Chemistry of Proteins

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 50

Applied Biochemistry

and Molecular Biology

(ABMB)
BCh 152

Chemistry of Proteins

and Nucleic Acids


A comprehensive survey of biochemistry, including

protein structure, enzyme kinetics, membrane

structure, and DNA structure. The laboratory

introduces basic techniques including buffer

preparation, and qualitative and quantitative

analyses of amino acids, proteins, and nucleic acids.

To explain and analyze the titration curves of acids,

bases, and amino acids. Protein isolation and

identification.
Amino Acids & Peptides 1
BIOMEDICAL IMPORTANCE more than 20 amino acids, its redundancy
In addition to providing the monomer units from limits the available codons to the 20 L--amino
which the long polypeptide chains of proteins are acids listed in Table 3–1, classified according to
synthesized, the L--amino acids and their the polarity of their R groups. Both one- and three-
derivatives participate in cellular functions as letter abbreviations for each amino acid can be
diverse as nerve transmission and the biosynthesis used to represent the amino acids in peptides
of porphyrins, purines, pyrimidines, and urea. (Table 3–1). Some proteins contain additional
Short polymers of amino acids called peptides amino acids that arise by modification of an
perform prominent roles in the neuroendocrine amino acid already present in a peptide. Examples
system as hormones, hormone-releasing factors, include conver- sion of peptidyl proline and lysine
neuromodula- tors, or neurotransmitters. While to 4-hydroxyproline and 5-hydroxylysine; the
proteins contain only L--amino acids, conversion of peptidyl gluta- mate to -
microorganisms elaborate peptides that contain carboxyglutamate; and the methylation,
formylation, acetylation, prenylation, and
both D- and L--amino acids. Several of these
phosphoryla- tion of certain aminoacyl residues.
peptides are of therapeutic value, including the an-
These modifications extend the biologic diversity
tibiotics bacitracin and gramicidin A and the
of proteins by altering their solubility, stability, and
antitumor agent bleomycin. Certain other
interaction with other proteins.
microbial peptides are toxic. The cyanobacterial
peptides microcystin and nodularin are lethal in
large doses, while small quantities promote the Only L-a-Amino Acids Occur in Proteins
formation of hepatic tumors. Neither hu- mans With the sole exception of glycine, the -
nor any other higher animals can synthesize 10 of carbon of amino acids is chiral. Although some
the 20 common L--amino acids in amounts protein amino acids are dextrorotatory and some
adequate to support infant growth or to maintain levorotatory, all share the absolute configuration of
health in adults. Consequently, the human diet L-glyceraldehyde and thus are L--amino acids.
must contain adequate quantities of these Several free L--amino acids fulfill important roles
nutritionally essential amino acids. in metabolic processes. Examples in- clude
ornithine, citrulline, and argininosuccinate that
PROPERTIES OF AMINO ACIDS participate in urea synthesis; tyrosine in
The Genetic Code Specifies formation of thyroid hormones; and glutamate in
20 L-a-Amino Acids neurotransmitter biosynthesis. D-Amino acids
that occur naturally in- clude free D-serine and
Of the over 300 naturally occurring amino acids, 20 D-aspartate in brain tissue, D-alanine and D-
con- stitute the monomer units of proteins. glutamate in the cell walls of gram- positive
While a nonre- dundant three-letter genetic code bacteria, and D-amino acids in some nonmam-
could accommodate malian peptides and certain antibiotics.
1
Table 1–1. L- -Amino acids present in proteins.

Name Symbol Structural Formula pK1 pK2 pK3


With Aliphatic Side Chains a-COOH a-NH3+ R Group
Glycine Gly [G] H CH COO–
2.4 9.8
+
NH3
Alanine Ala [A] – 2.4 9.9
CH3 CH COO
+
NH3
H3C
CH CH COO–
Valine Val [V] H3C + 2.2 9.7
NH
3

H3C
CH CH CH COO–
2
Leucine Leu [L] H3C + 2.3 9.7
NH
3

CH3
CH2
Isoleucine Ile [I] CH CH COO– 2.3 9.8
+
CH3 NH3

With Side Chains Containing Hydroxylic (OH) Groups


Serine Ser [S] CH CH COO
– 2.2 9.2 about 13
2

OH NH3+
Threonine Thr [T] – 2.1 9.1 about 13
CH3 CH CH COO

OH NH3+

Tyrosine Tyr [Y] See below.


With Side Chains Containing Sulfur Atoms

Cysteine Cys [C] CH2 CH COO 1.9 10.8 8.3

SH NH3+

Methionine Met [M] CH2 CH2 CH COO 2.1 9.3

S CH3 NH3+

With Side Chains Containing Acidic Groups or Their Amides


Aspartic acid Asp [D] –
OOC CH CH COO
– 2.0 9.9 3.9
2
+
NH3
Asparagine Asn [N] – 2.1 8.8
H2N C CH2 CH COO
+
O NH3

OOC CH2 CH2 CH COO–
Glutamic acid Glu [E] +
2.1 9.5 4.1
NH
3

HN C CH CH CH COO–
2 2 2
Glutamine Gln [Q] 2.2 9.1
O NH +
3

(continued )
2
Table 1–1. L--Amino acids present in proteins. (continued)

Name Symbol Structural Formula pK1 pK2 pK3


With Side Chains Containing Basic Groups a-COOH a-NH3 + R Group
Arginine Arg [R] H N CH CH CH CH COO– 1.8 9.0 12.5
2 2 2
+ +
C NH2 NH3
NH2
CH CH CH CH CH COO–
2 2 2 2
Lysine Lys [K] 2.2 9.2 10.8
NH3 + NH3 +

CH2 CH COO
Histidine His [H] +
1.8 9.3 6.0
HN N NH3

Containing Aromatic Rings


Histidine His [H] See above.

Phenylalanine Phe [F] CH2 CH COO 2.2 9.2
+
NH3
Tyrosine Tyr [Y] 2.2 9.1 10.1
HO CH2 CH COO–
+
NH3
Tryptophan Trp [W] 2.4 9.4
CH2 CH COO–
+
NH3
N
H

Imino Acid
Proline Pro [P] 2.0 10.6
+ –
N COO
H2

Amino Acids May Have Positive, Negative, Molecules that contain an equal number of ioniz-
or Zero Net Charge able groups of opposite charge and that therefore bear
no net charge are termed zwitterions. Amino acids in
Charged and uncharged forms of the ionizable blood and most tissues thus should be represented as in
 COOH and NH3+ weak acid groups exist in solu- A, below.
tion in protonic equilibrium:
NH + NH2
 
R — COOH  R — COO H 
 O– OH
R —NH3  R — NH2  H R R
O O
While both RCOOH and RNH3 are weak acids, + A B
RCOOH is a far stronger acid than RNH3+. At
physiologic pH (pH 7.4), carboxyl groups exist almost Structure B cannot exist in aqueous solution because at
entirely as RCOO and amino groups predomi- any pH low enough to protonate the carboxyl group
nantly as RNH3+. Figure 3–1 illustrates the effect of the amino group would also be protonated. Similarly,
pH on the charged state of aspartic acid. at any pH sufficiently high for an uncharged amino
O H+ O H+ O H+ O

OH OH O O–

pK1 = 2.09 + pK2 = 3.86 pK3 = 9.82


+
NH3 (-COOH) NH3 (-COOH) NH3+ (— NH )
+
NH2
–O –O –
HO O
O O O O
A B C D
In strong acid Around pH 3; Around pH 6–8; In strong alkali
(below pH 1); net charge = 0 net charge = –1 (above pH 11);
net charge = +1 net charge = –2

Figure 1–1. Protonic equilibria of aspartic acid.

group to predominate, a carboxyl group will be present At Its Isoelectric pH (pI), an Amino Acid
as RCOO. The uncharged representation B (above) Bears No Net Charge
is, however, often used for reactions that do not involve
protonic equilibria. The isoelectric species is the form of a molecule that
has an equal number of positive and negative charges
and thus is electrically neutral. The isoelectric pH, also
pKa Values Express the Strengths called the pI, is the pH midway between pKa values on
of Weak Acids either side of the isoelectric species. For an amino acid
The acid strengths of weak acids are expressed as their such as alanine that has only two dissociating groups,
pKa (Table 3–1). The imidazole group of histidine and there is no ambiguity. The first pKa (R COOH) is
the guanidino group of arginine exist as resonance hy- 2.35 and the second pKa (RNH3+) is 9.69. The iso-
brids with positive charge distributed between both ni- electric pH (pI) of alanine thus is
trogens (histidine) or all three nitrogens (arginine) (Fig-
ure 3–2). The net charge on an amino acid—the pK 1  pK 2 2.35  9.69
pl    6.02
algebraic sum of all the positively and negatively 2 2
charged groups present—depends upon the pKa values
of its functional groups and on the pH of the surround- For polyfunctional acids, pI is also the pH midway be-
ing medium. Altering the charge on amino acids and tween the pKa values on either side of the isoionic
their derivatives by varying the pH facilitates the physi- species. For example, the pI for aspartic acid is
cal separation of amino acids, peptides, and proteins
(see Chapter 4). pK 1  pK 2 2.09  3.96
pl    3.02
2 2

For lysine, pI is calculated from:


R
pK 2  pK 3
N H H pl 
2

H Similar considerations apply to all polyprotic acids (eg,


proteins), regardless of the number of dissociating
R R R
groups present. In the clinical laboratory, knowledge of
the pI guides selection of conditions for electrophoretic
NH NH NH separations. For example, electrophoresis at pH 7.0 will
C NH2 C NH2 C NH2 separate two molecules with pI values of 6.0 and 8.0
NH2 because at pH 8.0 the molecule with a pI of 6.0 will
NH2 NH2 have a net positive charge, and that with pI of 8.0 a net
negative charge. Similar considerations apply to under-
Figure 3–2. Resonance hybrids of the protonated standing chromatographic separations on ionic sup-
forms of the R groups of histidine and arginine. ports such as DEAE cellulose (see Chapter 4).
pKa Values Vary With the Environment THE a-R GROUPS DETERMINE THE
The environment of a dissociable group affects its pKa. PROPERTIES OF AMINO ACIDS
The pKa values of the R groups of free amino acids in Since glycine, the smallest amino acid, can be accommo-
aqueous solution (Table 3–1) thus provide only an ap- dated in places inaccessible to other amino acids, it often
proximate guide to the pKa values of the same amino occurs where peptides bend sharply. The hydrophobic R
acids when present in proteins. A polar environment groups of alanine, valine, leucine, and isoleucine and the
favors the charged form (R COO or RNH3+), aromatic R groups of phenylalanine, tyrosine, and tryp-
and a nonpolar environment favors the uncharged form tophan typically occur primarily in the interior of cy-
(R COOH or RNH2). A nonpolar environment tosolic proteins. The charged R groups of basic and
thus raises the pKa of a carboxyl group (making it a acidic amino acids stabilize specific protein conforma-
weaker acid) but lowers that of an amino group (making tions via ionic interactions, or salt bonds. These bonds
it a stronger acid). The presence of adjacent charged also function in ―charge relay‖ systems during enzymatic
groups can reinforce or counteract solvent effects. The catalysis and electron transport in respiring mitochon-
pKa of a functional group thus will depend upon its lo- dria. Histidine plays unique roles in enzymatic catalysis.
cation within a given protein. Variations in pKa can en- The pKa of its imidazole proton permits it to function at
compass whole pH units (Table 3–2). pKa values that neutral pH as either a base or an acid catalyst. The pri-
diverge from those listed by as much as three pH units mary alcohol group of serine and the primary thioalco-
are common at the active sites of enzymes. An extreme hol (SH) group of cysteine are excellent nucleophiles
example, a buried aspartic acid of thioredoxin, has a and can function as such during enzymatic catalysis.
pKa above 9—a shift of over six pH units! However, the secondary alcohol group of threonine,
while a good nucleophile, does not fulfill an analogous
The Solubility and Melting Points role in catalysis. The  OH groups of serine, tyrosine,
of Amino Acids Reflect and threonine also participate in regulation of the activ-
Their Ionic Character ity of enzymes whose catalytic activity depends on the
phosphorylation state of these residues.
The charged functional groups of amino acids ensure
that they are readily solvated by—and thus soluble in—
polar solvents such as water and ethanol but insoluble FUNCTIONAL GROUPS DICTATE THE
in nonpolar solvents such as benzene, hexane, or ether. CHEMICAL REACTIONS OF AMINO ACIDS
Similarly, the high amount of energy required to dis- Each functional group of an amino acid exhibits all of
rupt the ionic forces that stabilize the crystal lattice its characteristic chemical reactions. For carboxylic acid
account for the high melting points of amino acids groups, these reactions include the formation of esters,
(> 200 °C). amides, and acid anhydrides; for amino groups, acyla-
Amino acids do not absorb visible light and thus are
tion, amidation, and esterification; and for  OH and
colorless. However, tyrosine, phenylalanine, and espe-
 SH groups, oxidation and esterification. The most
cially tryptophan absorb high-wavelength (250–290
important reaction of amino acids is the formation of a
nm) ultraviolet light. Tryptophan therefore makes the
major contribution to the ability of most proteins to peptide bond (shaded blue).
absorb light in the region of 280 nm.
+H N
3 O
H
N O–
Table 3–2. Typical range of pKa values for N
ionizable groups in proteins. H
O O
SH

Alanyl Cysteinyl Valine

Amino Acid Sequence Determines


Primary Structure
The number and order of all of the amino acid residues
in a polypeptide constitute its primary structure.
-Amino
Amino acids present in peptides are called aminoacyl
residues and are named by replacing the -ate or -ine suf-
fixes of free amino acids with -yl (eg, alanyl, aspartyl, ty-
rosyl). Peptides are then named as derivatives of the SH
carboxyl terminal aminoacyl residue. For example, Lys-
O CH2 H
Leu-Tyr-Gln is called lysyl-leucyl-tyrosyl-glutamine.
The -ine ending on glutamine indicates that its -car- C CH N
boxyl group is not involved in peptide bond formation. CH2 CH2
N C

CH2 H O COO
Peptide Structures Are Easy to Draw
+
Prefixes like tri- or octa- denote peptides with three or H C NH
eight residues, respectively, not those with three or COO

eight peptide bonds. By convention, peptides are writ-


ten with the residue that bears the free -amino group Figure 3–3. Glutathione (-glutamyl-cysteinyl-
at the left. To draw a peptide, use a zigzag to represent glycine). Note the non- peptide bond that links
the main chain or backbone. Add the main chain atoms, Glu to Cys.
which occur in the repeating order: -nitrogen, -car-
bon, carbonyl carbon. Now add a hydrogen atom to
each -carbon and to each peptide nitrogen, and an
oxygen to the carbonyl carbon. Finally, add the appro- releasing hormone (TRH) is cyclized to pyroglutamic
priate R groups (shaded) to each -carbon atom. acid, and the carboxyl group of the carboxyl terminal
prolyl residue is amidated. Peptides elaborated by fungi,
N C C N C
bacteria, and lower animals can contain nonprotein
amino acids. The antibiotics tyrocidin and gramicidin S
C N C C 
are cyclic polypeptides that contain D-phenylalanine
 and ornithine. The heptapeptide opioids dermorphin
O H
H and deltophorin in the skin of South American tree
COO–
+
H3 N C C N frogs contain D-tyrosine and D-alanine.
C N C C
H H H CH2
CH2 O Peptides Are Polyelectrolytes
–OOC OH
The peptide bond is uncharged at any pH of physiologic
Three-letter abbreviations linked by straight lines interest. Formation of peptides from amino acids is
represent an unambiguous primary structure. Lines are therefore accompanied by a net loss of one positive and
omitted for single-letter abbreviations. one negative charge per peptide bond formed. Peptides
nevertheless are charged at physiologic pH owing to their
Glu- Ala - Lys - Gly - Tyr - Ala carboxyl and amino terminal groups and, where present,
E A K G Y A their acidic or basic R groups. As for amino acids, the net
charge on a peptide depends on the pH of its environ-
Where there is uncertainty about the order of a portion ment and on the pKa values of its dissociating groups.
of a polypeptide, the questionable residues are enclosed
in brackets and separated by commas. The Peptide Bond Has Partial
Glu- Lys - (Ala, Gly, Tyr)- His - Ala Double-Bond Character
Although peptides are written as if a single bond linked
Some Peptides Contain Unusual the -carboxyl and -nitrogen atoms, this bond in fact
exhibits partial double-bond character:
Amino Acids
In mammals, peptide hormones typically contain only O O–

the -amino acids of proteins linked by standard pep- C C +


tide bonds. Other peptides may, however, contain non- N N
protein amino acids, derivatives of the protein amino H H
acids, or amino acids linked by an atypical peptide
bond. For example, the amino terminal glutamate of There thus is no freedom of rotation about the bond
glutathione, which participates in protein folding and that connects the carbonyl carbon and the nitrogen of a
in the metabolism of xenobiotics (Chapter 53), is peptide bond. Consequently, all four of the colored
linked to cysteine by a non- peptide bond (Figure atoms of Figure 3–4 are coplanar. The imposed semi-
3–3). The amino terminal glutamate of thyrotropin- rigidity of the peptide bond has important conse-
O R H O

121 122
C
120
117
110
120 120

0.1 nm
O H R

0.36 nm

Figure 3–4. Dimensions of a fully extended poly-


peptide chain. The four atoms of the peptide bond
(colored blue) are coplanar. The unshaded atoms are
the -carbon atom, the -hydrogen atom, and the -R
group of the particular amino acid. Free rotation can
occur about the bonds that connect the -carbon with
the -nitrogen and with the -carbonyl carbon (blue
arrows). The extended polypeptide chain is thus a semi-
rigid structure with two-thirds of the atoms of the back-
bone held in a fixed planar relationship one to another.
The distance between adjacent -carbon atoms is 0.36
nm (3.6 Å). The interatomic distances and bond angles,
which are not equivalent, are also shown. (Redrawn and
reproduced, with permission, from Pauling L, Corey LP,
Branson HR: The structure of proteins: Two hydrogen-
bonded helical configurations of the polypeptide chain.
Proc Natl Acad Sci U S A 1951;37:205.)
BIOMEDICAL IMPORTANCE Column Chromatography
Proteins perform multiple critically important roles. An Column chromatography of proteins employs as the
internal protein network, the cytoskeleton (Chapter stationary phase a column containing small spherical
49), maintains cellular shape and physical integrity. beads of modified cellulose, acrylamide, or silica whose
Actin and myosin filaments form the contractile ma- surface typically has been coated with chemical func-
chinery of muscle (Chapter 49). Hemoglobin trans- tional groups. These stationary phase matrices interact
ports oxygen (Chapter 6), while circulating antibodies with proteins based on their charge, hydrophobicity,
search out foreign invaders (Chapter 50). Enzymes cat- and ligand-binding properties. A protein mixture is ap-
alyze reactions that generate energy, synthesize and de- plied to the column and the liquid mobile phase is per-
grade biomolecules, replicate and transcribe genes, colated through it. Small portions of the mobile phase
process mRNAs, etc (Chapter 7). Receptors enable cells or eluant are collected as they emerge (Figure 4–1).
to sense and respond to hormones and other environ-
mental cues (Chapters 42 and 43). An important goal
of molecular medicine is the identification of proteins Partition Chromatography
whose presence, absence, or deficiency is associated Column chromatographic separations depend on the
with specific physiologic states or diseases. The primary relative affinity of different proteins for a given station-
sequence of a protein provides both a molecular finger- ary phase and for the mobile phase. Association be-
print for its identification and information that can be tween each protein and the matrix is weak and tran-
used to identify and clone the gene or genes that en- sient. Proteins that interact more strongly with the
code it. stationary phase are retained longer. The length of time
that a protein is associated with the stationary phase is a
function of the composition of both the stationary and
PROTEINS & PEPTIDES MUST BE mobile phases. Optimal separation of the protein of in-
terest from other proteins thus can be achieved by care-
PURIFIED PRIOR TO ANALYSIS ful manipulation of the composition of the two phases.
Highly purified protein is essential for determination of
its amino acid sequence. Cells contain thousands of dif-
ferent proteins, each in widely varying amounts. The
Size Exclusion Chromatography
isolation of a specific protein in quantities sufficient for Size exclusion—or gel filtration—chromatography sep-
analysis thus presents a formidable challenge that may arates proteins based on their Stokes radius, the diam-
require multiple successive purification techniques. eter of the sphere they occupy as they tumble in solu-
Classic approaches exploit differences in relative solu- tion. The Stokes radius is a function of molecular mass
bility of individual proteins as a function of pH (iso- and shape. A tumbling elongated protein occupies a
electric precipitation), polarity (precipitation with larger volume than a spherical protein of the same mass.
ethanol or acetone), or salt concentration (salting out Size exclusion chromatography employs porous beads
with ammonium sulfate). Chromatographic separations (Figure 4–2). The pores are analogous to indentations
partition molecules between two phases, one mobile in a river bank. As objects move downstream, those that
and the other stationary. For separation of amino acids enter an indentation are retarded until they drift back
or sugars, the stationary phase, or matrix, may be a into the main current. Similarly, proteins with Stokes
sheet of filter paper (paper chromatography) or a thin radii too large to enter the pores (excluded proteins) re-
layer of cellulose, silica, or alumina (thin-layer chro- main in the flowing mobile phase and emerge before
matography; TLC). proteins that can enter the pores (included proteins).

21
22 /

Figure 4–1. Components of a simple liquid chromatography apparatus. R: Reser-


voir of mobile phase liquid, delivered either by gravity or using a pump. C: Glass or
plastic column containing stationary phase. F: Fraction collector for collecting por-
tions, called fractions, of the eluant liquid in separate test tubes.

Ion Exchange Chromatography


Absorption Chromatography
In ion exchange chromatography, proteins interact with
For absorption chromatography, the protein mixture is the stationary phase by charge-charge interactions. Pro-
applied to a column under conditions where the pro- teins with a net positive charge at a given pH adhere to
tein of interest associates with the stationary phase so beads with negatively charged functional groups such as
tightly that its partition coefficient is essentially unity. carboxylates or sulfates (cation exchangers). Similarly,
Nonadhering molecules are first eluted and discarded. proteins with a net negative charge adhere to beads with
Proteins are then sequentially released by disrupting the positively charged functional groups, typically tertiary or
forces that stabilize the protein-stationary phase com- quaternary amines (anion exchangers). Proteins, which
plex, most often by using a gradient of increasing salt are polyanions, compete against monovalent ions for
concentration. The composition of the mobile phase is binding to the support—thus the term ―ion exchange.‖
altered gradually so that molecules are selectively re- For example, proteins bind to diethylaminoethyl
leased in descending order of their affinity for the sta- (DEAE) cellulose by replacing the counter-ions (gener-
tionary phase. ally Cl or CH3COO) that neutralize the protonated
amine. Bound proteins are selectively displaced by grad-
ually raising the concentration of monovalent ions in
A B C

Figure 4–2. Size-exclusion chromatography. A: A mixture of large molecules


(diamonds) and small molecules (circles) are applied to the top of a gel filtration
column. B: Upon entering the column, the small molecules enter pores in the sta-
tionary phase matrix from which the large molecules are excluded. C: As the mo-
bile phase flows down the column, the large, excluded molecules flow with it
while the small molecules, which are temporarily sheltered from the flow when in-
side the pores, lag farther and farther behind.

the mobile phase. Proteins elute in inverse order of the fied by affinity chromatography using immobilized sub-
strength of their interactions with the stationary phase. strates, products, coenzymes, or inhibitors. In theory,
Since the net charge on a protein is determined by only proteins that interact with the immobilized ligand
the pH (see Chapter 3), sequential elution of proteins adhere. Bound proteins are then eluted either by compe-
may be achieved by changing the pH of the mobile tition with soluble ligand or, less selectively, by disrupt-
phase. Alternatively, a protein can be subjected to con- ing protein-ligand interactions using urea, guanidine
secutive rounds of ion exchange chromatography, each hydrochloride, mildly acidic pH, or high salt concentra-
at a different pH, such that proteins that co-elute at one tions. Stationary phase matrices available commercially
pH elute at different salt concentrations at another pH. contain ligands such as NAD+ or ATP analogs. Among
the most powerful and widely applicable affinity matri-
Hydrophobic Interaction Chromatography ces are those used for the purification of suitably modi-
fied recombinant proteins. These include a Ni 2+ matrix
Hydrophobic interaction chromatography separates that binds proteins with an attached polyhistidine ―tag‖
proteins based on their tendency to associate with a sta- and a glutathione matrix that binds a recombinant pro-
tionary phase matrix coated with hydrophobic groups tein linked to glutathione S-transferase.
(eg, phenyl Sepharose, octyl Sepharose). Proteins with
exposed hydrophobic surfaces adhere to the matrix via
hydrophobic interactions that are enhanced by a mobile Peptides Are Purified by Reversed-Phase
phase of high ionic strength. Nonadherent proteins are High-Pressure Chromatography
first washed away. The polarity of the mobile phase is
then decreased by gradually lowering the salt concentra- The stationary phase matrices used in classic column
tion. If the interaction between protein and stationary chromatography are spongy materials whose compress-
phase is particularly strong, ethanol or glycerol may be ibility limits flow of the mobile phase. High-pressure liq-
added to the mobile phase to decrease its polarity and uid chromatography (HPLC) employs incompressible
further weaken hydrophobic interactions. silica or alumina microbeads as the stationary phase and
pressures of up to a few thousand psi. Incompressible
matrices permit both high flow rates and enhanced reso-
Affinity Chromatography lution. HPLC can resolve complex mixtures of lipids or
Affinity chromatography exploits the high selectivity of peptides whose properties differ only slightly. Reversed-
most proteins for their ligands. Enzymes may be puri- phase HPLC exploits a hydrophobic stationary phase of
24 /

aliphatic polymers 3–18 carbon atoms in length. Peptide through the acrylamide matrix determines the rate of
mixtures are eluted using a gradient of a water-miscible migration. Since large complexes encounter greater re-
organic solvent such as acetonitrile or methanol. sistance, polypeptides separate based on their relative
molecular mass (Mr). Individual polypeptides trapped
Protein Purity Is Assessed by in the acrylamide gel are visualized by staining with
Polyacrylamide Gel Electrophoresis dyes such as Coomassie blue (Figure 4–4).
(PAGE)
Isoelectric Focusing (IEF)
The most widely used method for determining the pu-
rity of a protein is SDS-PAGE—polyacrylamide gel Ionic buffers called ampholytes and an applied electric
electrophoresis (PAGE) in the presence of the anionic field are used to generate a pH gradient within a poly-
detergent sodium dodecyl sulfate (SDS). Electrophore- acrylamide matrix. Applied proteins migrate until they
sis separates charged biomolecules based on the rates at reach the region of the matrix where the pH matches
which they migrate in an applied electrical field. For their isoelectric point (pI), the pH at which a peptide’s
SDS-PAGE, acrylamide is polymerized and cross- net charge is zero. IEF is used in conjunction with SDS-
linked to form a porous matrix. SDS denatures and PAGE for two-dimensional electrophoresis, which sepa-
binds to proteins at a ratio of one molecule of SDS per rates polypeptides based on pI in one dimension and
two peptide bonds. When used in conjunction with 2- based on Mr in the second (Figure 4–5). Two-dimen-
mercaptoethanol or dithiothreitol to reduce and break sional electrophoresis is particularly well suited for sepa-
disulfide bonds (Figure 4 –3), SDS separates the com- rating the components of complex mixtures of proteins.
ponent polypeptides of multimeric proteins. The large
number of anionic SDS molecules, each bearing a SANGER WAS THE FIRST TO DETERMINE
charge of 1, on each polypeptide overwhelms the
charge contributions of the amino acid functional
THE SEQUENCE OF A POLYPEPTIDE
groups. Since the charge-to-mass ratio of each SDS- Mature insulin consists of the 21-residue A chain and
polypeptide complex is approximately equal, the physi- the 30-residue B chain linked by disulfide bonds. Fred-
cal resistance each peptide encounters as it moves erick Sanger reduced the disulfide bonds (Figure 4–3),

NH
O HN
S
HN S
H
NH
O

O SH

HCOOH C2H5

OH

NH
O
HN
SO2
HN
HS H
NH
O
Figure 4–4. Use of SDS-PAGE to observe successive
purification of a recombinant protein. The gel was
Figure 4–3. Oxidative cleavage of adjacent polypep- stained with Coomassie blue. Shown are protein stan-
tide chains linked by disulfide bonds (shaded) by per- dards (lane S) of the indicated mass, crude cell extract
formic acid (left) or reductive cleavage by -mercap- (E), high-speed supernatant liquid (H), and the DEAE-
toethanol (right) forms two peptides that contain Sepharose fraction (D). The recombinant protein has a
cysteic acid residues or cysteinyl residues, respectively. mass of about 45 kDa.
pH = 3 pH = 10
IEF

SDS
PAGE
Figure 4–5. Two-dimensional IEF-SDS-PAGE. The
gel was stained with Coomassie blue. A crude bacter-
ial extract was first subjected to isoelectric focusing
(IEF) in a pH 3–10 gradient. The IEF gel was then
placed horizontally on the top of an SDS gel, and the
proteins then further resolved by SDS-PAGE. Notice
the greatly improved resolution of distinct polypep-
tides relative to ordinary SDS-
PAGE gel (Figure 4–4).

separated the A and B chains, and cleaved each chain Large Polypeptides Are First Cleaved Into
into smaller peptides using trypsin, chymotrypsin, and Smaller Segments
pepsin. The resulting peptides were then isolated and
treated with acid to hydrolyze peptide bonds and gener- While the first 20–30 residues of a peptide can readily
ate peptides with as few as two or three amino acids. be determined by the Edman method, most polypep-
Each peptide was reacted with 1-fluoro-2,4-dinitroben- tides contain several hundred amino acids. Conse-
zene (Sanger’s reagent), which derivatizes the exposed quently, most polypeptides must first be cleaved into
-amino group of amino terminal residues. The amino smaller peptides prior to Edman sequencing. Cleavage
acid content of each peptide was then determined. also may be necessary to circumvent posttranslational
While the -amino group of lysine also reacts with modifications that render a protein’s -amino group
Sanger’s reagent, amino-terminal lysines can be distin- ―blocked‖, or unreactive with the Edman reagent.
guished from those at other positions because they react It usually is necessary to generate several peptides
with 2 mol of Sanger’s reagent. Working backwards to using more than one method of cleavage. This reflects
larger fragments enabled Sanger to determine the com- both inconsistency in the spacing of chemically or enzy-
plete sequence of insulin, an accomplishment for which matically susceptible cleavage sites and the need for sets
he received a Nobel Prize in 1958. of peptides whose sequences overlap so one can infer
the sequence of the polypeptide from which they derive
(Figure 4–7). Reagents for the chemical or enzymatic
THE EDMAN REACTION ENABLES cleavage of proteins include cyanogen bromide (CNBr),
PEPTIDES & PROTEINS trypsin, and Staphylococcus aureus V8 protease (Table
TO BE SEQUENCED 4–1). Following cleavage, the resulting peptides are pu-
rified by reversed-phase HPLC—or occasionally by
Pehr Edman introduced phenylisothiocyanate (Edman’s SDS-PAGE—and sequenced.
reagent) to selectively label the amino-terminal residue
of a peptide. In contrast to Sanger’s reagent, the
phenylthiohydantoin (PTH) derivative can be removed MOLECULAR BIOLOGY HAS
under mild conditions to generate a new amino terminal REVOLUTIONIZED THE DETERMINATION
residue (Figure 4–6). Successive rounds of derivatization OF PRIMARY STRUCTURE
with Edman’s reagent can therefore be used to sequence
many residues of a single sample of peptide. Edman se- Knowledge of DNA sequences permits deduction of
quencing has been automated, using a thin film or solid the primary structures of polypeptides. DNA sequenc-
matrix to immobilize the peptide and HPLC to identify ing requires only minute amounts of DNA and can
PTH amino acids. Modern gas-phase sequencers can readily yield the sequence of hundreds of nucleotides.
analyze as little as a few picomoles of peptide. To clone and sequence the DNA that encodes a partic-
26 /

S Peptide X Peptide Y
C Peptide Z
N
+
O H NH2 Carboxyl terminal Amino terminal
N portion of portion of
N R peptide X peptide Y
H
R
Figure 4–7. The overlapping peptide Z is used to de-
Phenylisothiocyanate (Edman reagent) duce that peptides X and Y are present in the original
and a peptide protein in the order X  Y, not Y  X.

sequence can be determined and the genetic code used


to infer the primary structure of the encoded poly-
S peptide.
The hybrid approach enhances the speed and effi-
N NH ciency of primary structure analysis and the range of
H
proteins that can be sequenced. It also circumvents ob-
O H stacles such as the presence of an amino-terminal block-
N ing group or the lack of a key overlap peptide. Only a
N R
H few segments of primary structure must be determined
R
by Edman analysis.
A phenylthiohydantoic acid DNA sequencing reveals the order in which amino
acids are added to the nascent polypeptide chain as it is
H+, nitro- H2O synthesized on the ribosomes. However, it provides no
methane information about posttranslational modifications such
as proteolytic processing, methylation, glycosylation,
S O phosphorylation, hydroxylation of proline and lysine,
NH2 and disulfide bond formation that accompany matura-
NH + N
H
tion. While Edman sequencing can detect the presence
R of most posttranslational events, technical limitations
O often prevent identification of a specific modification.
A phenylthiohydantoin and a peptide
shorter by one residue
Table 4–1. Methods for cleaving polypeptides.
Figure 4–6. The Edman reaction. Phenylisothio-
cyanate derivatizes the amino-terminal residue of a Method Bond Cleaved
peptide as a phenylthiohydantoic acid. Treatment with
acid in a nonhydroxylic solvent releases a phenylthio- CNBr Met-X
hydantoin, which is subsequently identified by its chro- Trypsin Lys-X and Arg-X
matographic mobility, and a peptide one residue
Chymotrypsin Hydrophobic amino acid-X
shorter. The process is then repeated.
Endoproteinase Lys-C Lys-X
Endoproteinase Arg-C Arg-X
ular protein, some means of identifying the correct
clone—eg, knowledge of a portion of its nucleotide se- Endoproteinase Asp-N X-Asp
quence—is essential. A hybrid approach thus has V8 protease Glu-X, particularly where X is hydro-
emerged. Edman sequencing is used to provide a partial phobic
amino acid sequence. Oligonucleotide primers modeled
on this partial sequence can then be used to identify Hydroxylamine Asn-Gly
clones or to amplify the appropriate gene by the poly- o-Iodosobenzene Trp-X
merase chain reaction (PCR) (see Chapter 40). Once an
authentic DNA clone is obtained, its oligonucleotide Mild acid Asp-Pro
MASS SPECTROMETRY DETECTS
Figure 4–8. Basic components of a simple mass
COVALENT MODIFICATIONS
spectrometer. A mixture of molecules is vaporized in an
Mass spectrometry, which discriminates molecules ionized state in the sample chamber S. These mole-
based solely on their mass, is ideal for detecting the cules are then accelerated down the flight tube by an
phosphate, hydroxyl, and other groups on posttransla- electrical potential applied to accelerator grid A. An
tionally modified amino acids. Each adds a specific and ad-justable electromagnet, E, applies a magnetic field
readily identified increment of mass to the modified thatdeflects the flight of the individual ions until they
amino acid (Table 4–2). For analysis by mass spec- strike the detector, D. The greater the mass of the ion,
trometry, a sample in a vacuum is vaporized under the higher the magnetic field required to focus it onto
conditions where protonation can occur, imparting the detector.
positive charge. An electrical field then propels the
cations through a magnetic field which deflects them
at a right angle to their original direction of flight and
focuses them onto a detector (Figure 4–8). The mag-
netic force required to deflect the path of each ionic phase HPLC column are introduced directly into the
species onto the detector, measured as the current ap- mass spectrometer for immediate determination of
plied to the electromagnet, is recorded. For ions of their masses.
identical net charge, this force is proportionate to their Peptides inside the mass spectrometer are broken
mass. In a time-of-flight mass spectrometer, a briefly down into smaller units by collisions with neutral he-
applied electric field accelerates the ions towards a lium atoms (collision-induced dissociation), and the
de- tector that records the time at which each ion masses of the individual fragments are determined.
arrives. For molecules of identical charge, the velocity to Since peptide bonds are much more labile than carbon-
which they are accelerated—and hence the time carbon bonds, the most abundant fragments will differ
required to reach the detector—will be inversely from one another by units equivalent to one or two
proportionate to their mass. amino acids. Since—with the exception of leucine and
Conventional mass spectrometers generally are used isoleucine—the molecular mass of each amino acid is
to determine the masses of molecules of 1000 Da or unique, the sequence of the peptide can be recon-
less, whereas time-of-flight mass spectrometers are structed from the masses of its fragments.
suited for determining the large masses of proteins.
The analysis of peptides and proteins by mass spec-
tometry initially was hindered by difficulties in Two-Dimensional Electrophoresis &
volatilizing large organic molecules. However, matrix-
assisted laser-desorption (MALDI) and electrospray Gene Array Chips Are Used to Survey
dispersion (eg, nanospray) permit the masses of even Protein Expression
large polypeptides (> 100,000 Da) to be determined One goal of proteomics is the identification of proteins
with extraordinary accuracy (± 1 Da). Using electro- whose levels of expression correlate with medically sig-
spray dispersion, peptides eluting from a reversed- nificant events. The presumption is that proteins whose
appearance or disappearance is associated with a specific
physiologic condition or disease will provide insights
Table 4–2. Mass increases resulting from into root causes and mechanisms. Determination of the
common posttranslational modifications. proteomes characteristic of each cell type requires the
utmost efficiency in the isolation and identification of
individual proteins. The contemporary approach uti-
Modification Mass Increase (Da) lizes robotic automation to speed sample preparation
Phosphorylation 80 and large two-dimensional gels to resolve cellular pro-
teins. Individual polypeptides are then extracted and
Hydroxylation 16 analyzed by Edman sequencing or mass spectroscopy.
Methylation 14 While only about 1000 proteins can be resolved on a
single gel, two-dimensional electrophoresis has a major
Acetylation 42
advantage in that it examines the proteins themselves.
Myristylation 210 An alternative and complementary approach employs
Palmitoylation 238 gene arrays, sometimes called DNA chips, to detect the
expression of the mRNAs which encode proteins.
Glycosylation 162
Higher Orders of Protein Structures 3
BIOMEDICAL IMPORTANCE Globular proteins are compact, are roughly spherical
or ovoid in shape, and have axial ratios (the ratio of
Proteins catalyze metabolic reactions, power cellular their shortest to longest dimensions) of not over 3.
motion, and form macromolecular rods and cables that Most enzymes are globular proteins, whose large inter-
provide structural integrity to hair, bones, tendons, and nal volume provides ample space in which to con-
teeth. In nature, form follows function. The structural struct cavities of the specific shape, charge, and hy-
variety of human proteins therefore reflects the sophis- drophobicity or hydrophilicity required to bind
tication and diversity of their biologic roles. Maturation substrates and promote catalysis. By contrast, many
of a newly synthesized polypeptide into a biologically structural proteins adopt highly extended conforma-
functional protein requires that it be folded into a spe- tions. These fibrous proteins possess axial ratios of 10
cific three-dimensional arrangement, or conformation. or more.
During maturation, posttranslational modifications Lipoproteins and glycoproteins contain covalently
may add new chemical groups or remove transiently bound lipid and carbohydrate, respectively. Myoglobin,
needed peptide segments. Genetic or nutritional defi- hemoglobin, cytochromes, and many other proteins
ciencies that impede protein maturation are deleterious contain tightly associated metal ions and are termed
to health. Examples of the former include Creutzfeldt- metalloproteins. With the development and applica-
Jakob disease, scrapie, Alzheimer’s disease, and bovine tion of techniques for determining the amino acid se-
spongiform encephalopathy (mad cow disease). Scurvy quences of proteins (Chapter 4), more precise classifica-
represents a nutritional deficiency that impairs protein tion schemes have emerged based upon similarity, or
maturation. homology, in amino acid sequence and structure.
However, many early classification terms remain in
CONFORMATION VERSUS common use.
CONFIGURATION
The terms configuration and conformation are often
confused. Configuration refers to the geometric rela- PROTEINS ARE CONSTRUCTED USING
tionship between a given set of atoms, for example, MODULAR PRINCIPLES
those that distinguish L- from D-amino acids. Intercon-
version of configurational alternatives requires breaking Proteins perform complex physical and catalytic func-
covalent bonds. Conformation refers to the spatial re- tions by positioning specific chemical groups in a pre-
relationship of every atom in a molecule. cise three-dimensional arrangement. The polypeptide
Interconversion between conformers occurs without scaffold containing these groups must adopt a confor-
covalent bond rup- ture, with retention of mation that is both functionally efficient and phys-
configuration, and typically via rotation about single ically strong. At first glance, the biosynthesis of
bonds. polypeptides comprised of tens of thousands of indi-
vidual atoms would appear to be extremely challeng-
ing. When one considers that a typical polypeptide
PROTEINS WERE INITIALLY CLASSIFIED can adopt  1050 distinct conformations, folding into
BY THEIR GROSS CHARACTERISTICS the conformation appropriate to their biologic func-
tion would appear to be even more difficult. As de-
Scientists initially approached structure-function rela- scribed in Chapters 3 and 4, synthesis of the polypep-
tionships in proteins by separating them into classes tide backbones of proteins employs a small set of
based upon properties such as solubility, shape, or the common building blocks or modules, the amino acids,
presence of nonprotein groups. For example, the pro- joined by a common linkage, the peptide bond. A
teins that can be extracted from cells using solutions at stepwise modular pathway simplifies the folding and
physiologic pH and ionic strength are classified as sol- processing of newly synthesized polypeptides into ma-
uble. Extraction of integral membrane proteins re- ture proteins.
quires dissolution of the membrane with detergents.
30
THE FOUR ORDERS OF
PROTEIN STRUCTURE
The modular nature of protein synthesis and folding
are embodied in the concept of orders of protein struc-
ture: primary structure, the sequence of the amino
acids in a polypeptide chain; secondary structure, the
folding of short (3- to 30-residue), contiguous segments
of polypeptide into geometrically ordered units; ter-
tiary structure, the three-dimensional assembly of sec-
ondary structural units to form larger functional units
such as the mature polypeptide and its component do-
mains; and quaternary structure, the number and
types of polypeptide units of oligomeric proteins and
their spatial arrangement. 

SECONDARY STRUCTURE
Peptide Bonds Restrict Possible
Secondary Conformations
Free rotation is possible about only two of the three co-
valent bonds of the polypeptide backbone: the -car- occur in nature. Schematic diagrams of proteins
bon (C) to the carbonyl carbon (Co) bond and the
C to nitrogen bond (Figure 3–4). The partial double- repre-sent  helices as cylinders.
bond character of the peptide bond that links Co to the The stability of an  helix arises primarily from hy-
-nitrogen requires that the carbonyl carbon, carbonyl drogen bonds formed between the oxygen of the pep-
oxygen, and -nitrogen remain coplanar, thus prevent- tide bond carbonyl and the hydrogen atom of the pep-
ing rotation. The angle about the CN bond is tide bond nitrogen of the fourth residue down the
termed the phi () angle, and that about the CoC polypeptide chain (Figure 5–4). The ability to form the
bond the psi () angle. For amino acids other than maximum number of hydrogen bonds, supplemented
glycine, most combinations of phi and psi angles are by van der Waals interactions in the core of this tightly
disallowed because of steric hindrance (Figure 5–1). packed structure, provides the thermodynamic driving
The conformations of proline are even more restricted force for the formation of an  helix. Since the peptide
due to the absence of free rotation of the NC bond. bond nitrogen of proline lacks a hydrogen atom to con-
Regions of ordered secondary structure arise when a tribute to a hydrogen bond, proline can only be stably
series of aminoacyl residues adopt similar phi and psi accommodated within the first turn of an  helix.
angles. Extended segments of polypeptide (eg, loops) When present elsewhere, proline disrupts the confor-
can possess a variety of such angles. The angles that de- mation of the helix, producing a bend. Because of its
fine the two most common types of secondary struc- small size, glycine also often induces bends in  helices.
ture, the a helix and the β sheet, fall within the lower Many  helices have predominantly hydrophobic R
and upper left-hand quadrants of a Ramachandran groups on one side of the axis of the helix and predomi-
plot, respectively (Figure 5–1). nantly hydrophilic ones on the other. These amphi-
pathic helices are well adapted to the formation of in-
The Alpha Helix terfaces between polar and nonpolar regions such as the
hydrophobic interior of a protein and its aqueous envi-
The polypeptide backbone of an  helix is twisted by
an equal amount about each -carbon with a phi angle
of approximately 57 degrees and a psi angle of approx-
imately  47 degrees. A complete turn of the helix con-
tains an average of 3.6 aminoacyl residues, and the dis-
tance it rises per turn (its pitch) is 0.54 nm (Figure
5–2). The R groups of each aminoacyl residue in an 
helix face outward (Figure 5–3). Proteins contain only
L-amino acids, for which a right-handed  helix is by
far the more stable, and only right-handed  helices
32 /

R R

Figure 5–3. View down the axis of an  helix. The


C side chains (R) are on the outside of the helix. The van
N der Waals radii of the atoms are larger than shown here;
C hence, there is almost no free space inside the helix.
C (Slightly modified and reproduced, with permission, from
0.54-nm pitch Stryer L: Biochemistry, 3rd ed. Freeman, 1995. Copyright
(3.6 residues)
© 1995 by W.H. Freeman and Co.)
N
0.15 nm
polypeptide chain proceed in the same direction amino
to carboxyl, or an antiparallel sheet, in which they pro-
ceed in opposite directions (Figure 5–5). Either config-
uration permits the maximum number of hydrogen
bonds between segments, or strands, of the sheet. Most
Figure 5–2. Orientation of the main chain atoms of a  sheets are not perfectly flat but tend to have a right-
peptide about the axis of an  helix. handed twist. Clusters of twisted strands of  sheet
form the core of many globular proteins (Figure 5–6).
Schematic diagrams represent  sheets as arrows that
ronment. Clusters of amphipathic helices can create a point in the amino to carboxyl terminal direction.
channel, or pore, that permits specific polar molecules
to pass through hydrophobic cell membranes.
Loops & Bends
Roughly half of the residues in a ―typical‖ globular pro-
The Beta Sheet
tein reside in  helices and  sheets and half in loops,
The second (hence ―beta‖) recognizable regular sec- turns, bends, and other extended conformational fea-
ondary structure in proteins is the  sheet. The amino tures. Turns and bends refer to short segments of
acid residues of a  sheet, when viewed edge-on, form a amino acids that join two units of secondary structure,
zigzag or pleated pattern in which the R groups of adja- such as two adjacent strands of an antiparallel  sheet.
cent residues point in opposite directions. Unlike the A  turn involves four aminoacyl residues, in which the
compact backbone of the  helix, the peptide backbone first residue is hydrogen-bonded to the fourth, resulting
of the  sheet is highly extended. But like the  helix, in a tight 180-degree turn (Figure 5–7). Proline and
 sheets derive much of their stability from hydrogen glycine often are present in  turns.
bonds between the carbonyl oxygens and amide hydro- Loops are regions that contain residues beyond the
gens of peptide bonds. However, in contrast to the  minimum number necessary to connect adjacent re-
helix, these bonds are formed with adjacent segments of gions of secondary structure. Irregular in conformation,
 sheet (Figure 5–5). loops nevertheless serve key biologic roles. For many
Interacting  sheets can be arranged either to form a enzymes, the loops that bridge domains responsible for
parallel  sheet, in which the adjacent segments of the binding substrates often contain aminoacyl residues
C R

N O

R C

Figure 5–5. Spacing and bond angles of the hydro-


gen bonds of antiparallel and parallel pleated  sheets.
Arrows indicate the direction of each strand. The hydro-
gen-donating -nitrogen atoms are shown as blue cir-
cles. Hydrogen bonds are indicated by dotted lines. For
Figure 5–4. Hydrogen bonds (dotted lines) formed clarity in presentation, R groups and hydrogens are
between H and O atoms stabilize a polypeptide in an omitted. Top: Antiparallel  sheet. Pairs of hydrogen
-helical conformation. (Reprinted, with permission, bonds alternate between being close together and
from Haggis GH et al: Introduction to Molecular Biology. wide apart and are oriented approximately perpendicu-
Wiley, 1964.) lar to the polypeptide backbone. Bottom: Parallel 
sheet. The hydrogen bonds are evenly spaced but slant
in alternate directions.
that participate in catalysis. Helix-loop-helix motifs
provide the oligonucleotide-binding portion of DNA-
binding proteins such as repressors and transcription
factors. Structural motifs such as the helix-loop-helix dered regions assume an ordered conformation upon
motif that are intermediate between secondary and ter- binding of a ligand. This structural flexibility enables
tiary structures are often termed supersecondary struc- such regions to act as ligand-controlled switches that af-
tures. Since many loops and bends reside on the surface fect protein structure and function.
of proteins and are thus exposed to solvent, they consti-
tute readily accessible sites, or epitopes, for recognition
and binding of antibodies.
Tertiary & Quaternary Structure
While loops lack apparent structural regularity, they The term ―tertiary structure‖ refers to the entire three-
exist in a specific conformation stabilized through hy- dimensional conformation of a polypeptide. It indicates,
drogen bonding, salt bridges, and hydrophobic interac- in three-dimensional space, how secondary structural
tions with other portions of the protein. However, not features—helices, sheets, bends, turns, and loops—
all portions of proteins are necessarily ordered. Proteins assemble to form domains and how these domains re-
may contain ―disordered‖ regions, often at the extreme late spatially to one another. A domain is a section of
amino or carboxyl terminal, characterized by high con- protein structure sufficient to perform a particular
formational flexibility. In many instances, these disor- chemical or physical task such as binding of a substrate
34 /

COOH
H
H CH2
N
H C H
 C

H N O C O

CH3 C O H N
CH2OH
C C
H H

Figure 5–7. A -turn that links two segments of an-


tiparallel  sheet. The dotted line indicates the hydro-
gen bond between the first and fourth amino acids of
the four-residue segment Ala-Gly-Asp-Ser.

30
15
or other ligand. Other domains may anchor a protein to
55 a membrane or interact with a regulatory molecule that
N modulates its function. A small polypeptide such as
50
345 80 70 triose phosphate isomerase (Figure 5–6) or myoglobin
330 280 (Chapter 6) may consist of a single domain. By contrast,
90
protein kinases contain two domains. Protein kinases
150
185 350 catalyze the transfer of a phosphoryl group from ATP to
145 C
a peptide or protein. The amino terminal portion of the
230 377 polypeptide, which is rich in  sheet, binds ATP, while
the carboxyl terminal domain, which is rich in  helix,
310 245

110 320
220 260 binds the peptide or protein substrate (Figure 5–8). The
groups that catalyze phosphoryl transfer reside in a loop
300 258 positioned at the interface of the two domains.
205
120 In some cases, proteins are assembled from more
than one polypeptide, or protomer. Quaternary struc-
170
125
ture defines the polypeptide composition of a protein
and, for an oligomeric protein, the spatial relationships
Figure 5–6. Examples of tertiary structure of pro- between its subunits or protomers. Monomeric pro-
teins. Top: The enzyme triose phosphate isomerase. teins consist of a single polypeptide chain. Dimeric
Note the elegant and symmetrical arrangement of al- proteins contain two polypeptide chains. Homodimers
ternating  sheets and  helices. (Courtesy of J Richard- contain two copies of the same polypeptide chain,
son.) Bottom: Two-domain structure of the subunit of a
while in a heterodimer the polypeptides differ. Greek
letters (, ,  etc) are used to distinguish different sub-
homodimeric enzyme, a bacterial class II HMG-CoA re-
units of a heterooligomeric protein, and subscripts indi-
ductase. As indicated by the numbered residues, the
cate the number of each subunit type. For example, 4
single polypeptide begins in the large domain, enters
designates a homotetrameric protein, and 22 a pro-
the small domain, and ends in the large domain. (Cour- tein with five subunits of three different types.
tesy of C Lawrence, V Rodwell, and C Stauffacher, Purdue Since even small proteins contain many thousands
University.) of atoms, depictions of protein structure that indicate
the position of every atom are generally too complex to
be readily interpreted. Simplified schematic diagrams
thus are used to depict key features of a protein’s ter-
from water. Other significant contributors include hy-
drogen bonds and salt bridges between the carboxylates
of aspartic and glutamic acid and the oppositely
charged side chains of protonated lysyl, argininyl, and
histidyl residues. While individually weak relative to a
typical covalent bond of 80–120 kcal/mol, collectively
these numerous interactions confer a high degree of sta-
bility to the biologically functional conformation of a
protein, just as a Velcro fastener harnesses the cumula-
tive strength of multiple plastic loops and hooks.
Some proteins contain covalent disulfide (S S)
bonds that link the sulfhydryl groups of cysteinyl
residues. Formation of disulfide bonds involves oxida-
tion of the cysteinyl sulfhydryl groups and requires oxy-
gen. Intrapolypeptide disulfide bonds further enhance
the stability of the folded conformation of a peptide,
while interpolypeptide disulfide bonds stabilize the
quaternary structure of certain oligomeric proteins.

THREE-DIMENSIONAL STRUCTURE
IS DETERMINED BY X-RAY
CRYSTALLOGRAPHY OR BY
NMR SPECTROSCOPY
X-Ray Crystallography
Since the determination of the three-dimensional struc-
ture of myoglobin over 40 years ago, the three-dimen-
sional structures of thousands of proteins have been de-
Figure 5–8. Domain structure. Protein kinases con- termined by x-ray crystallography. The key to x-ray
tain two domains. The upper, amino terminal domain crystallography is the precipitation of a protein under
binds the phosphoryl donor ATP (light blue). The lower, conditions in which it forms ordered crystals that dif-
carboxyl terminal domain is shown binding a synthetic fract x-rays. This is generally accomplished by exposing
peptide substrate (dark blue).
small drops of the protein solution to various combina-
tions of pH and precipitating agents such as salts and
organic solutes such as polyethylene glycol. A detailed
three-dimensional structure of a protein can be con-
tiary and quaternary structure. Ribbon diagrams (Fig- structed from its primary structure using the pattern by
ures 5–6 and 5–8) trace the conformation of the which it diffracts a beam of monochromatic x-rays.
polypeptide backbone, with cylinders and arrows indi- While the development of increasingly capable com-
cating regions of  helix and  sheet, respectively. In an puter-based tools has rendered the analysis of complex
even simpler representation, line segments that link the x-ray diffraction patterns increasingly facile, a major
 carbons indicate the path of the polypeptide back- stumbling block remains the requirement of inducing
bone. These schematic diagrams often include the side highly purified samples of the protein of interest to
chains of selected amino acids that emphasize specific crystallize. Several lines of evidence, including the abil-
structure-function relationships. ity of some crystallized enzymes to catalyze chemical re-
actions, indicate that the vast majority of the structures
MULTIPLE FACTORS STABILIZE determined by crystallography faithfully represent the
TERTIARY & QUATERNARY STRUCTURE structures of proteins in free solution.

Higher orders of protein structure are stabilized primar- Nuclear Magnetic Resonance
ily—and often exclusively—by noncovalent interac-
tions. Principal among these are hydrophobic interac-
Spectroscopy
tions that drive most hydrophobic amino acid side Nuclear magnetic resonance (NMR) spectroscopy, a
chains into the interior of the protein, shielding them powerful complement to x-ray crystallography, mea-
36 /

sures the absorbance of radio frequency electromagnetic Folding Is Modular


energy by certain atomic nuclei. ―NMR-active‖ isotopes
of biologically relevant atoms include 1H, 13C, 15N, and Protein folding generally occurs via a stepwise process.
31
P. The frequency, or chemical shift, at which a partic- In the first stage, the newly synthesized polypeptide
ular nucleus absorbs energy is a function of both the emerges from ribosomes, and short segments fold into
functional group within which it resides and the prox- secondary structural units that provide local regions of
imity of other NMR-active nuclei. Two-dimensional organized structure. Folding is now reduced to the se-
NMR spectroscopy permits a three-dimensional repre- lection of an appropriate arrangement of this relatively
sentation of a protein to be constructed by determining small number of secondary structural elements. In the
the proximity of these nuclei to one another. NMR second stage, the forces that drive hydrophobic regions
spectroscopy analyzes proteins in aqueous solution, ob- into the interior of the protein away from solvent drive
viating the need to form crystals. It thus is possible to the partially folded polypeptide into a ―molten globule‖
observe changes in conformation that accompany lig- in which the modules of secondary structure rearrange
and binding or catalysis using NMR spectroscopy. to arrive at the mature conformation of the protein.
However, only the spectra of relatively small proteins, This process is orderly but not rigid. Considerable flexi-
 20 kDa in size, can be analyzed with current tech- bility exists in the ways and in the order in which ele-
nology. ments of secondary structure can be rearranged. In gen-
eral, each element of secondary or supersecondary
structure facilitates proper folding by directing the fold-
Molecular Modeling ing process toward the native conformation and away
An increasingly useful adjunct to the empirical determi- from unproductive alternatives. For oligomeric pro-
nation of the three-dimensional structure of proteins is teins, individual protomers tend to fold before they as-
the use of computer technology for molecular model- sociate with other subunits.
ing. The types of models created take two forms. In the
first, the known three-dimensional structure of a pro-
tein is used as a template to build a model of the proba- Chaperones
ble structure of a homologous protein. In the second, Chaperone proteins participate in the folding of over
computer software is used to manipulate the static half of mammalian proteins. The hsp70 (70-kDa heat
model provided by crystallography to explore how a shock protein) family of chaperones binds short se-
protein’s conformation might change when ligands are quences of hydrophobic amino acids in newly syn-
bound or when temperature, pH, or ionic strength is thesized polypeptides, shielding them from solvent.
altered. Scientists also are examining the library of Chaperones prevent aggregation, thus providing an op-
available protein structures in an attempt to devise portunity for the formation of appropriate secondary
computer programs that can predict the three-dimen- structural elements and their subsequent coalescence
sional conformation of a protein directly from its pri- into a molten globule. The hsp60 family of chaperones,
mary sequence. sometimes called chaperonins, differ in sequence and
structure from hsp70 and its homologs. Hsp60 acts
PROTEIN FOLDING later in the folding process, often together with an
hsp70 chaperone. The central cavity of the donut-
The Native Conformation of a Protein
Is Thermodynamically Favored
The number of distinct combinations of phi and psi
angles specifying potential conformations of even a rel-
atively small—15-kDa—polypeptide is unbelievably
vast. Proteins are guided through this vast labyrinth of
possibilities by thermodynamics. Since the biologically
relevant—or native—conformation of a protein gener-
ally is that which is most energetically favored, knowl-
edge of the native conformation is specified in the pri-
mary sequence. However, if one were to wait for a
polypeptide to find its native conformation by random
exploration of all possible conformations, the process
would require billions of years to complete. Clearly,
protein folding in cells takes place in a more orderly
and guided fashion.
shaped hsp60 chaperone provides a sheltered environ-
ment in which a polypeptide can fold until all hy-
drophobic regions are buried in its interior, eliminating
aggregation. Chaperone proteins can also ―rescue‖ pro-
teins that have become thermodynamically trapped in a
misfolded dead end by unfolding hydrophobic regions
and providing a second chance to fold productively.

Protein Disulfide Isomerase


Disulfide bonds between and within polypeptides stabi-
lize tertiary and quaternary structure. However, disul-
fide bond formation is nonspecific. Under oxidizing
conditions, a given cysteine can form a disulfide bond
with the SH of any accessible cysteinyl residue. By
catalyzing disulfide exchange, the rupture of an SS
bond and its reformation with a different partner cys-
teine, protein disulfide isomerase facilitates the forma-
tion of disulfide bonds that stabilize their native confor-
mation.
Enzymes Mechanism 4
BIOMEDICAL IMPORTANCE with the ability to simultaneously conduct and inde-
pendently control a broad spectrum of chemical
Enzymes are biologic polymers that catalyze the chemi- processes.
cal reactions which make life as we know it possible.
The presence and maintenance of a complete and bal-
anced set of enzymes is essential for the breakdown of ENZYMES ARE CLASSIFIED BY REACTION
nutrients to supply energy and chemical building TYPE & MECHANISM
blocks; the assembly of those building blocks into pro-
A system of enzyme nomenclature that is comprehen-
teins, DNA, membranes, cells, and tissues; and the har-
nessing of energy to power cell motility and muscle sive, consistent, and at the same time easy to use has
contraction. With the exception of a few catalytic RNA proved elusive. The common names for most enzymes
molecules, or ribozymes, the vast majority of enzymes derive from their most distinctive characteristic: their
are proteins. Deficiencies in the quantity or catalytic ac- ability to catalyze a specific chemical reaction. In gen-
tivity of key enzymes can result from genetic defects, eral, an enzyme’s name consists of a term that identifies
nutritional deficits, or toxins. Defective enzymes can re- the type of reaction catalyzed followed by the suffix
sult from genetic mutations or infection by viral or bac- -ase. For example, dehydrogenases remove hydrogen
terial pathogens (eg, Vibrio cholerae). Medical scientists atoms, proteases hydrolyze proteins, and isomerases cat-
address imbalances in enzyme activity by using pharma- alyze rearrangements in configuration. One or more
cologic agents to inhibit specific enzymes and are inves- modifiers usually precede this name. Unfortunately,
tigating gene therapy as a means to remedy deficits in while many modifiers name the specific substrate in-
enzyme level or function. volved (xanthine oxidase), others identify the source of
the enzyme (pancreatic ribonuclease), specify its mode
of regulation (hormone-sensitive lipase), or name a dis-
ENZYMES ARE EFFECTIVE & HIGHLY tinguishing characteristic of its mechanism (a cysteine
SPECIFIC CATALYSTS protease). When it was discovered that multiple forms
of some enzymes existed, alphanumeric designators
The enzymes that catalyze the conversion of one or were added to distinguish between them (eg, RNA
more compounds (substrates) into one or more differ- polymerase III; protein kinase C). To address the am-
ent compounds (products) enhance the rates of the biguity and confusion arising from these inconsistencies
corresponding noncatalyzed reaction by factors of at in nomenclature and the continuing discovery of new
least 106. Like all catalysts, enzymes are neither con- enzymes, the International Union of Biochemists (IUB)
sumed nor permanently altered as a consequence of developed a complex but unambiguous system of en-
their participation in a reaction. zyme nomenclature. In the IUB system, each enzyme
In addition to being highly efficient, enzymes are has a unique name and code number that reflect the
also extremely selective catalysts. Unlike most catalysts type of reaction catalyzed and the substrates involved.
used in synthetic chemistry, enzymes are specific both Enzymes are grouped into six classes, each with several
for the type of reaction catalyzed and for a single sub- subclasses. For example, the enzyme commonly called
strate or a small set of closely related substrates. En- ―hexokinase‖ is designated ―ATP:D-hexose-6-phospho-
zymes are also stereospecific catalysts and typically cat- transferase E.C. 2.7.1.1.‖ This identifies hexokinase as a
alyze reactions only of specific stereoisomers of a given member of class 2 (transferases), subclass 7 (transfer of a
compound—for example, D- but not L-sugars, L- but phosphoryl group), sub-subclass 1 (alcohol is the phos-
not D-amino acids. Since they bind substrates through phoryl acceptor). Finally, the term ―hexose-6‖ indicates
at least ―three points of attachment,‖ enzymes can even that the alcohol phosphorylated is that of carbon six of
convert nonchiral substrates to chiral products. Figure a hexose. Listed below are the six IUB classes of en-
7–1 illustrates why the enzyme-catalyzed reduction of zymes and the reactions they catalyze.
the nonchiral substrate pyruvate produces L-lactate
rather a racemic mixture of D- and L-lactate. The ex- 1. Oxidoreductases catalyze oxidations and reduc-
quisite specificity of enzyme catalysts imbues living cells tions.
49
50 /

Prosthetic Groups Are Tightly Integrated


Into an Enzyme’s Structure
Prosthetic groups are distinguished by their tight, stable
incorporation into a protein’s structure by covalent or
noncovalent forces. Examples include pyridoxal phos-
phate, flavin mononucleotide (FMN), flavin dinu-
cleotide (FAD), thiamin pyrophosphate, biotin, and
the metal ions of Co, Cu, Mg, Mn, Se, and Zn. Metals
are the most common prosthetic groups. The roughly
Enzyme site Substrate
one-third of all enzymes that contain tightly bound
Figure 7–1. Planar representation of the ―three- metal ions are termed metalloenzymes. Metal ions that
point attachment‖ of a substrate to the active site of an participate in redox reactions generally are complexed
enzyme. Although atoms 1 and 4 are identical, once
to prosthetic groups such as heme (Chapter 6) or iron-
sulfur clusters (Chapter 12). Metals also may facilitate
atoms 2 and 3 are bound to their complementary sites
the binding and orientation of substrates, the formation
on the enzyme, only atom 1 can bind. Once bound to
of covalent bonds with reaction intermediates (Co 2+ in
an enzyme, apparently identical atoms thus may be dis-
coenzyme B12), or interaction with substrates to render
tinguishable, permitting a stereospecific chemical
them more electrophilic (electron-poor) or nucleo-
change. philic (electron-rich).

Cofactors Associate Reversibly With


2. Transferases catalyze transfer of groups such as Enzymes or Substrates
methyl or glycosyl groups from a donor molecule
to an acceptor molecule. Cofactors serve functions similar to those of prosthetic
3. Hydrolases catalyze the hydrolytic cleavage of groups but bind in a transient, dissociable manner ei-
C C, C O, CN, P O, and certain other ther to the enzyme or to a substrate such as ATP. Un-
bonds, including acid anhydride bonds. like the stably associated prosthetic groups, cofactors
therefore must be present in the medium surrounding
4. Lyases catalyze cleavage of C C, C O, CN, the enzyme for catalysis to occur. The most common
and other bonds by elimination, leaving double cofactors also are metal ions. Enzymes that require a
bonds, and also add groups to double bonds. metal ion cofactor are termed metal-activated enzymes
5. Isomerases catalyze geometric or structural to distinguish them from the metalloenzymes for
changes within a single molecule. which metal ions serve as prosthetic groups.
6. Ligases catalyze the joining together of two mole-
cules, coupled to the hydrolysis of a pyrophospho-
ryl group in ATP or a similar nucleoside triphos- Coenzymes Serve as Substrate Shuttles
phate. Coenzymes serve as recyclable shuttles—or group
transfer reagents—that transport many substrates from
Despite the many advantages of the IUB system, their point of generation to their point of utilization.
texts tend to refer to most enzymes by their older and Association with the coenzyme also stabilizes substrates
shorter, albeit sometimes ambiguous names. such as hydrogen atoms or hydride ions that are unsta-
ble in the aqueous environment of the cell. Other
chemical moieties transported by coenzymes include
PROSTHETIC GROUPS, COFACTORS, methyl groups (folates), acyl groups (coenzyme A), and
& COENZYMES PLAY IMPORTANT oligosaccharides (dolichol).
ROLES IN CATALYSIS
Many enzymes contain small nonprotein molecules and Many Coenzymes, Cofactors, & Prosthetic
metal ions that participate directly in substrate binding Groups Are Derivatives of B Vitamins
or catalysis. Termed prosthetic groups, cofactors, and The water-soluble B vitamins supply important compo-
coenzymes, these extend the repertoire of catalytic ca- nents of numerous coenzymes. Many coenzymes con-
pabilities beyond those afforded by the limited number tain, in addition, the adenine, ribose, and phosphoryl
of functional groups present on the aminoacyl side moieties of AMP or ADP (Figure 7–2). Nicotinamide
chains of peptides. and riboflavin are components of the redox coenzymes
/ 51

O Arg 145

NH2 NH

+ OH C
N NH2 NH2

O CH2 O
H H
O C O O
N H C C
N H Tyr 248
H H H
HO OH N C
His 196
– O CH2
O P O
2+
Zn
NH2
NH2 O O His 69
C N
N
Glu 72
N
O N N
H
O P O CH2
Figure 7–3. Two-dimensional representation of a
O– O dipeptide substrate, glycyl-tyrosine, bound within the
active site of carboxypeptidase A.
H H
HO OR

Figure 7–2. Structure of NAD+ and NADP+. For


NAD+, R  H. For NADP+, R  PO .
2

NAD and NADP and FMN and FAD, respectively.


Pantothenic acid is a component of the acyl group car-
rier coenzyme A. As its pyrophosphate, thiamin partici-
pates in decarboxylation of -keto acids and folic acid
and cobamide coenzymes function in one-carbon me-
tabolism.
Enzyme Kinetics 5
BIOMEDICAL IMPORTANCE A  B  P+ Q (2)
Enzyme kinetics is the field of biochemistry concerned Unidirectional arrows are also used to describe reac-
with the quantitative measurement of the rates of en- tions in living cells where the products of reaction (2)
zyme-catalyzed reactions and the systematic study of fac- are immediately consumed by a subsequent enzyme-
tors that affect these rates. Kinetic analyses permit scien- catalyzed reaction. The rapid removal of product P or
tists to reconstruct the number and order of the Q therefore precludes occurrence of the reverse reac-
individual steps by which enzymes transform substrates tion, rendering equation (2) functionally irreversible
into products. The study of enzyme kinetics also repre- under physiologic conditions.
sents the principal way to identify potential therapeutic
agents that selectively enhance or inhibit the rates of spe-
cific enzyme-catalyzed processes. Together with site- CHANGES IN FREE ENERGY DETERMINE
directed mutagenesis and other techniques that probe THE DIRECTION & EQUILIBRIUM STATE
protein structure, kinetic analysis can also reveal details OF CHEMICAL REACTIONS
of the catalytic mechanism. A complete, balanced set of
enzyme activities is of fundamental importance for main- The Gibbs free energy change G (also called either the
taining homeostasis. An understanding of enzyme kinet- free energy or Gibbs energy) describes both the direc-
ics thus is important for understanding how physiologic tion in which a chemical reaction will tend to proceed
stresses such as anoxia, metabolic acidosis or alkalosis, and the concentrations of reactants and products that
toxins, and pharmacologic agents affect that balance. will be present at equilibrium. G for a chemical reac-
tion equals the sum of the free energies of formation of
CHEMICAL REACTIONS ARE DESCRIBED the reaction products GP minus the sum of the free
energies of formation of the substrates GS. G0 de-
USING BALANCED EQUATIONS notes the change in free energy that accompanies transi-
A balanced chemical equation lists the initial chemical tion from the standard state, one-molar concentrations
species (substrates) present and the new chemical of substrates and products, to equilibrium. A more use-
species (products) formed for a particular chemical re- ful biochemical term is G0, which defines G0 at a
action, all in their correct proportions or stoichiome- standard state of 107 M protons, pH 7.0 (Chapter 10).
try. For example, balanced equation (1) below describes If the free energy of the products is lower than that of
the reaction of one molecule each of substrates A and B the substrates, the signs of G0 and G0 will be nega-
to form one molecule each of products P and Q. tive, indicating that the reaction as written is favored in
the direction left to right. Such reactions are referred to
 P Q
A B  (1) as spontaneous. The sign and the magnitude of the
free energy change determine how far the reaction will
The double arrows indicate reversibility, an intrinsic proceed. Equation (3)—
property of all chemical reactions. Thus, for reaction
(1), if A and B can form P and Q, then P and Q can G0  RT ln Keq (3)
also form A and B. Designation of a particular reactant
as a ―substrate‖ or ―product‖ is therefore somewhat ar- —illustrates the relationship between the equilibrium
bitrary since the products for a reaction written in one constant Keq and G0, where R is the gas constant (1.98
direction are the substrates for the reverse reaction. The cal/mol/°K or 8.31 J/mol/°K) and T is the absolute
term ―products‖ is, however, often used to designate temperature in degrees Kelvin. Keq is equal to the prod-
the reactants whose formation is thermodynamically fa- uct of the concentrations of the reaction products, each
vored. Reactions for which thermodynamic factors raised to the power of their stoichiometry, divided by
strongly favor formation of the products to which the the product of the substrates, each raised to the power
arrow points often are represented with a single arrow of their stoichiometry.
as if they were ―irreversible‖:
60
/ 61

Raising the temperature increases For the reaction A + B  P + Q— characteristic changes in free energy, GF,
and GD are
[P][Q] associated with each partial reaction.
Keq  (4)
[A][B] E R L 
 ELRLL GF (8)
and for reaction (5)
ELRLL 
 E  R L GD (9)
(5)
AA 
 P  E R L G  G  G
E R L  F D (8-10)
[P] (6)
K eq 
[A]2 For the overall reaction (10), G is the sum of GF and
GD. As for any equation of two terms, it is not possi-
—G0 may be calculated from equation (3) if the con- ble to infer from G either the sign or the magnitude
centrations of substrates and products present at equi- of GF or GD.
librium are known. If G0 is a negative number, Keq Many reactions involve multiple transition states,
will be greater than unity and the concentration of each with an associated change in free energy. For these
products at equilibrium will exceed that of substrates. If reactions, the overall G represents the sum of all of
G0 is positive, Keq will be less than unity and the for- the free energy changes associated with the formation
mation of substrates will be favored. and decay of all of the transition states. Therefore, it is
Notice that, since G0 is a function exclusively of not possible to infer from the overall ΔG the num-
the initial and final states of the reacting species, it can ber or type of transition states through which the re-
provide information only about the direction and equi- action proceeds. Stated another way: overall thermo-
librium state of the reaction. G0 is independent of the dynamics tells us nothing about kinetics.
mechanism of the reaction and therefore provides no
information concerning rates of reactions. Conse- GF Defines the Activation Energy
quently—and as explained below—although a reaction
may have a large negative G0 or G0, it may never- Regardless of the sign or magnitude of G, GF for the
theless take place at a negligible rate. overwhelming majority of chemical reactions has a pos-
itive sign. The formation of transition state intermedi-
ates therefore requires surmounting of energy barriers.
THE RATES OF REACTIONS For this reason, GF is often termed the activation en-
ARE DETERMINED BY THEIR ergy, Eact, the energy required to surmount a given en-
ACTIVATION ENERGY ergy barrier. The ease—and hence the frequency—with
which this barrier is overcome is inversely related to
Reactions Proceed via Transition States Eact. The thermodynamic parameters that determine
how fast a reaction proceeds thus are the GF values for
The concept of the transition state is fundamental to formation of the transition states through which the re-
understanding the chemical and thermodynamic basis action proceeds. For a simple reaction, where means
of catalysis. Equation (7) depicts a displacement reac- ―proportionate to,‖
tion in which an entering group E displaces a leaving
group L, attached initially to R. E
act
(11)
E R L  Rate  e RT
 E  R L (7)
Midway through the displacement, the bond between The activation energy for the reaction proceeding in the
R and L has weakened but has not yet been completely opposite direction to that drawn is equal to GD.
severed, and the new bond between E and R is as yet
incompletely formed. This transient intermediate—in
which neither free substrate nor product exists—is NUMEROUS FACTORS AFFECT
termed the transition state, E···R···L. Dotted lines THE REACTION RATE
represent the ―partial‖ bonds that are undergoing for- The kinetic theory—also called the collision theory—
mation and rupture. of chemical kinetics states that for two molecules to
Reaction (7) can be thought of as consisting of two react they must (1) approach within bond-forming dis-
―partial reactions,‖ the first corresponding to the forma- tance of one another, or ―collide‖; and (2) must possess
tion (F) and the second to the subsequent decay (D) of sufficient kinetic energy to overcome the energy barrier
the transition state intermediate. As for all reactions, for reaching the transition state. It therefore follows
that anything which increases the frequency or energy of which can also be written as
collision between substrates will increase the rate of the
reaction in which they participate. ABBP (15)
the corresponding rate expression is
Temperature
the kinetic energy of molecules. As illustrated in Rate  [A][B][B] (16)
Figure 8–1, the total num- ber of molecules whose or
kinetic energy exceeds the en- ergy barrier Eact (vertical
bar) for formation of products increases from low (A), Rate  [A][B]
2
(17)
through intermediate (B), to
high (C) temperatures. Increasing the kinetic energy of For the general case when n molecules of A react with
molecules also increases their motion and therefore the m molecules of B,
frequency with which they collide. This combination of
more frequent and more highly energetic and produc- nA  mB  P (18)
tive collisions increases the reaction rate.
the rate expression is
Reactant Concentration
Rate  [A]n[B]m (19)
The frequency with which molecules collide is directly
proportionate to their concentrations. For two different Replacing the proportionality constant with an equal
molecules A and B, the frequency with which they col- sign by introducing a proportionality or rate constant
lide will double if the concentration of either A or B is k characteristic of the reaction under study gives equa-
doubled. If the concentrations of both A and B are dou- tions (20) and (21), in which the subscripts 1 and 1
bled, the probability of collision will increase fourfold. refer to the rate constants for the forward and reverse
For a chemical reaction proceeding at constant tem- reactions, respectively.
perature that involves one molecule each of A and B,
Rate 1  k1[A]n[B]m (20)
ABP (12)
the number of molecules that possess kinetic energy Rate1  k 1[P] (21)
sufficient to overcome the activation energy barrier will
be a constant. The number of collisions with sufficient
energy to produce product P therefore will be directly Keq Is a Ratio of Rate Constants
proportionate to the number of collisions between A While all chemical reactions are to some extent re-
and B and thus to their molar concentrations, denoted versible, at equilibrium the overall concentrations of re-
by square brackets. actants and products remain constant. At equilibrium,
the rate of conversion of substrates to products there-
Rate  [A][B] (13) fore equals the rate at which products are converted to
Similarly, for the reaction represented by substrates.

A  2B  P (14) Rate1  Rate1 (22)

Therefore,
k1[A]n[B]m  k 1[P] (23)
Energy barrier
 and
A B
Number of
molecules

k1  [P]
 (24)
k 1 [A]n[B]m

The ratio of k1 to k1 is termed the equilibrium con-


0 stant, Keq. The following important properties of a sys-

Kinetic energy tem at equilibrium must be kept in mind:
Figure 8–1. The energy barrier for chemical (1) The equilibrium constant is a ratio of the reaction
reactions. rate constants (not the reaction rates).
(2) At equilibrium, the reaction rates (not the rate
constants) of the forward and back reactions are Go  RT ln Keq (25)
equal. If we include the presence of the enzyme (E) in the cal-
(3) Equilibrium is a dynamic state. Although there is culation of the equilibrium constant for a reaction,
no net change in the concentration of substrates
or products, individual substrate and product A B  Enz 
 P+ Q +Enz (26)
molecules are continually being interconverted.
the expression for the equilibrium constant,
(4) The numeric value of the equilibrium constant
Keq can be calculated either from the concentra- [P][Q][Enz]
tions of substrates and products at equilibrium or K eq  (27)
[A][B][Enz]
from the ratio k1/k1.
reduces to one identical to that for the reaction in the
absence of the enzyme:
THE KINETICS OF
ENZYMATIC CATALYSIS [P][Q]
K eq  (28)
[A][B]
Enzymes Lower the Activation Energy
Barrier for a Reaction Enzymes therefore have no effect on Keq.
All enzymes accelerate reaction rates by providing tran-
sition states with a lowered GF for formation of the
transition states. However, they may differ in the way MULTIPLE FACTORS AFFECT THE RATES
this is achieved. Where the mechanism or the sequence OF ENZYME-CATALYZED REACTIONS
of chemical steps at the active site is essentially the same
as those for the same reaction proceeding in the absence Temperature
of a catalyst, the environment of the active site lowers Raising the temperature increases the rate of both uncat-
ΔGF by stabilizing the transition state intermediates. As alyzed and enzyme-catalyzed reactions by increasing the
discussed in Chapter 7, stabilization can involve (1) kinetic energy and the collision frequency of the react-
acid-base groups suitably positioned to transfer protons ing molecules. However, heat energy can also increase
to or from the developing transition state intermediate, the kinetic energy of the enzyme to a point that exceeds
(2) suitably positioned charged groups or metal ions the energy barrier for disrupting the noncovalent inter-
that stabilize developing charges, or (3) the imposition actions that maintain the enzyme’s three-dimensional
of steric strain on substrates so that their geometry ap- structure. The polypeptide chain then begins to unfold,
proaches that of the transition state. HIV protease (Fig- or denature, with an accompanying rapid loss of cat-
ure 7–6) illustrates catalysis by an enzyme that lowers alytic activity. The temperature range over which an
the activation barrier by stabilizing a transition state in- enzyme maintains a stable, catalytically competent con-
termediate. formation depends upon—and typically moderately
Catalysis by enzymes that proceeds via a unique re- exceeds—the normal temperature of the cells in which
action mechanism typically occurs when the transition it resides. Enzymes from humans generally exhibit sta-
state intermediate forms a covalent bond with the en- bility at temperatures up to 45–55 °C. By contrast,
zyme (covalent catalysis). The catalytic mechanism of enzymes from the thermophilic microorganisms that re-
the serine protease chymotrypsin (Figure 7–7) illus- side in volcanic hot springs or undersea hydrothermal
trates how an enzyme utilizes covalent catalysis to pro- vents may be stable up to or above 100 °C.
vide a unique reaction pathway. The Q10, or temperature coefficient, is the factor
by which the rate of a biologic process increases for a
ENZYMES DO NOT AFFECT Keq 10 °C increase in temperature. For the temperatures
over which enzymes are stable, the rates of most bio-
Enzymes accelerate reaction rates by lowering the acti- logic processes typically double for a 10 °C rise in tem-
vation barrier GF. While they may undergo transient perature (Q10 = 2). Changes in the rates of enzyme-
modification during the process of catalysis, enzymes catalyzed reactions that accompany a rise or fall in body
emerge unchanged at the completion of the reaction. temperature constitute a prominent survival feature for
The presence of an enzyme therefore has no effect on ―cold-blooded‖ life forms such as lizards or fish, whose
G0 for the overall reaction, which is a function solely body temperatures are dictated by the external environ-
of the initial and final states of the reactants. Equation ment. However, for mammals and other homeothermic
(25) shows the relationship between the equilibrium organisms, changes in enzyme reaction rates with tem-
constant for a reaction and the standard free energy perature assume physiologic importance only in cir-
change for that reaction: cumstances such as fever or hypothermia.
Hydrogen Ion Concentration the rate of the forward reaction. Assays of enzyme activ-
ity almost always employ a large (10 3–107) molar excess
The rate of almost all enzyme-catalyzed reactions ex- of substrate over enzyme. Under these conditions, v i is
hibits a significant dependence on hydrogen ion con- proportionate to the concentration of enzyme. Measur-
centration. Most intracellular enzymes exhibit optimal ing the initial velocity therefore permits one to estimate
activity at pH values between 5 and 9. The relationship the quantity of enzyme present in a biologic sample.
of activity to hydrogen ion concentration (Figure 8–2)
reflects the balance between enzyme denaturation at
high or low pH and effects on the charged state of the SUBSTRATE CONCENTRATION AFFECTS
enzyme, the substrates, or both. For enzymes whose REACTION RATE
mechanism involves acid-base catalysis, the residues in- In what follows, enzyme reactions are treated as if they
volved must be in the appropriate state of protonation had only a single substrate and a single product. While
for the reaction to proceed. The binding and recogni- most enzymes have more than one substrate, the princi-
tion of substrate molecules with dissociable groups also ples discussed below apply with equal validity to en-
typically involves the formation of salt bridges with the zymes with multiple substrates.
enzyme. The most common charged groups are the For a typical enzyme, as substrate concentration is
negative carboxylate groups and the positively charged increased, vi increases until it reaches a maximum value
groups of protonated amines. Gain or loss of critical Vmax (Figure 8–3). When further increases in substrate
charged groups thus will adversely affect substrate bind- concentration do not further increase vi, the enzyme is
ing and thus will retard or abolish catalysis. said to be ―saturated‖ with substrate. Note that the
shape of the curve that relates activity to substrate con-
ASSAYS OF ENZYME-CATALYZED centration (Figure 8–3) is hyperbolic. At any given in-
REACTIONS TYPICALLY MEASURE stant, only substrate molecules that are combined with
THE INITIAL VELOCITY the enzyme as an ES complex can be transformed into
product. Second, the equilibrium constant for the for-
Most measurements of the rates of enzyme-catalyzed re- mation of the enzyme-substrate complex is not infi-
actions employ relatively short time periods, conditions nitely large. Therefore, even when the substrate is pre-
that approximate initial rate conditions. Under these sent in excess (points A and B of Figure 8–4), only a
conditions, only traces of product accumulate, hence fraction of the enzyme may be present as an ES com-
the rate of the reverse reaction is negligible. The initial plex. At points A or B, increasing or decreasing [S]
velocity (vi) of the reaction thus is essentially that of therefore will increase or decrease the number of ES
complexes with a corresponding change in v i. At point
C (Figure 8–4), essentially all the enzyme is present as
X the ES complex. Since no free enzyme remains available
100 for forming ES, further increases in [S] cannot increase
the rate of the reaction. Under these saturating condi-
tions, vi depends solely on—and thus is limited by—
SH+ E– the rapidity with which free enzyme is released to com-
bine with more substrate.
%

Vmax
0
Low High
Vmax/2
pH
vi
B
Figure 8–2. Effect of pH on enzyme activity. Con-

sider, for example, a negatively charged enzyme (EH ) A Vmax/2
that binds a positively charged substrate (SH+). Shown
is the proportion (%) of SH+ [\\\] and of EH [///] as a K [S]
function of pH. Only in the cross-hatched area do both
the enzyme and the substrate bear an appropriate Figure 8–3. Effect of substrate concentration on the
charge. initial velocity of an enzyme-catalyzed reaction.
Nucleotides 6
BIOMEDICAL IMPORTANCE H H
6 4
C 5 C 5
Nucleotides—the monomer units or building blocks of 1
N 3
N CH
nucleic acids—serve multiple additional functions. They 2
CH
C HC CH
form a part of many coenzymes and serve as donors of H N 4 2 N 6
3 H 1
phosphoryl groups (eg, ATP or GTP), of sugars (eg,
UDP- or GDP-sugars), or of lipid (eg, CDP-acylglyc- Purine Pyrimidine
erol). Regulatory nucleotides include the second mes-
sengers cAMP and cGMP, the control by ADP of ox- Figure 33–1. Purine and pyrimidine. The atoms are
idative phosphorylation, and allosteric regulation of numbered according to the international system.
enzyme activity by ATP, AMP, and CTP. Synthetic
purine and pyrimidine analogs that contain halogens,
thiols, or additional nitrogen are employed for chemo-
therapy of cancer and AIDS and as suppressors of the Nucleosides & Nucleotides
immune response during organ transplantation. Nucleosides are derivatives of purines and pyrimidines
that have a sugar linked to a ring nitrogen. Numerals
PURINES, PYRIMIDINES, NUCLEOSIDES, with a prime (eg, 2 or 3) distinguish atoms of the
& NUCLEOTIDES sugar from those of the heterocyclic base. The sugar in
ribonucleosides is D-ribose, and in deoxyribonucleo-
Purines and pyrimidines are nitrogen-containing hete- sides it is 2-deoxy-D-ribose. The sugar is linked to the
rocycles, cyclic compounds whose rings contain both heterocyclic base via a β-N-glycosidic bond, almost al-
carbon and other elements (hetero atoms). Note that ways to N-1 of a pyrimidine or to N-9 of a purine (Fig-
the smaller pyrimidine has the longer name and the ure 33–3).
larger purine the shorter name and that their six-atom
rings are numbered in opposite directions (Figure
33–1). The planar character of purines and pyrimidines
facilitates their close association, or ―stacking,‖ which NH2 NH O OH
stabilizes double-stranded DNA (Chapter 36). The oxo
and amino groups of purines and pyrimidines exhibit
keto-enol and amine-imine tautomerism (Figure 33–2),
but physiologic conditions strongly favor the amino Figure 33–2. Tautomerism of the oxo and amino
and oxo forms. functional groups of purines and pyrimidines.

286
NUCLEOTIDES / 287

NH2 NH2 O O

HN
HN

O O
N N H2N N N

HO HO HO HO
O O O O

OH OH OH OH OH OH OH OH
Adenosine Cytidine Guanosine Uridine

Figure 33–3. Ribonucleosides, drawn as the syn conformers.

Mononucleotides are nucleosides with a phosphoryl cleotides. Both therefore exist as syn or anti conformers
group esterified to a hydroxyl group of the sugar. The (Figure 33–5). While both conformers occur in nature,
3- and 5-nucleotides are nucleosides with a phospho- anti conformers predominate. Table 33–1 lists the
ryl group on the 3- or 5-hydroxyl group of the sugar, major purines and pyrimidines and their nucleoside
respectively. Since most nucleotides are 5-, the prefix and nucleotide derivatives. Single-letter abbreviations
―5-‖ is usually omitted when naming them. UMP and are used to identify adenine (A), guanine (G), cytosine
dAMP thus represent nucleotides with a phosphoryl (C), thymine (T), and uracil (U), whether free or pre-
group on C-5 of the pentose. Additional phosphoryl sent in nucleosides or nucleotides. The prefix ―d‖
groups linked by acid anhydride bonds to the phos- (deoxy) indicates that the sugar is 2-deoxy-D-ribose
phoryl group of a mononucleotide form nucleoside (eg, dGTP) (Figure 33–6).
diphosphates and triphosphates (Figure 33–4).
Steric hindrance by the base restricts rotation about Nucleic Acids Also Contain
the -N-glycosidic bond of nucleosides and nu- Additional Bases
Small quantities of additional purines and pyrimidines
NH2 occur in DNA and RNAs. Examples include 5-methyl-
N
cytosine of bacterial and human DNA, 5-hydroxy-
Adenine methylcytosine of bacterial and viral nucleic acids, and
mono- and di-N-methylated adenine and guanine of
N N

CH2
O NH2 NH2
O Ribose
O– O O–

HO P O P O P
HO OH
O O– O N N

HO HO
Adenosine 5-monophosphate (AMP) O O

Adenosine 5-diphosphate (ADP)


Syn Anti
OH OH OH OH
Adenosine 5-triphosphate (ATP)
Figure 33–5. The syn and anti conformers of adeno-
Figure 33–4. ATP, its diphosphate, and its sine differ with respect to orientation about the N-gly-
monophosphate. cosidic bond.
Table 33–1. Bases, nucleosides, and nucleotides.

Nucleoside
Base Base X = Ribose or Nucleotide, Where
Formula X=H Deoxyribose X = Ribose Phosphate
NH2

N N
Adenine Adenosine Adenosine monophosphate
N
A A AMP
N
X

O
H N
N
Guanine Guanosine Guanosine monophosphate
H2N N
G G GMP
N
X

NH2

N Cytosine Cytidine Cytidine monophosphate


C C CMP
O N

O
H
N
Uracil Uridine Uridine monophosphate
U U UMP
O N

O
H CH3
N
Thymine Thymidine Thymidine monophosphate
O N
T T TMP

dX

NH2 NH2 O O
CH3
HN HN

N N N N O N O

O O O O O O O O O O O O
P P P P
–O O– –O O– –O O– –
O O

OH OH OH H OH OH OH H

AMP dAMP UMP TMP

Figure 33–6. AMP, dAMP, UMP, and TMP.


NUCLEOTIDES / 289
NH2 NH2
NH2 O
CH3 CH2OH
N
HN

O O N N H2N N N
N N
H H O
CH2 O CH2
5-Methylcytosine 5-Hydroxymethylcytosine O O
–O –O
P O P O

H3C CH3 O OH O
N O CH3 OH

N N
7
Figure 33–9. cAMP, 3,5-cyclic AMP, and cGMP.
HN

N H2N N N
H
Nucleotides Serve Diverse
Dimethylaminoadenine 7-Methylguanine Physiologic Functions
Figure 33–7. Four uncommon naturally occurring Nucleotides participate in reactions that fulfill physio-
pyrimidines and purines. logic functions as diverse as protein synthesis, nucleic
acid synthesis, regulatory cascades, and signal transduc-
tion pathways.
mammalian messenger RNAs (Figure 33–7). These
atypical bases function in oligonucleotide recognition
and in regulating the half-lives of RNAs. Free nu- Nucleoside Triphosphates Have High
cleotides include hypoxanthine, xanthine, and uric acid Group Transfer Potential
(see Figure 34–8), intermediates in the catabolism of Acid anhydrides, unlike phosphate esters, have high
adenine and guanine. Methylated heterocyclic bases of group transfer potential. 0 for the hydrolysis of each
plants include the xanthine derivatives caffeine of cof- of the terminal phosphates of nucleoside triphosphates
fee, theophylline of tea, and theobromine of cocoa (Fig- is about 7 kcal/mol (30 kJ/mol). The high group
ure 33–8). transfer potential of purine and pyrimidine nucleoside
Posttranslational modification of preformed polynu- triphosphates permits them to function as group trans-
cleotides can generate additional bases such as fer reagents. Cleavage of an acid anhydride bond typi-
pseudouridine, in which D-ribose is linked to C-5 of cally is coupled with a highly endergonic process such
uracil by a carbon-to-carbon bond rather than by a as covalent bond synthesis—eg, polymerization of nu-
-N-glycosidic bond. The nucleotide pseudouridylic cleoside triphosphates to form a nucleic acid.
acid  arises by rearrangement of UMP of a preformed In addition to their roles as precursors of nucleic
tRNA. Similarly, methylation by S-adenosylmethionine acids, ATP, GTP, UTP, CTP, and their derivatives
of a UMP of preformed tRNA forms TMP (thymidine each serve unique physiologic functions discussed in
monophosphate), which contains ribose rather than de- other chapters. Selected examples include the role of
oxyribose. ATP as the principal biologic transducer of free energy;
the second messenger cAMP (Figure 33–9); adenosine
CH 3-phosphate-5-phosphosulfate (Figure 33–10), the
O 3
sulfate donor for sulfated proteoglycans (Chapter 48)
H3C N and for sulfate conjugates of drugs; and the methyl
N
group donor S-adenosylmethionine (Figure 33–11).
O N
N
CH3
P
Figure 33–8. Caffeine, a trimethylxanthine. The di- Adenine Ribose P O SO 2–
methylxanthines theobromine and theophylline are
similar but lack the methyl group at N-1 and at N-7, re- Figure 33–10. Adenosine 3-phosphate-5-phos-
spectively. phosulfate.
NH2 Table 33–2. Many coenzymes and related
compounds are derivatives of adenosine
monophosphate.
N N NH2
N
COO– CH3 CH2 N Adenine
O
CH CH2 CH S N
+ N
O
NH3+
R O P O CH2
HO OH
O– O

Methionine Adenosine
R'' O OR'
Figure 33–11. S-Adenosylmethionine.
D-Ribose

Coenzyme R R' R" n


GTP serves as an allosteric regulator and as an energy Active methionine Methionine* H H 0
source for protein synthesis, and cGMP (Figure 33–9) Amino acid adenylates Amino acid H H 1
serves as a second messenger in response to nitric oxide Active sulfate SO32 H PO32 1
(NO) during relaxation of smooth muscle (Chapter 3,5-Cyclic AMP H PO32 1

48). UDP-sugar derivatives participate in sugar epimer- NAD* H H 2
izations and in biosynthesis of glycogen, glucosyl disac- NADP* †
PO32 H 2
charides, and the oligosaccharides of glycoproteins and FAD †
H H 2
proteoglycans (Chapters 47 and 48). UDP-glucuronic CoASH †
H PO32 2
acid forms the urinary glucuronide conjugates of biliru- *Replaces phosphoryl group.
bin (Chapter 32) and of drugs such as aspirin. CTP †
R is a B vitamin derivative.
participates in biosynthesis of phosphoglycerides,
sphingomyelin, and other substituted sphingosines
(Chapter 24). Finally, many coenzymes incorporate nu-
cleotides as well as structures similar to purine and nucleic acids thus often is expressed in terms of ―ab-
pyrimidine nucleotides (see Table 33–2). sorbance at 260 nm.‖

Nucleotides Are Polyfunctional Acids SYNTHETIC NUCLEOTIDE ANALOGS


Nucleosides or free purine or pyrimidine bases are un-
ARE USED IN CHEMOTHERAPY
charged at physiologic pH. By contrast, the primary Synthetic analogs of purines, pyrimidines, nucleosides,
phosphoryl groups (pK about 1.0) and secondary phos- and nucleotides altered in either the heterocyclic ring or
phoryl groups (pK about 6.2) of nucleotides ensure that the sugar moiety have numerous applications in clinical
they bear a negative charge at physiologic pH. Nu- medicine. Their toxic effects reflect either inhibition of
cleotides can, however, act as proton donors or accep- enzymes essential for nucleic acid synthesis or their in-
tors at pH values two or more units above or below corporation into nucleic acids with resulting disruption
neutrality. of base-pairing. Oncologists employ 5-fluoro- or 5-
iodouracil, 3-deoxyuridine, 6-thioguanine and 6-mer-
captopurine, 5- or 6-azauridine, 5- or 6-azacytidine,
Nucleotides Absorb Ultraviolet Light and 8-azaguanine (Figure 33–12), which are incorpo-
The conjugated double bonds of purine and pyrimidine rated into DNA prior to cell division. The purine ana-
derivatives absorb ultraviolet light. The mutagenic ef- log allopurinol, used in treatment of hyperuricemia and
fect of ultraviolet light results from its absorption by gout, inhibits purine biosynthesis and xanthine oxidase
nucleotides in DNA with accompanying chemical activity. Cytarabine is used in chemotherapy of cancer.
changes. While spectra are pH-dependent, at pH 7.0 all Finally, azathioprine, which is catabolized to 6-mercap-
the common nucleotides absorb light at a wavelength topurine, is employed during organ transplantation to
close to 260 nm. The concentration of nucleotides and suppress immunologic rejection.
NUCLEOTIDES / 291

O O

I HN
HN
6N
O O
N

HO HO O
O
F O HN
O HN 5 8
N

H2N N
2 O H
N
H HO OH
HO H
5-Iodo-2-deoxyuridine 5-Fluorouracil 6-Azauridine 8-Azaguanine

SH SH OH

6 N N
N

N H2N N N N
H H H
6-Mercaptopurine 6-Thioguanine Alloburinol

Figure 33–12. Selected synthetic pyrimidine and purine analogs.


O

Nucleic Acid Structure & Function 7


BIOMEDICAL IMPORTANCE
The discovery that genetic information is coded along
the length of a polymeric molecule composed of only
four types of monomeric units was one of the major sci-
entific achievements of the twentieth century. This
polymeric molecule, DNA, is the chemical basis of
heredity and is organized into genes, the fundamental
units of genetic information. The basic information
pathway—ie, DNA directs the synthesis of RNA,
which in turn directs protein synthesis—has been eluci-
dated. Genes do not function autonomously; their
replication and function are controlled by various gene
products, often in collaboration with components of
various signal transduction pathways. Knowledge of the
structure and function of nucleic acids is essential in
understanding genetics and many aspects of pathophys-
iology as well as the genetic basis of disease.

DNA CONTAINS THE


GENETIC INFORMATION
The demonstration that DNA contained the genetic in-
formation was first made in 1944 in a series of experi-
ments by Avery, MacLeod, and McCarty. They showed
that the genetic determination of the character (type) of
the capsule of a specific pneumococcus could be trans-
mitted to another of a different capsular type by intro-
ducing purified DNA from the former coccus into the
latter. These authors referred to the agent (later shown
to be DNA) accomplishing the change as ―transforming
factor.‖ Subsequently, this type of genetic manipulation
has become commonplace. Similar experiments have
recently been performed utilizing yeast, cultured mam-
malian cells, and insect and mammalian embryos as re-
cipients and cloned DNA as the donor of genetic infor-
mation.

DNA Contains Four Deoxynucleotides


The chemical nature of the monomeric deoxynucleo-
tide units of DNA—deoxyadenylate, deoxyguanylate,
deoxycytidylate, and thymidylate—is described in
Chapter 33. These monomeric units of DNA are held
in polymeric form by 3,5-phosphodiester bridges con-
stituting a single strand, as depicted in Figure 35–1.
O
N
NH
5 G

CH2 N NH2
N NH2

O
O
P
O O
CH2 N
H H
H H
H3C NH
O
H O
T
P
O
H H CH2 N NH2
H H
O N
O
H O
P
CH2 N N
H H
H H
O
O
H O

P
H H
H H
O
3
P

Figure 35–1. A segment of one strand of a DNA molecule in which the purine and pyrimidine bases guanine
(G), cytosine (C), thymine (T), and adenine (A) are held together by a phosphodiester backbone between 2-de-
oxyribosyl moieties attached to the nucleobases by an N-glycosidic bond. Note that the backbone has a polarity
(ie, a direction). Convention dictates that a single-stranded DNA sequence is written in the 5 to 3 direction (ie,
pGpCpTpA, where G, C, T, and A represent the four bases and p represents the interconnecting phosphates).

sides in the sequence of nucleotides on one strand, the dine nucleotide, whereas the other pair, the A–T pair, is
template strand. This is the strand of DNA that is held together by two hydrogen bonds. Thus, the G–C
copied during nucleic acid synthesis. It is sometimes re- bonds are much more resistant to denaturation, or
ferred to as the noncoding strand. The opposite strand ―melting,‖ than A–T-rich regions.
is considered the coding strand because it matches the
RNA transcript that encodes the protein. The Denaturation (Melting) of DNA
The two strands, in which opposing bases are held Is Used to Analyze Its Structure
together by hydrogen bonds, wind around a central axis
in the form of a double helix. Double-stranded DNA The double-stranded structure of DNA can be sepa-
exists in at least six forms (A–E and Z). The B form is rated into two component strands (melted) in solution
usually found under physiologic conditions (low salt, by increasing the temperature or decreasing the salt
high degree of hydration). A single turn of B-DNA concentration. Not only do the two stacks of bases pull
about the axis of the molecule contains ten base pairs. apart but the bases themselves unstack while still con-
The distance spanned by one turn of B-DNA is 3.4 nected in the polymer by the phosphodiester backbone.
nm. The width (helical diameter) of the double helix in Concomitant with this denaturation of the DNA mole-
B-DNA is 2 nm. cule is an increase in the optical absorbance of the
As depicted in Figure 35–3, three hydrogen bonds purine and pyrimidine bases—a phenomenon referred
hold the deoxyguanosine nucleotide to the deoxycyti- to as hyperchromicity of denaturation. Because of the
CH3

O
H H
N
H
O
Thymidine
Minor groove

T S Adenosine
P S A o
P 34 A
S T A S
P P
S G S H
P P
S G C S
Major groove H
O

O
H
Cytosine

H Guanosine
20 A
Figure 35–3. Base pairing between deoxyadenosine
Figure 35–2. A diagrammatic representation of the and thymidine involves the formation of two hydrogen
Watson and Crick model of the double-helical structure bonds. Three such bonds form between deoxycytidine
of the B form of DNA. The horizontal arrow indicates and deoxyguanosine. The broken lines represent hy-
the width of the double helix (20 Å), and the vertical drogen bonds.
arrow indicates the distance spanned by one complete
turn of the double helix (34 Å). One turn of B-DNA in-
cludes ten base pairs (bp), so the rise is 3.4 Å per bp. or DNA-RNA hybrids to be separated at much lower
The central axis of the double helix is indicated by the temperatures and minimizes the phosphodiester bond
vertical rod. The short arrows designate the polarity of breakage that occurs at high temperatures.
the antiparallel strands. The major and minor grooves
are depicted. (A, adenine; C, cytosine; G, guanine; Renaturation of DNA Requires
T, thymine; P, phosphate; S, sugar [deoxyribose].) Base Pair Matching
Separated strands of DNA will renature or reassociate
when appropriate physiologic temperature and salt con-
stacking of the bases and the hydrogen bonding be- ditions are achieved. The rate of reassociation depends
tween the stacks, the double-stranded DNA molecule upon the concentration of the complementary strands.
exhibits properties of a rigid rod and in solution is a vis- Reassociation of the two complementary DNA strands
cous material that loses its viscosity upon denaturation. of a chromosome after DNA replication is a physiologic
The strands of a given molecule of DNA separate example of renaturation (see below). At a given temper-
over a temperature range. The midpoint is called the ature and salt concentration, a particular nucleic acid
melting temperature, or Tm. The Tm is influenced by strand will associate tightly only with a complementary
the base composition of the DNA and by the salt con- strand. Hybrid molecules will also form under appro-
centration of the solution. DNA rich in G–C pairs, priate conditions. For example, DNA will form a hy-
which have three hydrogen bonds, melts at a higher tem- brid with a complementary DNA (cDNA) or with a
perature than that rich in A–T pairs, which have two hy- cognate messenger RNA (mRNA; see below). When
drogen bonds. A tenfold increase of monovalent cation combined with gel electrophoresis techniques that sepa-
concentration increases the Tm by 16.6 °C. Formamide, rate hybrid molecules by size and radioactive labeling to
which is commonly used in recombinant DNA experi- provide a detectable signal, the resulting analytic tech-
ments, destabilizes hydrogen bonding between bases, niques are called Southern (DNA/cDNA) and North-
thereby lowering the Tm. This allows the strands of DNA ern blotting (DNA/RNA), respectively. These proce-
dures allow for very specific identification of hybrids the cell and organism, and it provides the information
from mixtures of DNA or RNA (see Chapter 40). inherited by daughter cells or offspring. Both of these
functions require that the DNA molecule serve as a
There Are Grooves in the DNA Molecule template—in the first case for the transcription of the
information into RNA and in the second case for the
Careful examination of the model depicted in Figure replication of the information into daughter DNA mol-
35–2 reveals a major groove and a minor groove wind- ecules.
ing along the molecule parallel to the phosphodiester The complementarity of the Watson and Crick dou-
backbones. In these grooves, proteins can interact specif- ble-stranded model of DNA strongly suggests that
ically with exposed atoms of the nucleotides (usually by replication of the DNA molecule occurs in a semicon-
H bonding) and thereby recognize and bind to specific servative manner. Thus, when each strand of the dou-
nucleotide sequences without disrupting the base pair- ble-stranded parental DNA molecule separates from its
ing of the double-helical DNA molecule. As discussed in complement during replication, each serves as a tem-
Chapters 37 and 39, regulatory proteins control the ex- plate on which a new complementary strand is synthe-
pression of specific genes via such interactions. sized (Figure 35–4). The two newly formed double-
stranded daughter DNA molecules, each containing
DNA Exists in Relaxed one strand (but complementary rather than identical)
& Supercoiled Forms from the parent double-stranded DNA molecule, are
then sorted between the two daughter cells (Figure
In some organisms such as bacteria, bacteriophages, and 35–5). Each daughter cell contains DNA molecules
many DNA-containing animal viruses, the ends of the with information identical to that which the parent
DNA molecules are joined to create a closed circle with possessed; yet in each daughter cell the DNA molecule
no covalently free ends. This of course does not destroy of the parent cell has been only semiconserved.
the polarity of the molecules, but it eliminates all free 3
and 5 hydroxyl and phosphoryl groups. Closed circles
exist in relaxed or supercoiled forms. Supercoils are intro- THE CHEMICAL NATURE OF RNA DIFFERS
duced when a closed circle is twisted around its own axis FROM THAT OF DNA
or when a linear piece of duplex DNA, whose ends are
fixed, is twisted. This energy-requiring process puts the Ribonucleic acid (RNA) is a polymer of purine and
molecule under stress, and the greater the number of su- pyrimidine ribonucleotides linked together by 3,5-
percoils, the greater the stress or torsion (test this by phosphodiester bridges analogous to those in DNA
twisting a rubber band). Negative supercoils are formed (Figure 35–6). Although sharing many features with
when the molecule is twisted in the direction opposite DNA, RNA possesses several specific differences:
from the clockwise turns of the right-handed double
helix found in B-DNA. Such DNA is said to be under- (1) In RNA, the sugar moiety to which the phos-
wound. The energy required to achieve this state is, in a phates and purine and pyrimidine bases are attached is
sense, stored in the supercoils. The transition to another ribose rather than the 2-deoxyribose of DNA.
form that requires energy is thereby facilitated by the un- (2) The pyrimidine components of RNA differ from
derwinding. One such transition is strand separation, those of DNA. Although RNA contains the ribonu-
which is a prerequisite for DNA replication and tran- cleotides of adenine, guanine, and cytosine, it does not
scription. Supercoiled DNA is therefore a preferred form possess thymine except in the rare case mentioned
in biologic systems. Enzymes that catalyze topologic below. Instead of thymine, RNA contains the ribonu-
changes of DNA are called topoisomerases. Topoisom- cleotide of uracil.
erases can relax or insert supercoils. The best-character- (3) RNA exists as a single strand, whereas DNA ex-
ized example is bacterial gyrase, which induces negative ists as a double-stranded helical molecule. However,
supercoiling in DNA using ATP as energy source. Ho- given the proper complementary base sequence with
mologs of this enzyme exist in all organisms and are im- opposite polarity, the single strand of RNA—as
portant targets for cancer chemotherapy. demonstrated in Figure 35–7—is capable of folding
back on itself like a hairpin and thus acquiring double-
DNA PROVIDES A TEMPLATE FOR stranded characteristics.
(4) Since the RNA molecule is a single strand com-
REPLICATION & TRANSCRIPTION plementary to only one of the two strands of a gene, its
The genetic information stored in the nucleotide se- guanine content does not necessarily equal its cytosine
quence of DNA serves two purposes. It is the source of content, nor does its adenine content necessarily equal
information for the synthesis of all protein molecules of its uracil content.
OLD OLD
5 G C 3

G
Original
C G parent molecule

T A

A T

G C
G C

A
First-generation
A T
daughter molecules
T A

G C

G C
3 5
T A T A
C G C G

C C
Second-generation
daughter molecules
A T A T

A T A T

T A T A

G C G C
Figure 35–5. DNA replication is semiconservative.
G G
During a round of replication, each of the two strands
A T A T of DNA is used as a template for synthesis of a new,
complementary strand.
3 T A 5 3 T A 5
OLT NEW ETW OLD
A A
Figure 35–4. The double-stranded structure of DNA
and the template function of each old strand (dark
complementarity, an RNA molecule can bind specifi-
shading) on which a new (light shading) complemen-
cally via the base-pairing rules to its template DNA
tary strand is synthesized.
strand; it will not bind (―hybridize‖) with the other
(coding) strand of its gene. The sequence of the RNA
molecule (except for U replacing T) is the same as that
of the coding strand of the gene (Figure 35–8).
(5) RNA can be hydrolyzed by alkali to 2,3 cyclic
diesters of the mononucleotides, compounds that can- Nearly All of the Several Species of RNA
not be formed from alkali-treated DNA because of the Are Involved in Some Aspect of Protein
absence of a 2-hydroxyl group. The alkali lability of Synthesis
RNA is useful both diagnostically and analytically.
Those cytoplasmic RNA molecules that serve as tem-
Information within the single strand of RNA is con- plates for protein synthesis (ie, that transfer genetic in-
tained in its sequence (―primary structure‖) of purine formation from DNA to the protein-synthesizing ma-
and pyrimidine nucleotides within the polymer. The chinery) are designated messenger RNAs, or mRNAs.
sequence is complementary to the template strand of Many other cytoplasmic RNA molecules (ribosomal
the gene from which it was transcribed. Because of this RNAs; rRNAs) have structural roles wherein they con-
O

N
NH
5 G

CH2 N NH2 NH2


N

O
O
P
O O
CH2 N
H H
H H
O NH
HO O

P
O
H H CH2 N NH2
H H
O N
O
HO O
P
CH2 N N
H H
H H
O
O
O
HO
P
H H
H H
O
3 HO
P

Figure 35–6. A segment of a ribonucleic acid (RNA) molecule in which the purine and pyrimidine bases—
guanine (G), cytosine (C), uracil (U), and adenine (A)—are held together by phosphodiester bonds between ribo-
syl moieties attached to the nucleobases by N-glycosidic bonds. Note that the polymer has a polarity as indi-
cated by the labeled 3- and 5-attached phosphates.

tribute to the formation and function of ribosomes (the The genetic material for some animal and plant
organellar machinery for protein synthesis) or serve as viruses is RNA rather than DNA. Although some RNA
adapter molecules (transfer RNAs; tRNAs) for the viruses never have their information transcribed into a
translation of RNA information into specific sequences DNA molecule, many animal RNA viruses—specifi-
of polymerized amino acids. cally, the retroviruses (the HIV virus, for example)—are
Some RNA molecules have intrinsic catalytic activ- transcribed by an RNA-dependent DNA polymerase,
ity. The activity of these ribozymes often involves the the so-called reverse transcriptase, to produce a dou-
cleavage of a nucleic acid. An example is the role of ble-stranded DNA copy of their RNA genome. In
RNA in catalyzing the processing of the primary tran- many cases, the resulting double-stranded DNA tran-
script of a gene into mature messenger RNA. script is integrated into the host genome and subse-
Much of the RNA synthesized from DNA templates quently serves as a template for gene expression and
in eukaryotic cells, including mammalian cells, is de- from which new viral RNA genomes can be tran-
graded within the nucleus, and it never serves as either a scribed.
structural or an informational entity within the cellular
cytoplasm. RNA Is Organized in Several
In all eukaryotic cells there are small nuclear RNA Unique Structures
(snRNA) species that are not directly involved in pro-
tein synthesis but play pivotal roles in RNA processing. In all prokaryotic and eukaryotic organisms, three main
These relatively small molecules vary in size from 90 to classes of RNA molecules exist: messenger RNA
about 300 nucleotides (Table 35–1). (mRNA), transfer RNA (tRNA), and ribosomal RNA
Table 35–1. Some of the species of small stable
Loop
RNAs found in mammalian cells.

C G 

C G
G C
A U 

A U
A U
U G 
U G

Stem
C C
G C

U A
U A 
U C 
U A Messenger RNAs, particularly in eukaryotes, have
C G some unique chemical characteristics. The 5 terminal
G C of mRNA is ―capped‖ by a 7-methylguanosine triphos-
phate that is linked to an adjacent 2-O-methyl ribonu-
cleoside at its 5-hydroxyl through the three phosphates
5 3 (Figure 35–10). The mRNA molecules frequently con-
Figure 35–7. Diagrammatic representation of the tain internal 6-methyladenylates and other 2-O-ribose
methylated nucleotides. The cap is involved in the
secondary structure of a single-stranded RNA molecule
recognition of mRNA by the translating machinery,
in which a stem loop, or ―hairpin,‖ has been formed and
and it probably helps stabilize the mRNA by preventing
is dependent upon the intramolecular base pairing.
the attack of 5-exonucleases. The protein-synthesizing
Note that A forms hydrogen bonds with U in RNA. machinery begins translating the mRNA into proteins
beginning downstream of the 5 or capped terminal.
The other end of most mRNA molecules, the 3-hy-
(rRNA). Each differs from the others by size, function, droxyl terminal, has an attached polymer of adenylate
and general stability. residues 20–250 nucleotides in length. The specific
function of the poly(A) “tail” at the 3-hydroxyl termi-
nal of mRNAs is not fully understood, but it seems that
A. MESSENGER RNA (MRNA) it maintains the intracellular stability of the specific
This class is the most heterogeneous in size and stabil- mRNA by preventing the attack of 3-exonucleases.
ity. All members of the class function as messengers Some mRNAs, including those for some histones, do
conveying the information in a gene to the protein- not contain poly(A). The poly(A) tail, because it will
synthesizing machinery, where each serves as a template form a base pair with oligodeoxythymidine polymers
on which a specific sequence of amino acids is polymer- attached to a solid substrate like cellulose, can be used
ized to form a specific protein molecule, the ultimate to separate mRNA from other species of RNA, includ-
gene product (Figure 35–9). ing mRNA molecules that lack this tail.

DNA strands:
Coding 5 —T G G A A T T G T G A G C G G A T A A C A A T T T C A C A C A G G A A A C A G C T A T G A C C A T G — 3
Template 3 —A C C T T A A C A C T C G C C T A T T G T T A A A G T G T G T C C T T T G T C G A T A C T G G T A C — 5
RNA 5 p A U U G U G A G C G G A U A A C A A U U U C A C A C A G G A A A C A G C U A U G A C C A U G 3
transcript

Figure 35–8. The relationship between the sequences of an RNA transcript and its gene, in which the cod-
ing and template strands are shown with their polarities. The RNA transcript with a 5 to 3 polarity is comple-
mentary to the template strand with its 3 to 5 polarity. Note that the sequence in the RNA transcript and its
polarity is the same as that in the coding strand, except that the U of the transcript replaces the T of the gene.
DNA
5 3
3 5

mRNA
5 3

Protein synthesis on mRNA template
5 3
 Figure 35–9. The expression of genetic in-
Ribosome Completed
protein formation in DNA into the form of an mRNA
molecule transcript. This is subsequently translated by
ribosomes into a specific protein molecule.

In mammalian cells, including cells of humans, the The D, TWC, and extra arms help define a specific
mRNA molecules present in the cytoplasm are not the tRNA.
RNA products immediately synthesized from the DNA Although tRNAs are quite stable in prokaryotes, they
template but must be formed by processing from a pre- are somewhat less stable in eukaryotes. The opposite is
cursor molecule before entering the cytoplasm. Thus, true for mRNAs, which are quite unstable in prokary-
in mammalian nuclei, the immediate products of gene otes but generally stable in eukaryotic organisms.
transcription constitute a fourth class of RNA mole-
cules. These nuclear RNA molecules are very heteroge- C. RIBOSOMAL RNA (RRNA)
neous in size and are quite large. The heterogeneous A ribosome is a cytoplasmic nucleoprotein structure
nuclear RNA (hnRNA) molecules may have a molecu- that acts as the machinery for the synthesis of proteins
lar weight in excess of 107, whereas the molecular from the mRNA templates. On the ribosomes, the
weight of mRNA molecules is generally less than 2  mRNA and tRNA molecules interact to translate into a
106. As discussed in Chapter 37, hnRNA molecules are specific protein molecule information transcribed from
processed to generate the mRNA molecules which then the gene. In active protein synthesis, many ribosomes
enter the cytoplasm to serve as templates for protein are associated with an mRNA molecule in an assembly
synthesis. called the polysome.
The components of the mammalian ribosome,
B. TRANSFER RNA (TRNA) which has a molecular weight of about 4.2  106 and a
tRNA molecules vary in length from 74 to 95 nu- sedimentation velocity of 80S (Svedberg units), are
cleotides. They also are generated by nuclear processing shown in Table 35–2. The mammalian ribosome con-
of a precursor molecule (Chapter 37). The tRNA mole- tains two major nucleoprotein subunits—a larger one
cules serve as adapters for the translation of the infor- with a molecular weight of 2.8  106 (60S) and a
mation in the sequence of nucleotides of the mRNA smaller subunit with a molecular weight of 1.4  106
into specific amino acids. There are at least 20 species (40S). The 60S subunit contains a 5S ribosomal RNA
of tRNA molecules in every cell, at least one (and often (rRNA), a 5.8S rRNA, and a 28S rRNA; there are also
several) corresponding to each of the 20 amino acids re- probably more than 50 specific polypeptides. The 40S
quired for protein synthesis. Although each specific subunit is smaller and contains a single 18S rRNA and
tRNA differs from the others in its sequence of nu- approximately 30 distinct polypeptide chains. All of the
cleotides, the tRNA molecules as a class have many fea- ribosomal RNA molecules except the 5S rRNA are
tures in common. The primary structure—ie, the nu- processed from a single 45S precursor RNA molecule in
cleotide sequence—of all tRNA molecules allows the nucleolus (Chapter 37). 5S rRNA is independently
extensive folding and intrastrand complementarity to transcribed. The highly methylated ribosomal RNA
generate a secondary structure that appears like a molecules are packaged in the nucleolus with the spe-
cloverleaf (Figure 35–11). cific ribosomal proteins. In the cytoplasm, the ribo-
All tRNA molecules contain four main arms. The somes remain quite stable and capable of many transla-
acceptor arm terminates in the nucleotides CpCpAOH. tion cycles. The functions of the ribosomal RNA
These three nucleotides are added posttranscription- molecules in the ribosomal particle are not fully under-
ally. The tRNA-appropriate amino acid is attached to stood, but they are necessary for ribosomal assembly
the 3-OH group of the A moiety of the acceptor arm. and seem to play key roles in the binding of mRNA to
OH OH

C C

H H
HC CH

H2N –
H2C 5 O NH2
O
P O
HN O
O P O–
CH3 5
O
O
O– P CH2
O
O
O
CAP
HC CH
H H
2
3 C C O

OCH3 NH

5 O
O CH2 N
O
P
O O
mRNA
O–
HC H H CH

3 C C

OH

P
O
O–

Figure 35–10. The cap structure attached to the 5 terminal of most eukaryotic messen-
ger RNA molecules. A 7-methylguanosine triphosphate (black) is attached at the 5 terminal
of the mRNA (shown in blue), which usually contains a 2-O-methylpurine nucleotide.
These modifications (the cap and methyl group) are added after the mRNA is transcribed
from DNA.

ribosomes and its translation. Recent studies suggest size from 90 to 300 nucleotides and are present in
that an rRNA component performs the peptidyl trans- 100,000–1,000,000 copies per cell.
ferase activity and thus is an enzyme (a ribozyme). Small nuclear RNAs (snRNAs), a subset of these
RNAs, are significantly involved in mRNA processing
and gene regulation. Of the several snRNAs, U1, U2,
D. SMALL STABLE RNA U4, U5, and U6 are involved in intron removal and the
A large number of discrete, highly conserved, and small processing of hnRNA into mRNA (Chapter 37). The
stable RNA species are found in eukaryotic cells. The U7 snRNA may be involved in production of the cor-
majority of these molecules are complexed with pro- rect 3 ends of histone mRNA—which lacks a poly(A)
teins to form ribonucleoproteins and are distributed in tail. The U4 and U6 snRNAs may also be required for
the nucleus, in the cytoplasm, or in both. They range in poly(A) processing.

You might also like