molecule activates at least 500 molecules of transducin, each of which can activate a molecule of PDE. This phos- phodiesterase has a remarkably high turnover number, each activated molecule hydrolyzing 4,200 molecules of cGMP per second. The binding of cGMP to cGMP-gated ion channels is cooperative (at least three cGMP mole- cules must be bound to open one channel), and a rela- tively small change in [cGMP] therefore registers as a large change in ion conductance. The result of these amplifications is exquisite sensitivity to light. Absorption of a single photon closes 1,000 or more ion channels and changes the cell’s membrane potential by about 1 mV. The Visual Signal Is Quickly Terminated As your eyes move across this line, the images of the first words disappear rapidly—before you see the next series of words. In that short interval, a great deal of biochemistry has taken place. Very shortly after illumi- nation of the rod or cone cells stops, the photosensory system shuts off. The H9251 subunit of transducin (with bound GTP) has intrinsic GTPase activity. Within mil- liseconds after the decrease in light intensity, GTP is hy- drolyzed and T H9251 reassociates with T H9252H9253 . The inhibitory subunit of PDE, which had been bound to T H9251 -GTP, is re- leased and reassociates with PDE, strongly inhibiting that enzyme. To return [cGMP] to its “dark” level, the enzyme guanylyl cyclase converts GTP to cGMP (step 7 in Fig. 12–34) in a reaction that is inhibited by high [Ca 2H11001 ] (H11022100 nM). Calcium levels drop during illumina- tion, because the steady-state [Ca 2H11001 ] in the outer seg- ment is the result of outward pumping of Ca 2H11001 through the Na H11001 -Ca 2H11001 exchanger of the plasma membrane and inward movement of Ca 2H11001 through open cGMP-gated channels. In the dark, this produces a [Ca 2H11001 ] of about 500 nM—enough to inhibit cGMP synthesis. After brief illumination, Ca 2H11001 entry slows and [Ca 2H11001 ] declines (step Chapter 12 Biosignaling458 H9251 H9252 H9253 Cytosol Transducin Disk compartment Rhodopsin FIGURE 12–33 Likely structure of rhodopsin complexed with the G protein transducin. (PDB ID 1BAC) Rhodopsin (red) has seven trans- membrane helices embedded in the disk membranes of rod outer seg- ments and is oriented with its carboxyl terminus on the cytosolic side and its amino terminus inside the disk. The chromophore 11-cis reti- nal (blue), attached through a Schiff base linkage to Lys 256 of the sev- enth helix, lies near the center of the bilayer. (This location is similar to that of the epinephrine-binding site in the H9252-adrenergic receptor.) Several Ser and Thr residues near the carboxyl terminus are substrates for phosphorylations that are part of the desensitization mechanism for rhodopsin. Cytosolic loops that interact with the G protein trans- ducin are shown in orange; their exact positions are not yet known. The three subunits of transducin (green) are shown in their likely arrangement. Rhodopsin is palmitoylated at its carboxyl terminus, and both the H9251 and H9253 subunits of transducin have attached lipids (yellow) that assist in anchoring them to the membrane. 8885d_c12_458 2/20/04 1:26 PM Page 458 mac76 mac76:385_reb: 6 ). The inhibition of guanylyl cyclase by Ca 2H11001 is re- lieved, and the cyclase converts GTP to cGMP to return the system to its prestimulus state (step 7 ). Rhodopsin Is Desensitized by Phosphorylation Rhodopsin itself also undergoes changes in response to prolonged illumination. The conformational change in- duced by light absorption exposes several Thr and Ser residues in the carboxyl-terminal domain. These residues are quickly phosphorylated by rhodopsin ki- nase (step 8 in Fig. 12–34), which is functionally and structurally homologous to the H9252-adrenergic kinase (H9252ARK) that desensitizes the H9252-adrenergic receptor (Fig. 12–17). The Ca 2H11001 -binding protein recoverin in- hibits rhodopsin kinase at high [Ca 2H11001 ], but the inhibi- tion is relieved when [Ca 2H11001 ] drops after illumination, as described above. The phosphorylated carboxyl-terminal domain of rhodopsin is bound by the protein arrestin 1, preventing further interaction between activated rhodopsin and transducin. Arrestin 1 is a close homolog of arrestin 2 (H9252arr; Fig. 12–17). On a relatively long time scale (seconds to minutes), the all-trans-retinal of an excited rhodopsin molecule is removed and replaced by 11-cis-retinal, to produce rhodopsin that is ready for an- other round of excitation (step 9 in Fig. 12–34). Humans cannot synthesize retinal from simpler precursors and must obtain it in the diet in the form of vitamin A (see Fig. 10–21). Given the role of retinal in the process of vision, it is not surprising that dietary deficiency of vitamin A causes night blindness (poor vision at night or in dim light). ■ 12.7 Sensory Transduction in Vision, Olfaction, and Gustation 459 Ca 2+ Na + ,Ca 2+ 4 Na + Ca 2+ Light absorption converts 11-cis- retinal to all-trans-retinal, activating rhodopsin. 1 Activated rhodopsin catalyzes replacement of GDP by GTP on transducin (T), which then dissociates into T a -GTP and T bg . 2 T α -GTP activates cGMP phosphodiesterase (PDE) by binding and removing its inhibitory subunit (I). 3 Active PDE reduces [cGMP] to below the level needed to keep cation channels open. 4 Reduction of [Ca 2+ ] activates guanylyl cyclase (GC) and inhibits PDE; [cGMP] rises toward “dark” level, reopening cation channels and returning V m to prestimulus level. 7 Slowly, arrestin dissociates, rhodopsin is dephosphorylated, and all-trans-retinal is replaced with 11-cis-retinal. Rhodopsin is ready for another phototransduction cycle. 9 Rhodopsin kinase (RK) phosphorylates “bleached” rhodopsin; low [Ca 2+ ] and recoverin (Recov) stimulate this reaction. Arrestin (Arr) binds phosphorylated carboxyl terminus, inactivating rhodopsin. 8 Cation channels close, preventing influx of Na + and Ca 2+ ; membrane is hyperpolarized. This signal passes to the brain. Plasma membrane Disk membrane 5 Continued efflux of Ca 2+ through the Na + -Ca 2+ exchanger reduces cytosolic [Ca 2+ ]. 6 P P P P P RK Recov P GDP GTP cGMP ↓[Ca 2+ ] Recovery/Adaptation Excitation Rod I b g T Rh Rh Rh Arr Rh GC PDE PDE cGMP 5H11032-GMP cGMP cGMP cGMP cGMP I T a ? GTP T a ? GTP T a ? GDP GTP FIGURE 12–34 Molecular consequences of photon absorption by rhodopsin in the rod outer segment. The top half of the figure (steps 1 to 5 ) describes excitation; the bottom (steps 6 to 9 ), recovery and adaptation after illumination. 8885d_c12_459 2/20/04 1:26 PM Page 459 mac76 mac76:385_reb: Cone Cells Specialize in Color Vision Color vision in cone cells involves a path of sensory transduction essentially identical to that described above, but triggered by slightly different light receptors. Three types of cone cells are specialized to detect light from different regions of the spectrum, using three re- lated photoreceptor proteins (opsins). Each cone cell expresses only one kind of opsin, but each type is closely related to rhodopsin in size, amino acid sequence, and presumably three-dimensional structure. The differ- ences among the opsins, however, are great enough to place the chromophore, 11-cis-retinal, in three slightly different environments, with the result that the three photoreceptors have different absorption spectra (Fig. 12–35). We discriminate colors and hues by integrating the output from the three types of cone cells, each con- taining one of the three photoreceptors. Color blindness, such as the inability to distin- guish red from green, is a fairly common, genet- ically inherited trait in humans. The various types of color blindness result from different opsin mutations. One form is due to loss of the red photoreceptor; af- fected individuals are red H11546 dichromats (they see only two primary colors). Others lack the green pigment and are green H11002 dichromats. In some cases, the red and green photoreceptors are present but have a changed amino acid sequence that causes a change in their absorption spectra, resulting in abnormal color vision. Depending on which pigment is altered, such indivi- duals are red-anomalous trichromats or green- anomalous trichromats. Examination of the genes for the visual receptors has allowed the diagnosis of color blindness in a famous “patient” more than a century af- ter his death (Box 12–3)! ■ Vertebrate Olfaction and Gustation Use Mechanisms Similar to the Visual System The sensory cells used to detect odors and tastes have much in common with the rod and cone cells that de- tect light. Olfactory neurons have a number of long thin cilia extending from one end of the cell into a mucous layer that overlays the cell. These cilia present a large surface area for interaction with olfactory signals. The receptors for olfactory stimuli are ciliary membrane pro- teins with the familiar serpentine structure of seven transmembrane H9251 helices. The olfactory signal can be any one of the many volatile compounds for which there are specific receptor proteins. Our ability to discrimi- nate odors stems from hundreds of different olfactory receptors in the tongue and nasal passages and from the brain’s ability to integrate input from different types of olfactory receptors to recognize a “hybrid” pattern, ex- tending our range of discrimination far beyond the num- ber of receptors. The olfactory stimulus arrives at the sensory cells by diffusion through the air. In the mucous layer over- laying the olfactory neurons, the odorant molecule binds directly to an olfactory receptor or to a specific binding protein that carries the odorant to a receptor (Fig. 12–36). Interaction between odorant and receptor triggers a change in receptor conformation that results in the replacement of bound GDP by GTP on a G pro- tein, G olf , analogous to transducin and to G s of the H9252- adrenergic system. The activated G olf then activates adenylyl cyclase of the ciliary membrane, which syn- thesizes cAMP from ATP, raising the local [cAMP]. The cAMP-gated Na H11001 and Ca 2H11001 channels of the ciliary mem- brane open, and the influx of Na H11001 and Ca 2H11001 produces a small depolarization called the receptor potential. If a sufficient number of odorant molecules encounter re- ceptors, the receptor potential is strong enough to cause the neuron to fire an action potential. This is relayed to the brain in several stages and registers as a specific smell. All these events occur within 100 to 200 ms. Some olfactory neurons may use a second trans- duction mechanism. They have receptors coupled through G proteins to PLC rather than to adenylyl cy- clase. Signal reception in these cells triggers production of IP 3 (Fig. 12–19), which opens IP 3 -gated Ca 2H11001 chan- nels in the ciliary membrane. Influx of Ca 2H11001 then depo- larizes the ciliary membrane and generates a receptor potential or regulates Ca 2H11001 -dependent enzymes in the olfactory pathway. In either type of olfactory neuron, when the stimu- lus is no longer present, the transducing machinery shuts Chapter 12 Biosignaling460 Blue pigment Rhodopsin Red pigment 100 90 80 70 60 50 40 30 20 10 0 Relative absorbance 400 450 500 550 600 650 Green pigment Wavelength (nm) FIGURE 12–35 Absorption spectra of purified rhodopsin and the red, green, and blue receptors of cone cells. The spectra, obtained from individual cone cells isolated from cadavers, peak at about 420, 530, and 560 nm, and the maximum absorption for rhodopsin is at about 500 nm. For reference, the visible spectrum for humans is about 380 to 750 nm. 8885d_c12_460 2/20/04 1:26 PM Page 460 mac76 mac76:385_reb: 12.7 Sensory Transduction in Vision, Olfaction, and Gustation 461 Ciliary membrane Mucous layer Air cAMPATP GTP GDP Olfactory neuron Dendrite Axon Cilia AC Ca 2+ Cl – G olf b g O O BP O Odorant (O) arrives at the mucous layer and binds directly to an olfactory receptor (OR) or to a binding protein (BP) that carries it to the OR. 1 Activated OR catalyzes GDP-GTP exchange on a G protein (G olf ), causing its dissociation into a and bg. 2 Ca 2+ reduces the affinity of the cation channel for cAMP, lowering the sensitivity of the system to odorant. 6 G olf hydrolyzes GTP to GDP, shutting itself off. PDE hydrolyzes cAMP. Receptor kinase phosphorylates OR, inactivating it. Odorant is removed by metabolism. 7 Ca 2+ -gated chloride channels open. Efflux of Cl – depolarizes the cell, triggering an electrical signal to the brain. 5 G a -GTP activates adenylyl cyclase, which catalyzes cAMP synthesis, raising [cAMP]. 3 cAMP-gated cation channels open. Ca 2+ enters, raising internal [Ca 2+ ]. 4 OR O a GDP a GTP FIGURE 12–36 Molecular events of olfaction. These interactions oc- cur in the cilia of olfactory receptor cells. FIGURE 7 (a)H11021?/AuH11022 (b)H11021?/AuH11022 BOX 12–3 BIOCHEMISTRY IN MEDICINE Color Blindness: John Dalton’s Experiment from the Grave The chemist John Dalton (of atomic theory fame) was color-blind. He thought it probable that the vitreous humor of his eyes (the fluid that fills the eyeball be- hind the lens) was tinted blue, unlike the colorless fluid of normal eyes. He proposed that after his death, his eyes should be dissected and the color of the vit- reous humor determined. His wish was honored. The day after Dalton’s death in July 1844, Joseph Ransome dissected his eyes and found the vitreous humor to be perfectly colorless. Ransome, like many scientists, was reluctant to throw samples away. He placed Dalton’s eyes in a jar of preservative (Fig. 1), where they stayed for a century and a half. Then, in the mid-1990s, molecular biologists in England took small samples of Dalton’s retinas and ex- tracted DNA. Using the known gene sequences for the opsins of the red and green photopigments, they am- plified the relevant sequences (using techniques de- scribed in Chapter 9) and determined that Dalton had the opsin gene for the red photopigment but lacked the opsin gene for the green photopigment. Dalton was a green H11002 dichromat. So, 150 years after his death, the experiment Dalton started—by hypothesizing about the cause of his color blindness—was finally finished. FIGURE 1 Dalton’s eyes. 8885d_c12_461 2/20/04 1:26 PM Page 461 mac76 mac76:385_reb: itself off in several ways. A cAMP phosphodiesterase returns [cAMP] to the prestimulus level. G olf hydrolyzes its bound GTP to GDP, thereby inactivating itself. Phos- phorylation of the receptor by a specific kinase prevents its interaction with G olf , by a mechanism analogous to that used to desensitize the H9252-adrenergic receptor and rhodopsin. And lastly, some odorants are enzymatically destroyed by oxidases. The sense of taste in vertebrates reflects the activ- ity of gustatory neurons clustered in taste buds on the surface of the tongue. In these sensory neurons, ser- pentine receptors are coupled to the heterotrimeric G protein gustducin (very similar to the transducin of rod and cone cells). Sweet-tasting molecules are those that bind receptors in “sweet” taste buds. When the mole- cule (tastant) binds, gustducin is activated by replace- ment of bound GDP with GTP and then stimulates cAMP production by adenylyl cyclase. The resulting elevation of [cAMP] activates PKA, which phosphorylates K H11001 channels in the plasma membrane, causing them to close. Reduced efflux of K H11001 depolarizes the cell (Fig. 12–37). Other taste buds specialize in detecting bitter, sour, or salty tastants, using various combinations of sec- ond messengers and ion channels in the transduction mechanisms. G Protein–Coupled Serpentine Receptor Systems Share Several Features We have now looked at four systems (hormone signal- ing, vision, olfaction, and gustation) in which membrane receptors are coupled to second messenger–generating enzymes through G proteins. It is clear that signaling mechanisms arose early in evolution; serpentine recep- tors, heterotrimeric G proteins, and adenylyl cyclase are found in virtually all eukaryotic organisms. Even the common brewer’s yeast Saccharomyces uses serpentine receptors and G proteins to detect the opposite mating type. Overall patterns have been conserved, and the in- troduction of variety has given modern organisms the ability to respond to a wide range of stimuli (Table 12–8). Of the 35,000 or so genes in the human genome, as many as 1,000 encode serpentine receptors, including hun- dreds for olfactory stimuli and a number of “orphan re- ceptors” for which the natural ligand is not yet known. All well-studied transducing systems that act through heterotrimeric G proteins share certain com- mon features (Fig. 12–38). The receptors have seven transmembrane segments, a domain (generally the loop between transmembrane helices 6 and 7) that interacts with a G protein, and a carboxyl-terminal cytoplasmic domain that undergoes reversible phosphorylation on several Ser or Thr residues. The ligand-binding site (or, in the case of light reception, the light receptor) is buried deep in the membrane and includes residues from several of the transmembrane segments. Ligand binding (or light) induces a conformational change in the receptor, exposing a domain that can interact with a G protein. Heterotrimeric G proteins activate or in- hibit effector enzymes (adenylyl cyclase, PDE, or PLC), which change the concentration of a second messenger (cAMP, cGMP, IP 3 , or Ca 2H11001 ). In the hormone-detecting Chapter 12 Biosignaling462 Acetylcholine (muscarinic) Adenosine Angiotensin ATP (extracellular) Bradykinin Calcitonin Cannabinoids Catecholamines Cholecystokinin Corticotropin-releasing factor (CRF) Cyclic AMP (Dictyostelium discoideum) Dopamine Follicle-stimulating hormone (FSH) H9253-Aminobutyric acid (GABA) Glucagon Glutamate Growth hormone–releasing hormone (GHRH) Histamine Leukotrienes Light Luteinizing hormone (LH) Melatonin Odorants Opioids Oxytocin Platelet-activating factor Prostaglandins Secretin Serotonin Somatostatin Tastants Thyrotropin Thyrotropin-releasing hormone (TRH) Vasoactive intestinal peptide Vasopressin Yeast mating factors Some Signals Transduced by G Protein–Coupled Serpentine Receptors TABLE 12–8 8885d_c12_462 2/20/04 1:26 PM Page 462 mac76 mac76:385_reb: systems, the final output is an activated protein kinase that regulates some cellular process by phosphorylating a protein critical to that process. In sensory neurons, the output is a change in membrane potential and a con- sequent electrical signal that passes to another neuron in the pathway connecting the sensory cell to the brain. All these systems self-inactivate. Bound GTP is con- verted to GDP by the intrinsic GTPase activity of G pro- teins, often augmented by GTPase-activating proteins (GAPs) or RGS proteins (regulators of G-protein sig- naling). In some cases, the effector enzymes that are the targets of G protein modulation also serve as GAPs. 12.7 Sensory Transduction in Vision, Olfaction, and Gustation 463 Basolateral membrane Apical membrane cAMPATP GTP Olfactory neuron AC K H11545 G gust H9252 H9253 SR Sweet-tasting molecule (S) binds to sweet-taste receptor (SR), activating the G protein gustducin (G gust ). 1 Gustducin subunit activates adenylyl cyclase (AC) of the apical membrane, raising [cAMP]. H9251 Taste cell 2 PKA, activated by cAMP, phosphorylates a K H11545 channel in the basolateral membrane, causing it to close. The reduced efflux of K H11545 depolarizes the cell. 3 P S GTPGDP GDP PKA H9251 H9251 Vasopressin Epinephrine Light Odorants Sweet tastant ↓[cAMP] ↓ ↑[cAMP] ↑ P ↓[cGMP] ↑[IP 3 ] ↑[cAMP] ↑[cAMP] ↓P Ca 2H11545 ,Na H11545 ↑P Ca 2H11545 ↑P Ca 2H11545 ,Na H11545 ↓P K H11545 VR G i AC PKA -AR G s AC PKA ↑ PKA Rh T PDE OR 1 G olf PLC OR 2 G olf AC SR G gust AC H9252 FIGURE 12–38 Common features of signaling systems that detect hormones, light, smells, and tastes. Serpentine receptors provide sig- nal specificity, and their interaction with G proteins provides signal amplification. Heterotrimeric G proteins activate effector enzymes: adenylyl cyclase (AC), phospholipase C (PLC), and phosphodiesterases (PDE) that degrade cAMP or cGMP. Changes in concentration of the second messengers (cAMP, cGMP, IP 3 ) result in alterations of enzy- matic activities by phosphorylation or alterations in the permeability (P) of surface membranes to Ca 2H11001 , Na H11001 , and K H11001 . The resulting depo- larization or hyperpolarization of the sensory cell (the signal) is passed through relay neurons to sensory centers in the brain. In the best- studied cases, desensitization includes phosphorylation of the recep- tor and binding of a protein (arrestin) that interrupts receptor–G pro- tein interactions. VR is the vasopressin receptor; other receptor and G protein abbreviations are as used in earlier illustrations. FIGURE 12–37 Transduction mechanism for sweet tastants. 8885d_c12_463 2/20/04 1:27 PM Page 463 mac76 mac76:385_reb: Disruption of G-Protein Signaling Causes Disease Biochemical studies of signal transductions have led to an improved understanding of the patho- logical effects of toxins produced by the bacteria that cause cholera and pertussis (whooping cough). Both toxins are enzymes that interfere with normal signal transductions in the host animal. Cholera toxin, se- creted by Vibrio cholerae found in contaminated drink- ing water, catalyzes the transfer of ADP-ribose from NAD H11001 to the H9251 subunit of G s (Fig. 12–39), blocking its GTPase activity and thereby rendering G s permanently activated. This results in continuous activation of the adenylyl cyclase of intestinal epithelial cells and chron- ically high [cAMP], which triggers constant secretion of Cl H11002 , HCO 3 H11002 , and water into the intestinal lumen. The re- sulting dehydration and electrolyte loss are the major pathologies in cholera. The pertussis toxin, produced by Bordetella pertussis, catalyzes ADP-ribosylation of G i , preventing displacement of GDP by GTP and block- ing inhibition of adenylyl cyclase by G i . ■ SUMMARY 12.7 Sensory Transduction in Vision, Olfaction, and Gustation ■ Vision, olfaction, and gustation in vertebrates employ serpentine receptors, which act through heterotrimeric G proteins to change the V m of the sensory neuron. ■ In rod and cone cells of the retina, light activates rhodopsin, which stimulates replacement of GDP by GTP on the G protein transducin. The freed H9251 subunit of transducin activates cGMP phosphodiesterase, which lowers [cGMP] and thus closes cGMP-dependent ion channels in the outer segment of the neuron. The resulting hyperpolarization of the rod or cone cell carries the signal to the next neuron in the pathway, and eventually to the brain. ■ In olfactory neurons, olfactory stimuli, acting through serpentine receptors and G proteins, trigger either an increase in [cAMP] (by activating adenylyl cyclase) or an increase in [Ca 2H11001 ] (by activating PLC). These second messengers affect ion channels and thus the V m . ■ Gustatory neurons have serpentine receptors that respond to tastants by altering [cAMP], which in turn changes V m by gating ion channels. ■ There is a high degree of conservation of signaling proteins and transduction mechanisms across species. Chapter 12 Biosignaling464 H9252 H9253 H9251 G s H9252 H9253 H9251 G s CH 2 O OH P O H11002 H N H HH NH 2 O O H OO C O 2 P O H11002 O O Rib Adenine H11001 O NH 2 C NAD H11001 H11001 cholera toxin CH 2 OH P O H11002 H H HH O O H OO O P O H11002 O O Rib Adenine N Arg NH ADP-ribose Normal G s : GTPase activity terminates the signal from receptor to adenylyl cyclase. ADP-ribosylated G s : GTPase activity is inactivated; G s constantly activates adenylyl cyclase. NHArg FIGURE 12–39 Toxins produced by bacteria that cause cholera and whooping cough (pertussis). These toxins are enzymes that catalyze transfer of the ADP-ribose moiety of NAD H11001 to an Arg residue (cholera toxin) or a Cys residue (pertussis toxin) of G proteins: G s in the case of cholera (as shown here) and G I in whooping cough. The G proteins thus modified fail to respond to normal hormonal stimuli. The pathol- ogy of both diseases results from defective regulation of adenylyl cy- clase and overproduction of cAMP. 8885d_c12_464 2/20/04 1:27 PM Page 464 mac76 mac76:385_reb: 12.8 Regulation of Transcription by Steroid Hormones The large group of steroid, retinoic acid (retinoid), and thyroid hormones exert at least part of their effects by a mechanism fundamentally different from that of other hormones: they act in the nucleus to alter gene expres- sion. We therefore discuss their mode of action in detail in Chapter 28, along with other mechanisms for regu- lating gene expression. Here we give a brief overview. Steroid hormones (estrogen, progesterone, and cor- tisol, for example), too hydrophobic to dissolve readily in the blood, are carried on specific carrier proteins from their point of release to their target tissues. In target cells, these hormones pass through the plasma mem- branes by simple diffusion and bind to specific receptor proteins in the nucleus (Fig. 12–40). Hormone binding triggers changes in the conformation of the receptor proteins so that they become capable of interacting with specific regulatory sequences in DNA called hormone response elements (HREs), thus altering gene ex- pression (see Fig. 28–31). The bound receptor-hormone complex can either enhance or suppress the expression of specific genes adjacent to HREs. Hours or days are required for these regulators to have their full effect— the time required for the changes in RNA synthesis and subsequent protein synthesis to become evident in al- tered metabolism. The specificity of the steroid-receptor interac- tion is exploited in the use of the drug tamox- ifen to treat breast cancer. In some types of breast can- cer, division of the cancerous cells depends on the continued presence of the hormone estrogen. Tamox- ifen competes with estrogen for binding to the estrogen receptor, but the tamoxifen-receptor complex has little or no effect on gene expression; tamoxifen is an antag- onist of estrogen. Consequently, tamoxifen administered after surgery or during chemotherapy for hormone- dependent breast cancer slows or stops the growth of remaining cancerous cells. 12.8 Regulation of Transcription by Steroid Hormones 465 1 1 Hormone (H), carried to the target tissue on serum binding proteins, diffuses across the plasma membrane and binds to its specific receptor protein (Rec) in the nucleus. 2 3 3 Binding regulates transcription of the adjacent gene(s), increasing or decreasing the rate of mRNA formation. 4 4 Altered levels of the hormone- regulated gene product produce the cellular response to the hormone. Plasma membrane Nucleus Rec RNA polymerase HRE Gene transcription mRNA translation on ribosomes New protein Altered cell function H Serum binding protein with bound hormone 2 Hormone binding changes the conformation of Rec; it forms homo- or heterodimers with other hormone- receptor complexes and binds to specific regulatory regions called hormone response elements (HREs) in the DNA adjacent to specific genes. FIGURE 12–40 General mechanism by which steroid and thyroid hormones, retinoids, and vitamin D regulate gene expression. The de- tails of transcription and protein synthesis are discussed in Chapters 26 and 27. At least some steroids also act through plasma membrane receptors by a completely different mechanism. O CH 3 N CH 3 CH 3 Tamoxifen 8885d_c12_465 2/20/04 1:27 PM Page 465 mac76 mac76:385_reb: Another steroid analog, the drug RU486, is used to ter- minate early (preimplantation) pregnancies. An antag- onist of the hormone progesterone, RU486 binds to the progesterone receptor and blocks hormone actions es- sential to implantation of the fertilized ovum in the uterus. ■ The classic mechanism for steroid hormone action through nuclear receptors does not explain certain ef- fects of steroids that are too fast to be the result of al- tered protein synthesis. For example, the estrogen- mediated dilation of blood vessels is known to be independent of gene transcription or protein synthesis, as is the steroid-induced decrease in cellular [cAMP]. Another transduction mechanism is probably responsi- ble for some of these effects. A plasma membrane pro- tein predicted to have seven transmembrane helical seg- ments binds progesterone with very high affinity and mediates the inhibition of adenylyl cyclase by that hor- mone, accounting for the decrease in [cAMP]. A second nonclassical mechanism involves the rapid activation of the MAPK cascade by progesterone, acting through the soluble progesterone receptor. This is the same recep- tor that, in the nucleus, causes the much slower changes in gene expression that constitute the classic mecha- nism of progesterone action. How the MAPK cascade is activated is not yet clear. SUMMARY 12.8 Regulation of Transcription by Steroid Hormones ■ Steroid hormones enter cells and bind to specific receptor proteins. ■ The hormone-receptor complex binds specific regions of DNA, the hormone response elements, and regulates the expression of nearby genes by interacting with transcription factors. ■ Two other, faster-acting mechanisms produce some of the effects of steroids. Progesterone triggers a rapid drop in [cAMP], mediated by a plasma membrane receptor, and binding of progesterone to the classic soluble steroid receptor activates a MAPK cascade. 12.9 Regulation of the Cell Cycle by Protein Kinases One of the most dramatic roles for protein phosphory- lation is the regulation of the eukaryotic cell cycle. Dur- ing embryonic growth and later development, cell divi- sion occurs in virtually every tissue. In the adult organism most tissues become quiescent. A cell’s “deci- sion” to divide or not is of crucial importance to the or- ganism. When the regulatory mechanisms that limit cell division are defective and cells undergo unregulated division, the result is catastrophic—cancer. Proper cell division requires a precisely ordered sequence of bio- chemical events that assures every daughter cell a full complement of the molecules required for life. Investi- gations into the control of cell division in diverse eu- karyotic cells have revealed universal regulatory mech- anisms. Protein kinases and protein phosphorylation are central to the timing mechanism that determines entry into cell division and ensures orderly passage through these events. The Cell Cycle Has Four Stages Cell division in eukaryotes occurs in four well-defined stages (Fig. 12–41). In the S (synthesis) phase, the DNA is replicated to produce copies for both daughter Chapter 12 Biosignaling466 O N CH 3 CH 3 RU486 (mifepristone) OH CC CH 3 G1 6–12 h S 6–8 h G2 3–4 h M 1 h G1 Phase RNA and protein synthesis. No DNA synthesis. Restriction point A cell that passes this point is committed to pass into S phase. Reentry point A cell returning from G0 enters at early G1 phase. G0 Phase Terminally differentiated cells withdraw from cell cycle indefinitely. M Phase Mitosis (nuclear division) and cytokinesis (cell division) yield two daughter cells. G2 Phase No DNA synthesis. RNA and protein synthesis continue. S Phase DNA synthesis doubles the amount of DNA in the cell. RNA and protein also synthesized. G0 FIGURE 12–41 Eukaryotic cell cycle. The durations (in hours) of the four stages vary, but those shown are typical. 8885d_c12_466 2/20/04 1:27 PM Page 466 mac76 mac76:385_reb: cells. In the G2 phase (G indicates the gap between divisions), new proteins are synthesized and the cell approximately doubles in size. In the M phase (mitosis), the maternal nuclear envelope breaks down, matching chromosomes are pulled to opposite poles of the cell, each set of daughter chromosomes is surrounded by a newly formed nuclear envelope, and cytokinesis pinches the cell in half, producing two daughter cells. In em- bryonic or rapidly proliferating tissue, each daughter cell divides again, but only after a waiting period (G1). In cultured animal cells the entire process takes about 24 hours. After passing through mitosis and into G1, a cell ei- ther continues through another division or ceases to di- vide, entering a quiescent phase (G0) that may last hours, days, or the lifetime of the cell. When a cell in G0 begins to divide again, it reenters the division cycle through the G1 phase. Differentiated cells such as he- patocytes or adipocytes have acquired their specialized function and form; they remain in the G0 phase. Levels of Cyclin-Dependent Protein Kinases Oscillate The timing of the cell cycle is controlled by a family of protein kinases with activities that change in response to cellular signals. By phosphorylating specific proteins at precisely timed intervals, these protein kinases or- chestrate the metabolic activities of the cell to produce orderly cell division. The kinases are heterodimers with a regulatory subunit, cyclin, and a catalytic subunit, cyclin-dependent protein kinase (CDK). In the ab- sence of cyclin, the catalytic subunit is virtually inac- tive. When cyclin binds, the catalytic site opens up, a residue essential to catalysis becomes accessible (Fig. 12–42), and the activity of the catalytic subunit in- creases 10,000-fold. Animal cells have at least ten dif- ferent cyclins (designated A, B, and so forth) and at least eight cyclin-dependent kinases (CDK1 through CDK8), which act in various combinations at specific points in the cell cycle. Plants also use a family of CDKs to regulate their cell division. 12.9 Regulation of the Cell Cycle by Protein Kinases 467 (b) (a) (c) FIGURE 12–42 Activation of cyclin-dependent protein kinases (CDKs) by cyclin and phosphorylation. CDKs, a family of related enzymes, are active only when associated with cyclins, another protein family. The crystal structure of CDK2 with and without cyclin reveals the basis for this activation. (a) Without cyclin (PDB ID 1HCK), CDK2 folds so that one segment, the T loop (red), obstructs the binding site for protein substrates and thus inhibits protein kinase activity. The binding site for ATP (blue) is also near the T loop. (b) When cyclin binds (PDB ID 1FIN), it forces conformational changes that move the T loop away from the active site and reorient an amino-terminal helix (green), bringing a residue critical to catalysis (Glu 51 ) into the active site. (c) Phosphorylation of a Thr residue (dark orange space-filling structure) in the T loop produces a negatively charged residue that is stabilized by interaction with three Arg residues (red ball-and-stick struc- tures), holding CDK in its active conformation (PDB ID 1JST). 8885d_c12_467 2/23/04 9:12 AM Page 467 mac76 mac76: In a population of animal cells undergoing synchro- nous division, some CDK activities show striking oscil- lations (Fig. 12–43). These oscillations are the result of four mechanisms for regulating CDK activity: phosphor- ylation or dephosphorylation of the CDK, controlled degradation of the cyclin subunit, periodic synthesis of CDKs and cyclins, and the action of specific CDK- inhibiting proteins. Regulation of CDKs by Phosphorylation The activity of a CDK is strikingly affected by phosphorylation and de- phosphorylation of two critical residues in the protein (Fig. 12–44a). Phosphorylation of Tyr 15 near the amino terminus renders CDK2 inactive; the P –Tyr residue is in the ATP-binding site of the kinase, and the negatively charged phosphate group blocks the entry of ATP. A specific phosphatase dephosphorylates this P –Tyr residue, permitting the binding of ATP. Phosphorylation Chapter 12 Biosignaling468 Kinase activity Time G1 S G2 M Cyclin E–CDK2 Cyclin A–CDK2 Cyclin B–CDK1 G1 FIGURE 12–43 Variations in the activities of specific CDKs during the cell cycle in animals. Cyclin E–CDK2 activity peaks near the G1 phase–S phase boundary, when the active enzyme triggers synthesis of enzymes required for DNA synthesis (see Fig. 12–46). Cyclin A–CDK2 activity rises during the S and G2 phases, then drops sharply in the M phase, as cyclin B–CDK1 peaks. U Cyclin U U U U Cyclin Tyr No cyclin present; CDK is inactive. 2 Cyclin synthesis leads to its accumulation. Cyclin-CDK complex forms, but phosphorylation on Tyr 15 blocks ATP-binding site; still inactive. 3 Phosphorylation of Thr 160 in T loop and removal of Tyr 15 phosphoryl group activates cyclin-CDK manyfold. 4 1 Cyclin is degraded by proteasome, leaving CDK inactive. 8 CDK phosphorylates phosphatase, which activates more CDK. 5 CDK phosphorylates DBRP, activating it. 6 DBRP triggers addition of ubiquitin molecules to cyclin by ubiquitin ligase. 7 P i CDKCDK CDK Tyr Thr Thr Cyclin CDK PhosphatasePhosphatase DBRPDBRP P P P P P P (a) (b) FIGURE 12–44 Regulation of CDK by phosphorylation and prote- olysis. (a) The cyclin-dependent protein kinase activated at the time of mitosis (the M phase CDK) has a “T loop” that can fold into the substrate-binding site. When Thr 160 in the T loop is phosphorylated, the loop moves out of the substrate-binding site, activating the CDK manyfold. (b) The active cyclin-CDK complex triggers its own inac- tivation by phosphorylation of DBRP (destruction box recognizing protein). DBRP and ubiquitin ligase then attach several molecules of ubiquitin (U) to cyclin, targeting it for destruction by proteasomes, proteolytic enzyme complexes. 8885d_c12_468 2/20/04 1:28 PM Page 468 mac76 mac76:385_reb: of Thr 160 in the “T loop” of CDK, catalyzed by the CDK- activating kinase, forces the T loop out of the substrate- binding cleft, permitting substrate binding and catalytic activity. One circumstance that triggers this control mecha- nism is the presence of single-strand breaks in DNA, which leads to arrest of the cell cycle in G2. A specific protein kinase (called Rad3 in yeast), which is activated by single-strand breaks, triggers a cascade leading to the inactivation of the phosphatase that dephosphorylates Tyr 15 of CDK. The CDK remains inactive and the cell is arrested in G2. The cell will not divide until the DNA is repaired and the effects of the cascade are reversed. Controlled Degradation of Cyclin Highly specific and pre- cisely timed proteolytic breakdown of mitotic cyclins regulates CDK activity throughout the cell cycle. Progress through mitosis requires first the activation then the destruction of cyclins A and B, which activate the catalytic subunit of the M-phase CDK. These cyclins contain near their amino terminus the sequence Arg–Thr–Ala–Leu–Gly–Asp–Ile–Gly–Asn, the “destruc- tion box,” which targets them for degradation. (This us- age of “box” derives from the common practice, in dia- gramming the sequence of a nucleic acid or protein, of enclosing within a box a short sequence of nucleotide or amino acid residues with some specific function. It does not imply any three-dimensional structure.) The protein DBRP (destruction box recognizing protein) recognizes this sequence and initiates the process of cy- clin degradation by bringing together the cyclin and an- other protein, ubiquitin. Cyclin and activated ubiqui- tin are covalently joined by the enzyme ubiquitin ligase (Fig. 12–44b). Several more ubiquitin molecules are then appended, providing the signal for a proteolytic en- zyme complex, or proteasome, to degrade cyclin. What controls the timing of cyclin breakdown? A feedback loop occurs in the overall process shown in Figure 12–44. Increased CDK activity activates cyclin proteolysis. Newly synthesized cyclin associates with and activates CDK, which phosphorylates and activates DBRP. Active DBRP then causes proteolysis of cyclin. Lowered [cyclin] causes a decline in CDK activity, and the activity of DBRP also drops through slow, constant dephosphorylation and inactivation by a DBRP phos- phatase. The cyclin level is ultimately restored by syn- thesis of new cyclin molecules. The role of ubiquitin and proteasomes is not limited to the regulation of cyclin; as we shall see in Chapter 27, both also take part in the turnover of cellular proteins, a process fundamental to cellular housekeeping. Regulated Synthesis of CDKs and Cyclins The third mech- anism for changing CDK activity is regulation of the rate of synthesis of cyclin or CDK or both. For example, cy- clin D, cyclin E, CDK2, and CDK4 are synthesized only when a specific transcription factor, E2F, is present in the nucleus to activate transcription of their genes. Syn- thesis of E2F is in turn regulated by extracellular sig- nals such as growth factors and cytokines (inducers of cell division), compounds found to be essential for the division of mammalian cells in culture. These growth factors induce the synthesis of specific nuclear tran- scription factors essential to the production of the enzymes of DNA synthesis. Growth factors trigger phos- phorylation of the nuclear proteins Jun and Fos, tran- scription factors that promote the synthesis of a variety of gene products, including cyclins, CDKs, and E2F. In turn, E2F controls production of several enzymes es- sential for the synthesis of deoxynucleotides and DNA, enabling cells to enter the S phase (Fig. 12–45). Inhibition of CDKs Finally, specific protein inhibitors bind to and inactivate specific CDKs. One such protein is p21, which we discuss below. These four control mechanisms modulate the ac- tivity of specific CDKs that, in turn, control whether a cell will divide, differentiate, become permanently qui- escent, or begin a new cycle of division after a period of quiescence. The details of cell cycle regulation, such as the number of different cyclins and kinases and the 12.9 Regulation of the Cell Cycle by Protein Kinases 469 Growth factors, cytokines Phosphorylation of Jun and Fos in nucleus transcriptional regulation Passage from G1 to S phase MAPK cascade Transcription factor E2F transcriptional regulation Cyclins, CDKs Enzymes for DNA synthesis FIGURE 12–45 Regulation of cell division by growth factors. The path from growth factors to cell division leads through the enzyme cascade that activates MAPK; phosphorylation of the nuclear tran- scription factors Jun and Fos; and the activity of the transcription fac- tor E2F, which promotes synthesis of several enzymes essential for DNA synthesis. 8885d_c12_469 2/20/04 1:28 PM Page 469 mac76 mac76:385_reb: combinations in which they act, differ from species to species, but the basic mechanism has been conserved in the evolution of all eukaryotic cells. CDKs Regulate Cell Division by Phosphorylating Critical Proteins We have examined how cells maintain close control of CDK activity, but how does the activity of CDK control the cell cycle? The list of target proteins that CDKs are known to act upon continues to grow, and much remains to be learned. But we can see a general pattern behind CDK regulation by inspecting the effect of CDKs on the structures of laminin and myosin and on the activity of retinoblastoma protein. The structure of the nuclear envelope is maintained in part by highly organized meshworks of intermediate filaments composed of the protein laminin. Breakdown of the nuclear envelope before segregation of the sister chromatids in mitosis is partly due to the phosphoryla- tion of laminin by a CDK, which causes laminin filaments to depolymerize. A second kinase target is the ATP-driven actin- myosin contractile machinery that pinches a dividing cell into two equal parts during cytokinesis. After the division, CDK phosphorylates a small regulatory subunit of myosin, causing dissociation of myosin from actin fil- aments and inactivating the contractile machinery. Sub- sequent dephosphorylation allows reassembly of the contractile apparatus for the next round of cytokinesis. A third and very important CDK substrate is the retinoblastoma protein, pRb; when DNA damage is detected, this protein participates in a mechanism that arrests cell division in G1 (Fig. 12–46). Named for the retinal tumor cell line in which it was discovered, pRb functions in most, perhaps all, cell types to regulate cell division in response to a variety of stimuli. Unphosphor- ylated pRb binds the transcription factor E2F; while bound to pRb, E2F cannot promote transcription of a group of genes necessary for DNA synthesis (the genes for DNA polymerase H9251, ribonucleotide reductase, and other proteins; Chapter 25). In this state, the cell cycle cannot proceed from the G1 to the S phase, the step that commits a cell to mitosis and cell division. The pRb- E2F blocking mechanism is relieved when pRb is phos- phorylated by cyclin E–CDK2, which occurs in response to a signal for cell division to proceed. When the protein kinases ATM and ATR detect dam- age to DNA, such as a single-strand break, they activate p53 to serve as a transcription factor that stimulates the synthesis of the protein p21 (Fig. 12–46). This protein inhibits the protein kinase activity of cyclin E–CDK2. In the presence of p21, pRb remains unphosphorylated and bound to E2F, blocking the activity of this transcription factor, and the cell cycle is arrested in G1. This gives the cell time to repair its DNA before entering the S phase, thereby avoiding the potentially disastrous trans- fer of a defective genome to one or both daughter cells. SUMMARY 12.9 Regulation of the Cell Cycle by Protein Kinases ■ Progression through the cell cycle is regulated by the cyclin-dependent protein kinases (CDKs), which act at specific points in the cycle, phosphorylating key proteins and modulating their activities. The catalytic subunit of CDKs is inactive unless associated with the regulatory cyclin subunit. ■ The activity of a cyclin-CDK complex changes during the cell cycle through differential synthesis of CDKs, specific degradation of cyclin, phosphorylation and dephosphorylation of critical residues in CDKs, and binding of inhibitory proteins to specific cyclin-CDKs. Chapter 12 Biosignaling470 Cell division blocked by p53 Cell division occurs normally CDK2 Inactive CDK2 Active Cyclin E Active p53 DNA damage ↑[p21] p21 p21 pRb P pRb pRb E2F Inactive E2F Enzymes for DNA synthesis transcriptional regulation transcriptional regulation Passage from G1 to S Active Cyclin E FIGURE 12–46 Regulation of passage from G1 to S by phosphory- lation of pRb. When the retinoblastoma protein, pRb, is phosphory- lated, it cannot bind and inactivate EF2, a transcription factor that pro- motes synthesis of enzymes essential to DNA synthesis. If the regulatory protein p53 is activated by ATM and ATR, protein kinases that detect damaged DNA, it stimulates the synthesis of p21, which can bind to and inhibit cyclin E–CDK2 and thus prevent phosphory- lation of pRb. Unphosphorylated pRb binds and inactivates E2F, block- ing passage from G1 to S until the DNA has been repaired. 8885d_c12_470 2/20/04 1:28 PM Page 470 mac76 mac76:385_reb: 12.10 Oncogenes, Tumor Suppressor Genes, and Programmed Cell Death Tumors and cancer are the result of uncontrolled cell division. Normally, cell division is regulated by a family of extracellular growth factors, proteins that cause rest- ing cells to divide and, in some cases, differentiate. Defects in the synthesis, regulation, or recognition of growth factors can lead to cancer. Oncogenes Are Mutant Forms of the Genes for Proteins That Regulate the Cell Cycle Oncogenes were originally discovered in tumor-causing viruses, then later found to be closely similar to or de- rived from genes in the animal host cells, proto- oncogenes, which encode growth-regulating proteins. During viral infections, the DNA sequence of a proto- oncogene is sometimes copied by the virus and incor- porated into its genome (Fig. 12–47). At some point during the viral infection cycle, the gene can become defective by truncation or mutation. When this viral oncogene is expressed in its host cell during a subse- quent infection, the abnormal protein product interferes with normal regulation of cell growth, sometimes re- sulting in a tumor. Proto-oncogenes can become oncogenes without a viral intermediary. Chromosomal rearrangements, chem- ical agents, and radiation are among the factors that can cause oncogenic mutations. The mutations that produce oncogenes are genetically dominant; if either of a pair of chromosomes contains a defective gene, that gene product sends the signal “divide” and a tumor will re- sult. The oncogenic defect can be in any of the proteins involved in communicating the “divide” signal. We know of oncogenes that encode secreted proteins, growth fac- tors, transmembrane proteins (receptors), cytoplasmic proteins (G proteins and protein kinases), and the nu- clear transcription factors that control the expression of genes essential for cell division (Jun, Fos). 12.10 Oncogenes, Tumor Suppressor Genes, and Programmed Cell Death 471 Normal cell is infected with retrovirus. Retrovirus Gene for regulatory growth protein (proto-oncogene) Host cell now has retroviral genome incorporated near proto-oncogene. Forming virus encapsulates proto-oncogene and viral genome. Retrovirus with proto-oncogene infection cycles Mutation creates oncogene. Retrovirus with oncogene invades normal cell. Transformed cell, producing defective regulatory protein 3 4 5 2 1 FIGURE 12–47 Conversion of a regulatory gene to a viral oncogene. 1 A normal cell is infected by a retrovirus (Chapter 26), which 2 inserts its own genome into the chromosome of the host cell, near the gene for a regulatory protein (the proto-oncogene). 3 Viral par- ticles released from the infected cell sometimes “capture” a host gene, in this case a proto-oncogene. 4 During several cycles of infection, a mutation occurs in the viral proto-oncogene, converting it to an oncogene. 5 When the virus subsequently infects a cell, it introduces the oncogene into the cell’s DNA. Transcription of the oncogene leads to the production of a defective regulatory protein that continuously gives the signal for cell division, overriding normal regulatory mech- anisms. Host cells infected with oncogene-carrying viruses undergo unregulated cell division—they form tumors. Proto-oncogenes can also undergo mutation to oncogenes without the intervention of a retrovirus, as described in the text. 8885d_c12_471 2/20/04 1:29 PM Page 471 mac76 mac76:385_reb: Some oncogenes encode surface receptors with de- fective or missing signal-binding sites such that their in- trinsic Tyr kinase activity is unregulated. For example, the protein ErbB is essentially identical to the normal receptor for epidermal growth factor, except that ErbB lacks the amino-terminal domain that normally binds EGF (Fig. 12–48) and as a result sends the “divide” sig- nal whether EGF is present or not. Mutations in erbB2, the gene for a receptor Tyr kinase related to ErbB, are commonly associated with cancers of the glandular ep- ithelium in breast, stomach, and ovary. (For an expla- nation of the use of abbreviations in naming genes and their products, see Chapter 25.) Mutant forms of the G protein Ras are common in tumor cells. The ras oncogene encodes a protein with normal GTP binding but no GTPase activity. The mu- tant Ras protein is therefore always in its activated (GTP-bound) form, regardless of the signals arriving through normal receptors. The result can be unregu- lated growth. Mutations in ras are associated with 30% to 50% of lung and colon carcinomas and more than 90% of pancreatic carcinomas. Defects in Tumor Suppressor Genes Remove Normal Restraints on Cell Division Tumor suppressor genes encode proteins that normally restrain cell division. Mutation in one or more of these genes can lead to tumor formation. Unregulated growth due to defective tumor suppressor genes, unlike that due to oncogenes, is genetically re- cessive; tumors form only if both chromosomes of a pair contain a defective gene. In a person who inherits one correct copy and one defective copy, every cell has one defective copy of the gene. If any one of those 10 12 so- matic cells undergoes mutation in the one good copy, a tumor may grow from that doubly mutant cell. Muta- tions in both copies of the genes for pRb, p53, or p21 yield cells in which the normal restraint on cell division is lost and a tumor forms. Retinoblastoma is a cancer of the retina that occurs in children who have two defective Rb alleles. Very young children who develop retinoblastoma commonly have multiple tumors in both eyes. Each tumor is de- rived from a single retinal cell that has undergone a mu- tation in its one good copy of the Rb gene. (A fetus with two mutant alleles in every cell is nonviable.) Retinoblastoma patients also have a high incidence of cancers of the lung, prostate, and breast. A far less likely event is that a person born with two good copies of a gene will have two independent muta- tions in the same gene in the same cell, but this does occur. Some individuals develop retinoblastomas later in childhood, usually with only one tumor in only one eye. These individuals were presumably born with two good copies of Rb in every cell, but both Rb genes in a single retinal cell have undergone mutation, leading to a tumor. Mutations in the gene for p53 also cause tumors; in more than 90% of human cutaneous squamous cell car- cinomas (skin cancers) and about 50% of all other hu- man cancers, p53 is defective. Those very rare individ- uals who inherit one defective copy of p53 commonly have the Li-Fraumeni cancer syndrome, in which mul- tiple cancers (of the breast, brain, bone, blood, lung, and skin) occur at high frequency and at an early age. The explanation for multiple tumors in this case is the same as that for Rb mutations: an individual born with one defective copy of p53 in every somatic cell is likely to suffer a second p53 mutation in more than one cell in his or her lifetime. Mutations in oncogenes and tumor suppressor genes do not have an all-or-none effect. In some can- cers, perhaps in all, the progression from a normal cell to a malignant tumor requires an accumulation of mutations (sometimes over several decades), none of which, alone, is responsible for the end effect. For ex- ample, the development of colorectal cancer has several recognizable stages, each associated with a mutation (Fig. 12–49). If a normal epithelial cell in the colon un- dergoes mutation of both copies of the tumor suppres- sor gene APC (adenomatous polyposis coli), it begins to divide faster than normal and produces a clone of itself, a benign polyp (early adenoma). For reasons not yet known, the APC mutation results in chromosomal in- stability; whole regions of a chromosome are lost or re- Chapter 12 Biosignaling472 EGF-binding domain EGF-binding site empty; tyrosine kinase is inactive. Binding of EGF activates tyrosine kinase. Tyrosine kinase is constantly active. Tyrosine kinase domain EGF Extracellular space Normal EGF receptor ErbB protein FIGURE 12–48 Oncogene-encoded defective EGF receptor. The product of the erbB oncogene (the ErbB protein) is a truncated ver- sion of the normal receptor for epidermal growth factor (EGF). Its in- tracellular domain has the structure normally induced by EGF bind- ing, but the protein lacks the extracellular binding site for EGF. Unregulated by EGF, ErbB continuously signals cell division. 8885d_c12_472 2/20/04 1:30 PM Page 472 mac76 mac76:385_reb: arranged during cell division. This instability can lead to another mutation, commonly in ras, that converts the clone into an intermediate adenoma. A third mutation (probably in the tumor suppressor gene DCC) leads to a late adenoma. Only when both copies of p53 become defective does this cell mass become a carcinoma, a ma- lignant, life-threatening cancer. The full sequence there- fore requires at least seven genetic “hits”: two on each of three tumor suppressor genes (APC, DCC, and p53) and one on the protooncogene ras. There are probably several other routes to colorectal cancer as well, but the principle that full malignancy results only from multiple mutations is likely to hold. When a polyp is detected in the early adenoma stage and the cells containing the first mutations are removed surgically, late adenomas and carcinomas will not develop; hence the importance of early detection. ■ Apoptosis Is Programmed Cell Suicide Many cells can precisely control the time of their own death by the process of programmed cell death, or apoptosis (appH11032-a-toeH11032-sis; from the Greek for “drop- ping off,” as in leaves dropping in the fall). In the de- velopment of an embryo, for example, some cells must die. Carving fingers from stubby limb buds requires the precisely timed death of cells between developing fin- ger bones. During development of the nematode Caenorhabditis elegans from a fertilized egg, exactly 131 cells (of a total of 1,090 somatic cells in the em- bryo) must undergo programmed death in order to con- struct the adult body. Apoptosis also has roles in processes other than de- velopment. When an antibody-producing cell begins to make antibodies against an antigen normally present in the body, that cell undergoes programmed death in the thymus gland—an essential mechanism for eliminating anti-self antibodies. The monthly sloughing of cells of the uterine wall (menstruation) is another case of apop- tosis mediating normal cell death. Sometimes cell sui- cide is not programmed but occurs in response to bio- logical circumstances that threaten the rest of the organism. For example, a virus-infected cell that dies before completion of the infection cycle prevents spread of the virus to nearby cells. Severe stresses such as heat, hyperosmolarity, UV light, and gamma irradiation also trigger cell suicide; presumably the organism is better off with aberrant cells dead. The regulatory mechanisms that trigger apoptosis involve some of the same proteins that regulate the cell cycle. The signal for suicide often comes from outside, through a surface receptor. Tumor necrosis factor (TNF), produced by cells of the immune system, inter- acts with cells through specific TNF receptors. These receptors have TNF-binding sites on the outer face of the plasma membrane and a “death domain” of about 80 amino acid residues that passes the self-destruct sig- nal through the membrane to cytosolic proteins such as TRADD (TNF receptor-associated death domain) (Fig. 12–50). Another receptor, Fas, has a similar death do- main that allows it to interact with the cytosolic protein FADD (Fas-associated death domain), which activates a cytosolic protease called caspase 8. This enzyme be- longs to a family of proteases that participate in apop- tosis; all are synthesized as inactive proenzymes, all have a critical Cys residue at the active site, and all hydrolyze their target proteins on the carboxyl-terminal side of specific Asp residues (hence the name caspase). When caspase 8, an “initiator” caspase, is activated by an apoptotic signal carried through FADD, it further self-activates by cleaving its own proenzyme form. Mi- tochondria are one target of active caspase 8. The pro- tease causes the release of certain proteins contained between the inner and outer mitochondrial membranes: 12.10 Oncogenes, Tumor Suppressor Genes, and Programmed Cell Death 473 APC ras DCC? p53 ? ? Normal colorectal epithelium Early adenoma Intermediate adenoma Advanced adenoma Colorectal carcinoma Invasive carcinoma Metastatic carcinoma Tumor suppressor gene Oncogene Unknown status FIGURE 12–49 From normal epithelial cell to colorectal cancer. In the colon, mutations in both copies of the tumor suppressor gene APC lead to benign clusters of epithelial cells that multiply too rapidly (early adenoma). If a cell already defective in APC suffers a second muta- tion in the proto-oncogene ras, the doubly mutant cell gives rise to an intermediate adenoma, forming a benign polyp of the colon. When one of these cells undergoes further mutations in the tumor suppres- sor genes DCC (probably) and p53, increasingly aggressive tumors form. Finally, mutations in genes not yet characterized lead to a ma- lignant tumor and finally to a metastatic tumor that can spread to other tissues. Most malignant tumors probably result from a series of muta- tions such as this. 8885d_c12_473 2/20/04 1:30 PM Page 473 mac76 mac76:385_reb: cytochrome c (Chapter 19) and several “effector” cas- pases. Cytochrome c binds to the proenzyme form of the effector enzyme caspase 9 and stimulates its proteolytic activation. The activated caspase 9 in turn catalyzes wholesale destruction of cellular proteins—a major cause of apoptotic cell death. One specific target of caspase action is a caspase-activated deoxyribonuclease. In apoptosis, the monomeric products of protein and DNA degradation (amino acids and nucleotides) are released in a controlled process that allows them to be taken up and reused by neighboring cells. Apoptosis thus allows the organism to eliminate a cell without wasting its components. SUMMARY 12.10 Oncogenes, Tumor Suppressor Genes, and Programmed Cell Death ■ Oncogenes encode defective signaling proteins. By continually giving the signal for cell division, they lead to tumor formation. Oncogenes are genetically dominant and may encode defective growth factors, receptors, G proteins, protein kinases, or nuclear regulators of transcription. ■ Tumor suppressor genes encode regulatory proteins that normally inhibit cell division; mutations in these genes are genetically recessive but can lead to tumor formation. ■ Cancer is generally the result of an accumulation of mutations in oncogenes and tumor suppressor genes. ■ Apoptosis can be triggered by extracellular signals such as TNF through plasma membrane receptors. Chapter 12 Biosignaling474 FIGURE 12–50 Initial events of apoptosis. Receptors in the plasma membrane (Fas, TNF-R1) receive signals from outside the cell (the Fas ligand or tumor necrosis factor (TNF), respectively). Activated recep- tors foster interaction between the “death domain” (an 80 amino acid sequence) in Fas or TNF-R1 and a similar death domain in the cy- tosolic proteins FADD or TRADD. FADD activates a cytosolic pro- tease, caspase 8, that proteolytically activates other cellular proteases. TRADD also activates proteases. The resulting proteolysis is a primary factor in cell death. Fas Fas ligand FADD TNF-R1 TNF TRADD Mitochondrion Plasma membrane Effector caspases Activation of DNase Cytochrome c Death domains Protein degradation Cell death Caspase 8 (initiator) Key Terms signal transduction 421 enzyme cascade 422 desensitization 422 ligand-gated receptor channel 426 voltage-gated ion channel 427 second messenger 428 autophosphorylation 429 SH2 domain 429 G proteins 429 MAPK cascade 430 receptor Tyr kinase 432 serpentine receptors 435 G protein–coupled receptors (GPCR) 435 7 transmembrane segment (7tm) receptors 435 stimulatory G protein (G s ) 436 H9252-adrenergic receptor kinase (H9252ARK) 441 H9252-arrestin (H9252arr; arrestin 2) 441 G protein–coupled receptor kinases (GRKs) 441 scaffold proteins 441 inhibitory G protein (G i ) 441 calmodulin (CaM) 444 Ca 2+ /calmodulin-dependent protein kinases (CaM kinases I–IV) 444 two-component signaling systems 452 receptor His kinase 452 response regulator 452 receptorlike kinase (RLK) 455 hormone response element (HRE) 465 tamoxifen 465 RU486 466 cyclin 467 cyclin-dependent protein kinase (CDK) 467 ubiquitin 469 proteasome 469 growth factors 469 cytokine 469 retinoblastoma protein (pRb) 470 oncogene 471 tumor suppressor genes 472 programmed cell death 473 apoptosis 473 Terms in bold are defined in the glossary. 8885d_c12_474 2/20/04 1:30 PM Page 474 mac76 mac76:385_reb: Chapter 12 Further Reading 475 Further Reading General Cohen, P. (2001) The role of protein phosphorylation in human health and disease: the Sir Hans Krebs Medal Lecture. Eur. J. Biochem. 268, 5001–5010. An intermediate-level review of the role of protein kinases, their alteration in disease, and the drugs that affect their activity. Cohen, P. (2000) The regulation of protein function by multisite phosphorylation—a 25 year update. Trends Biochem. Sci. 25, 596–601. Historical account of the developments in protein phosphorylation. Heilmeyer, L. & Friedrich, P. (eds) (2001) Protein Modules in Cellular Signalling, IOS Press, Washington, DC. Receptor Ion Channels See also Chapter 11, Further Reading, Ion Channels. Aidley, D.J. & Stanfield, P.R. (1996) Ion Channels: Molecules in Action, Cambridge University Press, Cambridge. Clear, concise introduction to the physics, chemistry, and molecular biology used in research on ion channels; emphasis is on molecular approaches. Lehmann-Horn, F. & Jurkat-Rott, K. (1999) Voltage-gated ion channels and hereditary disease. Physiol. Rev. 79, 1317–1372. Advanced review of the structure and function of ion channels, with special emphasis on cases in which ion-channel defects produce human diseases. Receptor Enzymes Foster, D.C., Wedel, B.J., Robinson, S.W., & Garbers, D.L. (1999) Mechanisms of regulation and functions of guanylyl cyclases. Rev. Physiol. Biochem. Pharmacol. 135, 1–39. Advanced review of the structure and function of signal- transducing guanylyl cyclases. Saltiel, A.R. & Pessin, J.E. (2002) Insulin signaling pathways in time and space. Trends Biochem Sci. 12, 65–71. Short, intermediate-level review. Schaeffer, H.J. & Weber, M.J. (1999) Mitogen-activated protein kinases: specific messages from ubiquitous messengers. Mol. Cell. Biol. 19, 2435–2444. Intermediate-level review of MAPKs and the basis for specific signaling through these general signaling proteins. Schlessinger, J. (2000) Cell signaling by receptor tyrosine kinases. Cell 103, 211–225. Shepherd, P.R., Withers, D.J., & Siddle, K. (1998) Phospho- inositide 3-kinase: the key switch mechanism in insulin signalling. Biochem. J. 333, 471–490. Intermediate-level review of the importance of PKB and PI-3K in metabolic regulation by insulin. Shields, J.M., Pruitt, K., McFall, A., Shaub, A., & Der, C.J. (2000) Understanding Ras: it ain’t over ’til it’s over. Trends Cell Biol. 10, 147–154. Intermediate-level review of the monomeric G protein Ras. Widmann, C., Gibson, S., Jarpe, M.B., & Johnson, G.L. (1999) Mitogen-activated protein kinase: conservation of a three- kinase module from yeast to human. Physiol. Rev. 79, 143–180. Advanced review of the roles of MAPKs in diverse organisms, from yeast, slime mold, and nematode to vertebrates and plants. Zajchowski, L.D. & Robbins, S.M. (2002) Lipid rafts and little caves: compartmentalized signalling in membrane microdomains. Eur. J. Biochem. 269, 737–752. Serpentine Receptors Hahn, K. & Toutchkine, A. (2002) Live-cell fluorescent biosen- sors for activated signaling proteins. Curr. Opin. Cell Biol. 14, 167–172. Brief, intermediate-level review. Hamm, H.E. (1998) The many faces of G protein signaling. J. Biol. Chem. 273, 669–672. Introduction to a series of short reviews on G proteins. Lodowski, D.T., Pitcher, J.A., Capel, W.D., Lefkowitz, R.J., & Tesmer, J.J.G. (2003) Keeping G proteins at bay: a complex be- tween G protein–coupled receptor kinase 2 and G H9252H9253 . Science 300, 1256–1262. Martin, T.F.J. (1998) Phosphoinositide lipids as signaling mole- cules. Annu. Rev. Cell Dev. Biol. 14, 231–264. Discussion of the roles of phosphatidylinositol derivatives in signal transduction, cytoskeletal regulation, and membrane trafficking. Perry, S.J. & Lefkowitz, R.J. (2002) Arresting developments in heptahelical receptor signaling and regulation. Trends Cell Biol. 12, 130–138. Role of arrestins in serpentine receptor adaptation. Pinna, L.A. & Ruzzene, M. (1996) How do protein kinases rec- ognize their substrates? Biochim. Biophys. Acta 1314, 191–225. Advanced review of the factors, including consensus sequences, that give protein kinases their specificity. Roach, P.J. (1991) Multisite and hierarchal protein phosphoryla- tion. J. Biol. Chem. 266, 14,139–14,142. Report on the importance of multiple phosphorylation sites in the fine regulation of protein function. Skiba, N.P. & Hamm, H.E. (1998) How G sH9251 activates adenylyl cyclase. Nat. Struct. Biol. 5, 88–92. Intermediate-level review of the mechanism of G-protein action, based on structural studies. Zaccolo, M., De Giorgi, F., Cho, C.Y., Feng, L., Knapp, T., Negulescu, P.A., Taylor, S.S., Tsien, R.Y., & Pozzan, T. (2000) A genetically encoded, fluorescent indicator for cyclic AMP in liv- ing cells. Nat. Cell Biol. 2, 25–29. Describes the technique shown in Figure 5 of Box 12–2. Zhang, J., Campbell, R.E., Ting, A.Y., & Tsien, R.Y. (2002) Creating new fluorescent probes for cell biology. Nat. Rev. Molec. Cell Biol. 3, 906–918. The basis for techniques such as those described in Box 12–2. Scaffold Proteins and Membrane Rafts See also Chapter 11, Further Reading, Membrane Dynamics. Filipek, S., Stenkamp, R.E., Teller, D.C., & Palczewski, K. (2003) G protein-coupled receptor rhodopsin: A prospectus. Annu. Rev. Physiol. 65, 851–879. Advanced review. 8885d_c12_475 2/20/04 1:30 PM Page 475 mac76 mac76:385_reb: Chapter 12 Biosignaling476 Lim, W.A. (2002) The modular logic of signaling proteins: building allosteric switches from simple binding domains. Curr. Opin. Struct. Biol. 12, 61–68. Michel, J.J.C. & Scott, J.D. (2002) AKAP mediated signal transduction. Annu. Rev. Pharmacol. Toxicol. 42, 235–257. Advanced review of the proteins that target PKA to subcellular compartments. Neel, B.G., Gu, H., & Pao, L. (2003) The “Shp”ing news: SH2 domain–containing tyrosine phosphatases in cell signaling. Trends Biochem. Sci. 28, 284–293. Pawson, T. & Nash, P. (2003) Assembly of cell regulatory systems through protein interaction domains. Science 300, 445–452. Intermediate-level review of multivalent proteins and the com- plexes they form. Saltiel, A.R. & Pessin, J.E. (2002) Insulin signaling pathways in time and space. Trends Cell Biol. 12, 65–71. Siegal, G. (1999) The surprisingly flexible PTB domain. Nat. Struct. Biol. 6, 7–10. Tsui-Pierchala, B.A., Encinas, M., Milbrandt, J., & Johnson, E.M., Jr. (2002) Lipid rafts in neuronal signaling and function. Trends Neurosci. 25, 412–417. Yaffe, M.D. & Elia, A.E.H. (2001) Phosphoserine/threonine- binding domains. Curr. Opin. Cell Biol. 13, 131–138. Calcium Ions in Signaling Berridge, M.J. (1993) Inositol triphosphate and calcium sig- nalling. Nature 361, 315–325. Classic description of the IP 3 signaling system. Berridge, M.J., Lipp, P., & Bootman, M.D. (2000) The versatil- ity and universality of calcium signaling. Nat. Rev. Molec. Cell Biol. 1, 11–21. Intermediate review. Chin, D. & Means, A.R. (2000) Calmodulin: a prototypical calcium receptor. Trends Cell Biol. 10, 322–328. Nowycky, M.C. & Thomas, A.P. (2002) Intracellular calcium signaling. J. Cell Sci. 115, 3715–3716. A comprehensive poster of all elements of Ca 2H11001 signaling. Takahashi, A., Camacho, P., Lechleiter, J.D., & Herman, B. (1999) Measurement of intracellular calcium. Physiol. Rev. 79, 1089–1125. Advanced review of methods for estimating intracellular Ca 2H11001 levels in real time. Thomas, A.P., Bird, G.S.J., Hajnoczky, G., Robb-Gaspers, L.D., & Putney, J.W., Jr. (1996) Spatial and temporal aspects of cellular calcium signaling. FASEB J. 10, 1505–1517. Signaling in Plants and Bacteria Bakal, C.J. & Davies, J.E. (2000) No longer an exclusive club: eukaryotic signaling domains in bacteria. Trends Cell Biol. 10, 32–38. Intermediate review. Becraft, P.W. (2002) Receptor kinase signaling in plant develop- ment. Annu. Rev. Cell Dev. Biol. 18, 163–192. Advanced review. Chang, C. & Stadler, R. (2001) Ethylene hormone receptor action in Arabidopsis. Bioessays 23, 619–627. Cock, J.M., Vanoosthuyse, V., & Gaude, T. (2002) Receptor kinase signalling in plants and animals: distinct molecular systems with mechanistic similarities. Curr. Opin. Cell Biol. 14, 230–236. Hwang, I., Chen, H.-C., & Sheen, J. (2002) Two-component circuitry in Arabidopsis cytokinin signal transduction. Nature 413, 383–389. Jones, A.M. (2002) G-protein–coupled signaling in Arabidopsis. Curr. Opin. Plant Biol. 5, 402–407. Matsubayashi, Y., Yang, H., & Sakagami, Y. (2001) Peptide signals and their receptors in higher plants. Trends Plant Sci. 6, 573–577. McCarty, D.R. & Chory, J. (2000) Conservation and innovation in plant signaling pathways. Cell 103, 201–209. Meijer, H.J.G. & Munnik, T. (2003) Phospholipid-based signaling in plants. Annu. Rev. Plant Biol. 54, 265–306. Advanced review. Ouaked, F., Rozhon, W., Lecourieus, D., & Hirt, H. (2003) A MAPK pathway mediates ethylene signaling in plants. EMBO J. 22, 1282–1288. Talke, I.N., Blaudez, D., Maathuis, F.J.M., & Sanders, D. (2003) CNGCs: prime targets of plant cyclic nucleotide signalling? Trends Plant Sci. 8, 286–293. Tichtinsky, G., Vanoosthuyse, V., Cock, J.M., & Gaude, T. (2003) Making inroads into plant receptor kinase signalling path- ways. Trends Plant Sci. 8, 231–237. Vision, Olfaction, and Gustation Baylor, D. (1996) How photons start vision. Proc. Natl. Acad. Sci. USA 93, 560–565. One of six short reviews on vision in this journal issue. Margolskee, R.F. (2002) Molecular mechanisms of bitter and sweet taste transduction. J. Biol. Chem. 277, 1–4. Menon, S.T., Han, M., & Sakmar, T.P. (2001) Rhodopsin: struc- tural basis of molecular physiology. Physiol. Rev. 81, 1659–1688. Advanced review. Mombaerts, P. (2001) The human repertoire of odorant receptor genes and pseudogenes. Annu. Rev. Genomics Hum. Genet. 2, 493–510. Advanced review. Nathans, J. (1989) The genes for color vision. Sci. Am. 260 (February), 42–49. Ronnett, G.V. & Moon, C. (2002) G proteins and olfactory signal transduction. Annu. Rev. Physiol. 64, 189–222. Advanced review. Scott, K. & Zuker, C. (1997) Lights out: deactivation of the pho- totransduction cascade. Trends Biochem. Sci. 22, 350–354. Steroid Hormone Receptors and Action Carson-Jurica, M.A., Schrader, W.T., & O’Malley, B.W. (1990) Steroid receptor family—structure and functions. Endocr. Rev. 11, 201–220. Advanced discussion of the structure of nuclear hormone receptors and the mechanisms of their action. Hall, J.M., Couse, J.F., & Korach, K.S. (2001) The multifac- eted mechanisms of estradiol and estrogen receptor signaling. J. Biol. Chem. 276, 36,869–36,872. Brief, intermediate-level review. 8885d_c12_476 2/20/04 1:30 PM Page 476 mac76 mac76:385_reb: Chapter 12 Problems 477 1. Therapeutic Effects of Albuterol The respi- ratory symptoms of asthma result from constriction of the bronchi and bronchioles of the lungs due to contrac- tion of the smooth muscle of their walls. This constriction can be reversed by raising the [cAMP] in the smooth muscle. Ex- plain the therapeutic effects of albuterol, a H9252-adrenergic ag- onist taken (by inhalation) for asthma. Would you expect this drug to have any side effects? How might one design a bet- ter drug that did not have these effects? 2. Amplification of Hormonal Signals Describe all the sources of amplification in the insulin receptor system. 3. Termination of Hormonal Signals Signals carried by hormones must eventually be terminated. Describe several different mechanisms for signal termination. 4. Specificity of a Signal for a Single Cell Type Dis- cuss the validity of the following proposition. A signaling molecule (hormone, growth factor, or neurotransmitter) elic- its identical responses in different types of target cells if they contain identical receptors. 5. Resting Membrane Potential A variety of unusual in- vertebrates, including giant clams, mussels, and polychaete worms, live on the fringes of hydrothermal vents on the ocean bottom, where the temperature is 60 H11034C. (a) The adductor muscle of a deep-sea giant clam has a resting membrane potential of H1100295 mV. Given the intracellu- lar and extracellular ionic compositions shown below, would you have predicted this membrane potential? Why or why not? (b) Assume that the adductor muscle membrane is per- meable to only one of the ions listed above. Which ion could determine the V m ? Concentration (mM) Ion Intracellular Extracellular Na H11001 50 440 K H11001 400 20 Cl H11002 21 560 Ca 2H11001 0.4 10 Problems Jordan, V.C. (1998) Designer estrogens. Sci. Am. 279 (October), 60–67. Introductory review of the mechanism of estrogen action and the effects of estrogenlike compounds in medicine. L?sel, R.M., Falkenstein, E., Feuring, M., Schultz, A., Tillmann, H.-C., Rossol-Haseroth, K., & Wehling, M. (2003) Nongenomic steroid action: controversies, questions, and answers. Physiol. Rev. 83, 965–1016. Detailed review of the evidence for steroid hormone action through plasma membrane receptors. Cell Cycle and Cancer Cavenee, W.K. & White, R.L. (1995) The genetic basis of cancer. Sci. Am. 272 (March), 72–79. Chau, B.N. & Wang, J.Y.J. (2003) Coordinated regulation of life and death by RB. Nat. Rev. Cancer 3, 130–138. Fearon, E.R. (1997) Human cancer syndromes—clues to the ori- gin and nature of cancer. Science 278, 1043–1050. Intermediate-level review of the role of inherited mutations in the development of cancer. Herwig, S. & Strauss, M. (1997) The retinoblastoma protein: a master regulator of cell cycle, differentiation and apoptosis. Eur. J. Biochem. 246, 581–601. Hunt, M. & Hunt, T. (1993) The Cell Cycle: An Introduction, W. H. Freeman and Company/Oxford University Press, New York/ Oxford. Kinzler, K.W. & Vogelstein, B. (1996) Lessons from hereditary colorectal cancer. Cell 87, 159–170. Evidence for multistep processes in the development of cancer. Levine, A.J. (1997) p53, the cellular gatekeeper for growth and division. Cell 88, 323–331. Intermediate coverage of the function of protein p53 in the normal cell cycle and in cancer. Morgan, D.O. (1997) Cyclin-dependent kinases: engines, clocks, and microprocessors. Annu. Rev. Cell Dev. Biol. 13, 261–291. Advanced review. Obaya, A.J. & Sedivy, J.M. (2002) Regulation of cyclin-Cdk activity in mammalian cells. Cell. Mol. Life Sci. 59, 126–142. Rajagopalan, H., Nowak, M.A., Vogelstein, B., & Lengauer, C. (2003) The significance of unstable chromosomes in colorectal cancer. Nat. Rev. Cancer 3, 695–701. Sherr, C.J. & McCormick, F. (2002) The RB and p53 pathways in cancer. Cancer Cell 2, 102–112. Weinberg, R.A. (1996) How cancer arises. Sci. Am. 275 (Septem- ber), 62–70. Yamasaki, L. (2003) Role of the RB tumor suppressor in cancer. Cancer Treatment Res. 115, 209–239. Advanced review. Apoptosis Anderson, P. (1997) Kinase cascades regulating entry into apoptosis. Microbiol. Mol. Biol. Rev. 61, 33–46. Ashkenazi, A. & Dixit, V.M. (1998) Death receptors: signaling and modulation. Science 281, 1305–1308. This and the papers by Green and Reed and by Thornberry and Lazebnik (below) are in an issue of Science devoted to apoptosis. Duke, R.C., Ojcius, D.M., & Young, J.D.-E. (1996) Cell suicide in health and disease. Sci. Am. 275 (December), 80–87. Green, D.R. & Reed, J.C. (1998) Mitochondria and apoptosis. Science 281, 1309–1312. Jacobson, M.D., Weil, M., & Raff, M.C. (1997) Programmed cell death in animal development. Cell 88, 347–354. Lawen, A. (2003) Apoptosis—an introduction. Bioessays 25, 888–896. Thornberry, N.A. & Lazebnik, Y. (1998) Caspases: enemies within. Science 281, 1312. 8885d_c12_477 2/20/04 1:31 PM Page 477 mac76 mac76:385_reb: Chapter 12 Biosignaling478 6. Membrane Potentials in Frog Eggs Fertilization of a frog oocyte by a sperm cell triggers ionic changes similar to those observed in neurons (during movement of the action potential) and initiates the events that result in cell division and development of the embryo. Oocytes can be stimulated to divide without fertilization by suspending them in 80 mM KCl (normal pond water contains 9 mM KCl). (a) Calculate how much the change in extracellular [KCl] changes the resting membrane potential of the oocyte. (Hint: Assume the oocyte contains 120 mM K H11001 and is perme- able only to K H11001 .) Assume a temperature of 20 H11034C. (b) When the experiment is repeated in Ca 2H11001 -free wa- ter, elevated [KCl] has no effect. What does this suggest about the mechanism of the KCl effect? 7. Excitation Triggered by Hyperpolarization In most neurons, membrane depolarization leads to the opening of voltage-dependent ion channels, generation of an action po- tential, and ultimately an influx of Ca 2H11001 , which causes release of neurotransmitter at the axon terminus. Devise a cellular strategy by which hyperpolarization in rod cells could pro- duce excitation of the visual pathway and passage of visual signals to the brain. (Hint: The neuronal signaling pathway in higher organisms consists of a series of neurons that relay information to the brain (see Fig. 12–31). The signal released by one neuron can be either excitatory or inhibitory to the following, postsynaptic neuron.) 8. Hormone Experiments in Cell-Free Systems In the 1950s, Earl W. Sutherland, Jr., and his colleagues carried out pioneering experiments to elucidate the mechanism of action of epinephrine and glucagon. Given what you have learned in this chapter about hormone action, interpret each of the ex- periments described below. Identify substance X and indicate the significance of the results. (a) Addition of epinephrine to a homogenate of normal liver resulted in an increase in the activity of glycogen phos- phorylase. However, if the homogenate was first centrifuged at a high speed and epinephrine or glucagon was added to the clear supernatant fraction that contains phosphorylase, no increase in the phosphorylase activity occurred. (b) When the particulate fraction from the centrifuga- tion in (a) was treated with epinephrine, substance X was produced. The substance was isolated and purified. Unlike epinephrine, substance X activated glycogen phosphorylase when added to the clear supernatant fraction of the cen- trifuged homogenate. (c) Substance X was heat-stable; that is, heat treatment did not affect its capacity to activate phosphorylase. (Hint: Would this be the case if substance X were a protein?) Sub- stance X was nearly identical to a compound obtained when pure ATP was treated with barium hydroxide. (Fig. 8–6 will be helpful.) 9. Effect of Cholera Toxin on Adenylyl Cyclase The gram-negative bacterium Vibrio cholerae pro- duces a protein, cholera toxin (M r 90,000), that is responsi- ble for the characteristic symptoms of cholera: extensive loss of body water and Na H11001 through continuous, debilitating di- arrhea. If body fluids and Na H11001 are not replaced, severe de- hydration results; untreated, the disease is often fatal. When the cholera toxin gains access to the human intestinal tract it binds tightly to specific sites in the plasma membrane of the epithelial cells lining the small intestine, causing adenyl- yl cyclase to undergo prolonged activation (hours or days). (a) What is the effect of cholera toxin on [cAMP] in the intestinal cells? (b) Based on the information above, suggest how cAMP normally functions in intestinal epithelial cells. (c) Suggest a possible treatment for cholera. 10. Effect of Dibutyryl cAMP versus cAMP on Intact Cells The physiological effects of epinephrine should in principle be mimicked by addition of cAMP to the target cells. In practice, addition of cAMP to intact target cells elicits only a minimal physiological response. Why? When the structurally related derivative dibutyryl cAMP (shown below) is added to intact cells, the expected physiological response is readily ap- parent. Explain the basis for the difference in cellular re- sponse to these two substances. Dibutyryl cAMP is widely used in studies of cAMP function. 11. Nonhydrolyzable GTP Analogs Many enzymes can hydrolyze GTP between the H9252 and H9253 phosphates. The GTP analog H9252,H9253-imidoguanosine 5H11032-triphosphate Gpp(NH)p, shown below, cannot be hydrolyzed between the H9252 and H9253 phosphates. Predict the effect of microinjection of Gpp(NH)p into a myo- cyte on the cell’s response to H9252-adrenergic stimulation. 12. G Protein Differences Compare the G proteins G s , which acts in transducing the signal from H9252-adrenergic re- ceptors, and Ras. What properties do they share? How do they differ? What is the functional difference between G s and G I ? Gpp(NH)p CH 2 O O OH H9253H9252( , -imidoguanosine 5H11032-triphosphate) OH H HH H N O N HN H 2 N N O H11002 P O O O H11002 P O H11002 O P O H11002 O H N Dibutyryl cAMP (N 6 ,O 2 H11032 -Dibutyryl adenosine 3H11032,5H11032-cyclic monophosphate) (CH 2 ) 2 CH 3 CH 2 C (CH 2 ) 2 CH 3 C O OO O O H HH H N NH N N N O P O O H11002 8885d_c12_478 2/20/04 2:03 PM Page 478 mac76 mac76:385_reb: Chapter 12 Problems 479 13. EGTA Injection EGTA (ethylene glycol-bis(H9252-amino- ethyl ether)-N,N,NH11032,NH11032-tetraacetic acid) is a chelating agent with high affinity and specificity for Ca 2H11001 . By microinjecting a cell with an appropriate Ca 2H11001 -EDTA solution, an experi- menter can prevent cytosolic [Ca 2H11001 ] from rising above 10 H110027 M. How would EGTA microinjection affect a cell’s response to vasopressin (see Table 12–5)? To glucagon? 14. Visual Desensitization Oguchi’s disease is an inherited form of night blindness. Affected individu- als are slow to recover vision after a flash of bright light against a dark background, such as the headlights of a car on the freeway. Suggest what the molecular defect(s) might be in Oguchi’s disease. Explain in molecular terms how this de- fect accounts for the night blindness. 15. Mutations in PKA Explain how mutations in the R or C subunit of cAMP-dependent protein kinase (PKA) might lead to (a) a constantly active PKA or (b) a constantly inac- tive PKA. 16. Mechanisms for Regulating Protein Kinases Iden- tify eight general types of protein kinases found in eukary- otic cells, and explain what factor is directly responsible for activating each type. 17. Mutations in Tumor Suppressor Genes and Onco- genes Explain why mutations in tumor suppressor genes are recessive (both copies of the gene must be defective for the regulation of cell division to be defective) whereas mu- tations in oncogenes are dominant. 18. Retinoblastoma in Children Explain why some children with retinoblastoma develop multiple tumors of the retina in both eyes, whereas others have a sin- gle tumor in only one eye. 19. Mutations in ras How does a mutation in the ras gene that leads to formation of a Ras protein with no GTPase activity affect a cell’s response to insulin? 8885d_c12_479 2/20/04 1:31 PM Page 479 mac76 mac76:385_reb: