Sie sind auf Seite 1von 25

chapter

17
FATTY ACID CATABOLISM
17.1 Digestion, Mobilization, and Transport that make them especially suitable as storage fuels. The
of Fats 632 long alkyl chains of their constituent fatty acids are es-
17.2 Oxidation of Fatty Acids 637 sentially hydrocarbons, highly reduced structures with
an energy of complete oxidation (~38 kJ/g) more than
17.3 Ketone Bodies 650 twice that for the same weight of carbohydrate or pro-
tein. This advantage is compounded by the extreme
insolubility of lipids in water; cellular triacylglycerols
Jack Sprat could eat no fat, aggregate in lipid droplets, which do not raise the
His wife could eat no lean, osmolarity of the cytosol, and they are unsolvated. (In
And so between them both you see, storage polysaccharides, by contrast, water of solvation
They licked the platter clean. can account for two-thirds of the overall weight of the
John Clarke, Paroemiologia Anglo-Latina stored molecules.) And because of their relative chem-
(Proverbs English and Latin), 1639 ical inertness, triacylglycerols can be stored in large
quantity in cells without the risk of undesired chemical
reactions with other cellular constituents.
he oxidation of long-chain fatty acids to acetyl-CoA
T is a central energy-yielding pathway in many organ-
isms and tissues. In mammalian heart and liver, for
The properties that make triacylglycerols good stor-
age compounds, however, present problems in their role
as fuels. Because they are insoluble in water, ingested
example, it provides as much as 80% of the energetic triacylglycerols must be emulsified before they can be
needs under all physiological circumstances. The elec- digested by water-soluble enzymes in the intestine, and
trons removed from fatty acids during oxidation pass triacylglycerols absorbed in the intestine or mobilized
through the respiratory chain, driving ATP synthesis; from storage tissues must be carried in the blood bound
the acetyl-CoA produced from the fatty acids may be to proteins that counteract their insolubility. To over-
completely oxidized to CO2 in the citric acid cycle, re- come the relative stability of the COC bonds in a fatty
sulting in further energy conservation. In some species acid, the carboxyl group at C-1 is activated by attach-
and in some tissues, the acetyl-CoA has alternative fates. ment to coenzyme A, which allows stepwise oxidation
In liver, acetyl-CoA may be converted to ketone bod- of the fatty acyl group at the C-3, or , positionhence
ieswater-soluble fuels exported to the brain and other the name  oxidation.
tissues when glucose is not available. In higher plants, We begin this chapter with a brief discussion of the
acetyl-CoA serves primarily as a biosynthetic precursor, sources of fatty acids and the routes by which they travel
only secondarily as fuel. Although the biological role of to the site of their oxidation, with special emphasis on
fatty acid oxidation differs from organism to organism, the process in vertebrates. We then describe the chem-
the mechanism is essentially the same. The repetitive ical steps of fatty acid oxidation in mitochondria. The
four-step process, called  oxidation, by which fatty complete oxidation of fatty acids to CO2 and H2O takes
acids are converted into acetyl-CoA is the main topic of place in three stages: the oxidation of long-chain fatty
this chapter. acids to two-carbon fragments, in the form of acetyl-CoA
In Chapter 10 we described the properties of tria- ( oxidation); the oxidation of acetyl-CoA to CO2 in
cylglycerols (also called triglycerides or neutral fats) the citric acid cycle (Chapter 16); and the transfer of

631
632 Chapter 17 Fatty Acid Catabolism

electrons from reduced electron carriers to the mitochon- Micelle formation enormously increases the fraction of
drial respiratory chain (Chapter 19). In this chapter we lipid molecules accessible to the action of water-soluble
focus on the first of these stages. We begin our discus- lipases in the intestine, and lipase action converts tria-
sion of  oxidation with the simple case in which a fully cylglycerols to monoacylglycerols (monoglycerides) and
saturated fatty acid with an even number of carbon diacylglycerols (diglycerides), free fatty acids, and glyc-
atoms is degraded to acetyl-CoA. We then look briefly erol (step 2 ). These products of lipase action diffuse
at the extra transformations necessary for the degrada- into the epithelial cells lining the intestinal surface (the
tion of unsaturated fatty acids and fatty acids with an intestinal mucosa) (step 3 ), where they are recon-
odd number of carbons. Finally, we discuss variations verted to triacylglycerols and packaged with dietary
on the -oxidation theme in specialized organelles cholesterol and specific proteins into lipoprotein aggre-
peroxisomes and glyoxysomesand two less common gates called chylomicrons (Fig. 172; see also Fig.
pathways of fatty acid catabolism,  and  oxidation. The 171, step 4 ).
chapter concludes with a description of an alternative Apolipoproteins are lipid-binding proteins in the
fate for the acetyl-CoA formed by  oxidation in verte- blood, responsible for the transport of triacylglycerols,
brates: the production of ketone bodies in the liver. phospholipids, cholesterol, and cholesteryl esters be-
tween organs. Apolipoproteins (apo means detached
or separate, designating the protein in its lipid-free
17.1 Digestion, Mobilization, and form) combine with lipids to form several classes of
Transport of Fats lipoprotein particles, spherical aggregates with hy-
drophobic lipids at the core and hydrophilic protein side
Cells can obtain fatty acid fuels from three sources: fats
chains and lipid head groups at the surface. Various
consumed in the diet, fats stored in cells as lipid
combinations of lipid and protein produce particles of
droplets, and fats synthesized in one organ for export
different densities, ranging from chylomicrons and very-
to another. Some species use all three sources under
low-density lipoproteins (VLDL) to very-high-density
various circumstances, others use one or two. Verte-
lipoproteins (VHDL), which can be separated by ultra-
brates, for example, obtain fats in the diet, mobilize fats
centrifugation. The structures of these lipoprotein par-
stored in specialized tissue (adipose tissue, consisting
ticles and their roles in lipid transport are detailed in
of cells called adipocytes), and, in the liver, convert ex-
Chapter 21.
cess dietary carbohydrates to fats for export to other
The protein moieties of lipoproteins are recognized
tissues. On average, 40% or more of the daily energy re-
by receptors on cell surfaces. In lipid uptake from the
quirement of humans in highly industrialized countries
intestine, chylomicrons, which contain apolipoprotein
is supplied by dietary triacylglycerols (although most
C-II (apoC-II), move from the intestinal mucosa into the
nutritional guidelines recommend no more than 30% of
lymphatic system, and then enter the blood, which car-
daily caloric intake from fats). Triacylglycerols provide
ries them to muscle and adipose tissue (Fig. 171, step
more than half the energy requirements of some organs,
5 ). In the capillaries of these tissues, the extracellular
particularly the liver, heart, and resting skeletal muscle.
enzyme lipoprotein lipase, activated by apoC-II, hy-
Stored triacylglycerols are virtually the sole source of
drolyzes triacylglycerols to fatty acids and glycerol (step
energy in hibernating animals and migrating birds. Pro-
6 ), which are taken up by cells in the target tissues
tists obtain fats by consuming organisms lower in the
(step 7 ). In muscle, the fatty acids are oxidized for en-
food chain, and some also store fats as cytosolic lipid
ergy; in adipose tissue, they are reesterified for storage
droplets. Vascular plants mobilize fats stored in seeds
as triacylglycerols (step 8 ).
during germination, but do not otherwise depend on fats
The remnants of chylomicrons, depleted of most of
for energy.
their triacylglycerols but still containing cholesterol and
apolipoproteins, travel in the blood to the liver, where
Dietary Fats Are Absorbed in the Small Intestine
they are taken up by endocytosis, mediated by recep-
In vertebrates, before ingested triacylglycerols can be tors for their apolipoproteins. Triacylglycerols that en-
absorbed through the intestinal wall they must be con- ter the liver by this route may be oxidized to provide
verted from insoluble macroscopic fat particles to finely energy or to provide precursors for the synthesis of ke-
dispersed microscopic micelles. This solubilization is tone bodies, as described in Section 17.3. When the diet
carried out by bile salts, such as taurocholic acid (p. contains more fatty acids than are needed immediately
355), which are synthesized from cholesterol in the liver, for fuel or as precursors, the liver converts them to
stored in the gallbladder, and released into the small triacylglycerols, which are packaged with specific
intestine after ingestion of a fatty meal. Bile salts are apolipoproteins into VLDLs. The VLDLs are transported
amphipathic compounds that act as biological deter- in the blood to adipose tissues, where the triacylglyc-
gents, converting dietary fats into mixed micelles erols are removed and stored in lipid droplets within
of bile salts and triacylglycerols (Fig. 171, step 1 ). adipocytes.
17.1 Digestion, Mobilization, and Transport of Fats 633

Fats ingested
in diet

8 Fatty acids are oxidized


as fuel or reesterified
for storage.

Gallbladder Myocyte or
adipocyte

CO2

ATP

Small
intestine 7 Fatty acids enter cells.

1 Bile salts emulsify


dietary fats in the
small intestine, forming Lipoprotein lipase
mixed micelles.
6 Lipoprotein lipase,
activated by
apoC-II in the capillary,
2 Intestinal lipases Capillary converts triacylglycerols
degrade triacylglycerols. to fatty acids and glycerol.
Intestinal
mucosa 5 Chylomicrons move
through the lymphatic
system and
bloodstream
ApoC-II
to tissues.
3 Fatty acids and other breakdown
products are taken up by the Chylomicron
intestinal mucosa and converted
into triacylglycerols. 4 Triacylglycerols are incorporated,
with cholesterol and apolipoproteins,
into chylomicrons.

FIGURE 171 Processing of dietary lipids in vertebrates. Digestion


and absorption of dietary lipids occur in the small intestine, and the Apolipoproteins
fatty acids released from triacylglycerols are packaged and delivered
to muscle and adipose tissues. The eight steps are discussed in the
text. B-48

C-III

C-II
FIGURE 172 Molecular structure of a chylomicron. The surface is
a layer of phospholipids, with head groups facing the aqueous phase.
Triacylglycerols sequestered in the interior (yellow) make up more than
80% of the mass. Several apolipoproteins that protrude from the sur-
face (B-48, C-III, C-II) act as signals in the uptake and metabolism of Phospholipids
Cholesterol
chylomicron contents. The diameter of chylomicrons ranges from Triacylglycerols and
about 100 to 500 nm. cholesteryl esters
634 Chapter 17 Fatty Acid Catabolism

Hormones Trigger Mobilization acids can be oxidized for energy production. The hor-
of Stored Triacylglycerols mones epinephrine and glucagon, secreted in response
to low blood glucose levels, activate the enzyme adenylyl
Neutral lipids are stored in adipocytes (and in steroid- cyclase in the adipocyte plasma membrane (Fig. 173),
synthesizing cells of the adrenal cortex, ovary, and which produces the intracellular second messenger
testes) in the form of lipid droplets, with a core of sterol cyclic AMP (cAMP; see Fig. 1213). Cyclic AMP
esters and triacylglycerols surrounded by a monolayer dependent protein kinase (PKA) phosphorylates
of phospholipids. The surface of these droplets is coated perilipin A, and the phosphorylated perilipin causes
with perilipins, a family of proteins that restrict access hormone-sensitive lipase in the cytosol to move to
to lipid droplets, preventing untimely lipid mobilization. the lipid droplet surface, where it can begin hydrolyz-
When hormones signal the need for metabolic energy, ing triacylglycerols to free fatty acids and glycerol. PKA
triacylglycerols stored in adipose tissue are mobilized also phosphorylates hormone-sensitive lipase, doubling
(brought out of storage) and transported to tissues or tripling its activity, but the more than 50-fold increase
(skeletal muscle, heart, and renal cortex) in which fatty in fat mobilization triggered by epinephrine is due pri-
marily to perilipin phosphorylation. Cells with defective
perilipin genes have almost no response to increases in
Hormone Adenylyl
cyclase cAMP concentration; their hormone-sensitive lipase
1 does not associate with lipid droplets.
As hormone-sensitive lipase hydrolyzes triacylglyc-
Receptor
G5 erol in adipocytes, the fatty acids thus released (free
2
fatty acids, FFA) pass from the adipocyte into the
ATP cAMP
blood, where they bind to the blood protein serum al-
bumin. This protein (Mr 66,000), which makes up about
half of the total serum protein, noncovalently binds as
Fatty acid many as 10 fatty acids per protein monomer. Bound to
4 transporter
PKA
this soluble protein, the otherwise insoluble fatty acids
3 are carried to tissues such as skeletal muscle, heart, and
P Hormone-
renal cortex. In these target tissues, fatty acids dissoci-
7
P P sensitive ate from albumin and are moved by plasma membrane
P lipase b oxidation, transporters into cells to serve as fuel.
Perilipin P citric acid cycle,
respiratory chain About 95% of the biologically available energy of tri-
P
P 8
acylglycerols resides in their three long-chain fatty acids;
Lipid ATP only 5% is contributed by the glycerol moiety. The glyc-
droplet 6 CO2 erol released by lipase action is phosphorylated by glyc-
5 erol kinase (Fig. 174), and the resulting glycerol
Serum
Fatty acids albumin
3-phosphate is oxidized to dihydroxyacetone phosphate.
Triacyl- The glycolytic enzyme triose phosphate isomerase con-
glycerol Adipocyte Myocyte verts this compound to glyceraldehyde 3-phosphate,
which is oxidized via glycolysis.

Bloodstream
Fatty Acids Are Activated and Transported
FIGURE 173 Mobilization of triacylglycerols stored in adipose tis- into Mitochondria
sue. When low levels of glucose in the blood trigger the release of
The enzymes of fatty acid oxidation in animal cells are
glucagon, 1 the hormone binds its receptor in the adipocyte mem-
located in the mitochondrial matrix, as demonstrated in
brane and thus 2 stimulates adenylyl cyclase, via a G protein, to
1948 by Eugene P. Kennedy and Albert Lehninger. The
produce cAMP. This activates PKA, which phosphorylates 3 the
hormone-sensitive lipase and 4 perilipin molecules on the surface
fatty acids with chain lengths of 12 or fewer carbons
of the lipid droplet. Phosphorylation of perilipin permits hormone-
enter mitochondria without the help of membrane trans-
sensitive lipase access to the surface of the lipid droplet, where 5 it porters. Those with 14 or more carbons, which consti-
hydrolyzes triacylglycerols to free fatty acids. 6 Fatty acids leave the tute the majority of the FFA obtained in the diet or
adipocyte, bind serum albumin in the blood, and are carried in the released from adipose tissue, cannot pass directly
blood; they are released from the albumin and 7 enter a myocyte through the mitochondrial membranesthey must first
via a specific fatty acid transporter. 8 In the myocyte, fatty acids are undergo the three enzymatic reactions of the carnitine
oxidized to CO2, and the energy of oxidation is conserved in ATP, shuttle. The first reaction is catalyzed by a family of
which fuels muscle contraction and other energy requiring metabo- isozymes (different isozymes specific for fatty acids hav-
lism in the myocyte. ing short, intermediate, or long carbon chains) present
17.1 Digestion, Mobilization, and Transport of Fats 635

CH2OH
HO C H Glycerol in the outer mitochondrial membrane, the acyl-CoA
synthetases, which promote the general reaction
CH2OH
Fatty acid  CoA  ATP y8
8z fatty acylCoA  AMP  PPi
ATP
glycerol
kinase Thus, acyl-CoA synthetases catalyze the formation of a
ADP
thioester linkage between the fatty acid carboxyl group
CH2OH and the thiol group of coenzyme A to yield a fatty
acylCoA, coupled to the cleavage of ATP to AMP and
HO C H O L-Glycerol PPi. (Recall the description of this reaction in Chapter
CH2 O P O 3-phosphate 13, to illustrate how the free energy released by cleav-
O age of phosphoanhydride bonds in ATP could be cou-
NAD  pled to the formation of a high-energy compound; p.
glycerol 3-phosphate
dehydrogenase
XXX.) The reaction occurs in two steps and involves a
NADH  H fatty acyladenylate intermediate (Fig. 175).
CH2OH
Fatty acylCoAs, like acetyl-CoA, are high-energy
compounds; their hydrolysis to FFA and CoA has a large,
O C O Dihydroxyacetone negative standard free-energy change (G 31
CH2 O P O phosphate kJ/mol). The formation of a fatty acylCoA is made more
O
favorable by the hydrolysis of two high-energy bonds in
triose phosphate
ATP; the pyrophosphate formed in the activation reaction
isomerase is immediately hydrolyzed by inorganic pyrophosphatase
(left side of Fig. 175), which pulls the preceding activa-
H O
tion reaction in the direction of fatty acylCoA formation.
C
The overall reaction is
D-Glyceraldehyde
H C OH O
3-phosphate Fatty acid  CoA  ATP On
CH2 O P O fatty acylCoA  AMP  2Pi (171)
O G  34 kJ/mol

Fatty acylCoA esters formed at the cytosolic side


Glycolysis
of the outer mitochondrial membrane can be trans-
ported into the mitochondrion and oxidized to produce
FIGURE 174 Entry of glycerol into the glycolytic pathway. ATP, or they can be used in the cytosol to synthesize

O O O

O P O P O P O Adenosine ATP
MECHANISM FIGURE 175
  
Conversion of a fatty acid to a fatty O O O
acylCoA. The conversion is catalyzed O
by fatty acylCoA synthetase and R C Fatty acid
inorganic pyrophosphatase. Fatty acid O
activation by formation of the fatty
fatty acylCoA
acylCoA derivative occurs in two 1
synthetase

steps. In step 1 , the carboxylate ion
displaces the outer two ( and ) O
phosphates of ATP to form a fatty O O O P O Adenosine
acyladenylate, the mixed anhydride 
O P O P O  R C O 
Fatty acyladenylate
of a carboxylic acid and a phosphoric
O O O (enzyme-bound)
acid. The other product is PPi, an
Pyrophosphate CoA-SH
excellent leaving group that is
2
immediately hydrolyzed to two Pi, fatty acylCoA
inorganic synthetase AMP
pulling the reaction in the forward pyrophosphatase

direction. In step 2 , the thiol group of
O
coenzyme A carries out nucleophilic
2Pi R C Fatty acylCoA
attack on the enzyme-bound mixed
S-CoA
anhydride, displacing AMP and forming
the thioester fatty acylCoA. The G  19 kJ/mol G  15 kJ/mol
overall reaction is highly exergonic. (for the two-step process)
636 Chapter 17 Fatty Acid Catabolism

Outer mitochondrial Inner mitochondrial


membrane membrane

Cytosol Intermembrane Matrix


space
Carnitine
acyltransferase II
O O
R C R C
S-CoA Carnitine
Carnitine S-CoA
O
R C O
Carnitine R C CoA-SH
CoA-SH Carnitine

Carnitine Transporter
acyltransferase I

FIGURE 176 Fatty acid entry into mitochondria via the acyl-carnitine/ A, freeing carnitine to return to the intermembrane space through the
carnitine transporter. After fatty acylcarnitine is formed at the outer same transporter. Acyltransferase I is inhibited by malonyl-CoA, the
membrane or in the intermembrane space, it moves into the matrix first intermediate in fatty acid synthesis (see Fig. 211). This inhibition
by facilitated diffusion through the transporter in the inner membrane. prevents the simultaneous synthesis and degradation of fatty acids.
In the matrix, the acyl group is transferred to mitochondrial coenzyme

membrane lipids. Fatty acids destined for mitochondrial This three-step process for transferring fatty acids
oxidation are transiently attached to the hydroxyl group into the mitochondrionesterification to CoA, transes-
of carnitine to form fatty acylcarnitinethe second terification to carnitine followed by transport, and trans-
reaction of the shuttle. This transesterification is cat- esterification back to CoAlinks two separate pools of
alyzed by carnitine acyltransferase I (Mr 88,000), in coenzyme A and of fatty acylCoA, one in the cytosol,
the outer membrane. Either the acyl-CoA passes the other in mitochondria. These pools have different
through the outer membrane and is converted to the functions. Coenzyme A in the mitochondrial matrix is
carnitine ester in the intermembrane space (Fig. 176), largely used in oxidative degradation of pyruvate, fatty
or the carnitine ester is formed on the cytosolic face of acids, and some amino acids, whereas cytosolic coen-
the outer membrane, then moved across the outer mem- zyme A is used in the biosynthesis of fatty acids (see
brane to the intermembrane spacethe current evi- Fig. 2110). Fatty acylCoA in the cytosolic pool can be
dence does not reveal which. In either case, passage into used for membrane lipid synthesis or can be moved into
the intermembrane space (the space between the outer the mitochondrial matrix for oxidation and ATP pro-
and inner membranes) occurs through large pores duction. Conversion to the carnitine ester commits the
(formed by the protein porin) in the outer membrane. fatty acyl moiety to the oxidative fate.
The fatty acylcarnitine ester then enters the matrix by The carnitine-mediated entry process is the rate-
facilitated diffusion through the acyl-carnitine/carni- limiting step for oxidation of fatty acids in mitochondria
tine transporter of the inner mitochondrial membrane and, as discussed later, is a regulation point. Once in-
(Fig. 176). side the mitochondrion, the fatty acylCoA is acted upon
CH3
by a set of enzymes in the matrix.

CH3 N CH2 CH CH2 COO


CH3 OH SUMMARY 17.1 Digestion, Mobilization,
Carnitine
and Transport of Fats
In the third and final step of the carnitine shuttle,
the fatty acyl group is enzymatically transferred from The fatty acids of triacylglycerols furnish a
carnitine to intramitochondrial coenzyme A by carni- large fraction of the oxidative energy in
tine acyltransferase II. This isozyme, located on the animals. Dietary triacylglycerols are emulsified
inner face of the inner mitochondrial membrane, re- in the small intestine by bile salts, hydrolyzed
generates fatty acylCoA and releases it, along with free by intestinal lipases, absorbed by intestinal
carnitine, into the matrix (Fig. 176). Carnitine reen- epithelial cells, reconverted into
ters the intermembrane space via the acyl-carnitine/car- triacylglycerols, then formed into chylomicrons
nitine transporter. by combination with specific apolipoproteins.
17.2 Oxidation of Fatty Acids 637

Chylomicrons deliver triacylglycerols to tissues, Stage 1 Stage 2


where lipoprotein lipase releases free fatty CH3
acids for entry into cells. Triacylglycerols CH2
stored in adipose tissue are mobilized by a CH2  Oxidation
8 Acetyl-CoA
hormone-sensitive triacylglycerol lipase. The CH2
released fatty acids bind to serum albumin and CH2
are carried in the blood to the heart, skeletal CH2
muscle, and other tissues that use fatty acids CH2
for fuel. e CH2
Once inside cells, fatty acids are activated at CH2
the outer mitochondrial membrane by CH2 Citric
conversion to fatty acylCoA thioesters. Fatty CH2 acid cycle
acylCoA to be oxidized enters mitochondria in CH2
three steps, via the carnitine shuttle. CH2
CH2 64e
16CO2
CH2
17.2 Oxidation of Fatty Acids C O
As noted earlier, mitochondrial oxidation of fatty acids O
takes place in three stages (Fig. 177). In the first
stage oxidationfatty acids undergo oxidative re-
moval of successive two-carbon units in the form of NADH, FADH2
Stage 3
acetyl-CoA, starting from the carboxyl end of the fatty e
acyl chain. For example, the 16-carbon palmitic acid  1
(palmitate at pH 7) undergoes seven passes through the 2H  2 O2
Respiratory
oxidative sequence, in each pass losing two carbons as (electron-transfer)
acetyl-CoA. At the end of seven cycles the last two car- chain
H2O
bons of palmitate (originally C-15 and C-16) remain as
acetyl-CoA. The overall result is the conversion of the
16-carbon chain of palmitate to eight two-carbon acetyl ADP  Pi ATP
groups of acetyl-CoA molecules. Formation of each
acetyl-CoA requires removal of four hydrogen atoms FIGURE 177 Stages of fatty acid oxidation. Stage 1: A long-chain
(two pairs of electrons and four H) from the fatty acyl fatty acid is oxidized to yield acetyl residues in the form of acetyl-
moiety by dehydrogenases. CoA. This process is called  oxidation. Stage 2: The acetyl groups are
In the second stage of fatty acid oxidation, the oxidized to CO2 via the citric acid cycle. Stage 3: Electrons derived
from the oxidations of stages 1 and 2 pass to O2 via the mitochon-
acetyl groups of acetyl-CoA are oxidized to CO2 in the
drial respiratory chain, providing the energy for ATP synthesis by
citric acid cycle, which also takes place in the mito-
oxidative phosphorylation.
chondrial matrix. Acetyl-CoA derived from fatty acids
thus enters a final common pathway of oxidation with
the acetyl-CoA derived from glucose via glycolysis and The  Oxidation of Saturated Fatty Acids Has Four
pyruvate oxidation (see Fig. 161). The first two stages
Basic Steps
of fatty acid oxidation produce the reduced electron car-
riers NADH and FADH2, which in the third stage donate Four enzyme-catalyzed reactions make up the first stage
electrons to the mitochondrial respiratory chain, of fatty acid oxidation (Fig. 178a). First, dehydro-
through which the electrons pass to oxygen with the genation of fatty acylCoA produces a double bond
concomitant phosphorylation of ADP to ATP (Fig. between the  and  carbon atoms (C-2 and C-3), yield-
177). The energy released by fatty acid oxidation is ing a trans-2-enoyl-CoA (the symbol 2 designates
thus conserved as ATP. the position of the double bond; you may want to re-
We now take a closer look at the first stage of fatty view fatty acid nomenclature, p. 343.) Note that the new
acid oxidation, beginning with the simple case of a sat- double bond has the trans configuration, whereas the
urated fatty acyl chain with an even number of carbons, double bonds in naturally occurring unsaturated fatty
then turning to the slightly more complicated cases of acids are normally in the cis configuration. We consider
unsaturated and odd-number chains. We also consider the significance of this difference later.
the regulation of fatty acid oxidation, the -oxidative This first step is catalyzed by three isozymes of
processes as they occur in organelles other than mito- acyl-CoA dehydrogenase, each specific for a range of
chondria, and, finally, two less-general modes of fatty fatty-acyl chain lengths: very-long-chain acyl-CoA de-
acid catabolism,  oxidation and  oxidation. hydrogenase (VLCAD), acting on fatty acids of 12 to 18
638 Chapter 17 Fatty Acid Catabolism

carbons; medium-chain (MCAD), acting on fatty acids in the citric acid cycle (p. XXX); in both reactions the
of 4 to 14 carbons; and short-chain (SCAD), acting on enzyme is bound to the inner membrane, a double bond
fatty acids of 4 to 8 carbons. All three isozymes are flavo- is introduced into a carboxylic acid between the  and
proteins with FAD (see Fig. 1318) as a prosthetic  carbons, FAD is the electron acceptor, and electrons
group. The electrons removed from the fatty acylCoA from the reaction ultimately enter the respiratory chain
are transferred to FAD, and the reduced form of the de- and pass to O2, with the concomitant synthesis of about
hydrogenase immediately donates its electrons to an 1.5 ATP molecules per electron pair.
electron carrier of the mitochondrial respiratory chain, In the second step of the -oxidation cycle (Fig.
the electron-transferring flavoprotein (ETF) (see 178a), water is added to the double bond of the
Fig. 198). The oxidation catalyzed by an acyl-CoA de- trans-2-enoyl-CoA to form the L stereoisomer of
hydrogenase is analogous to succinate dehydrogenation -hydroxyacyl-CoA (3-hydroxyacyl-CoA). This re-
action, catalyzed by enoyl-CoA hydratase, is for-
mally analogous to the fumarase reaction in the citric
(a) 
(C16) R CH2 CH2 CH2 C S-CoA acid cycle, in which H2O adds across an  double
bond (p. XXX).
O Palmitoyl-CoA
In the third step, L--hydroxyacyl-CoA is dehydro-
acyl-CoA
FAD genated to form -ketoacyl-CoA, by the action of
dehydrogenase -hydroxyacyl-CoA dehydrogenase; NAD is the
FADH 2
electron acceptor. This enzyme is absolutely specific for
H the L stereoisomer of hydroxyacyl-CoA. The NADH
R CH2 C C C S-CoA
formed in the reaction donates its electrons to NADH
trans-2-
dehydrogenase, an electron carrier of the respiratory
H O chain, and ATP is formed from ADP as the electrons pass
Enoyl-CoA
H 2O to O2. The reaction catalyzed by -hydroxyacyl-CoA de-
enoyl-CoA
hydratase hydrogenase is closely analogous to the malate dehy-
drogenase reaction of the citric acid cycle (p. XXX).
OH The fourth and last step of the -oxidation cycle is
catalyzed by acyl-CoA acetyltransferase, more com-
R CH2 C CH2 C S-CoA
monly called thiolase, which promotes reaction of -
L- -Hydroxy-
H O ketoacyl-CoA with a molecule of free coenzyme A to
acyl-CoA
NAD split off the carboxyl-terminal two-carbon fragment of
 -hydroxyacyl-CoA
dehydrogenase
the original fatty acid as acetyl-CoA. The other product
NADH  H is the coenzyme A thioester of the fatty acid, now short-
ened by two carbon atoms (Fig. 178a). This reaction
R CH2 C CH2 C S-CoA
is called thiolysis, by analogy with the process of hy-
-Ketoacyl-CoA
O O drolysis, because the -ketoacyl-CoA is cleaved by re-
acyl-CoA CoA-SH
action with the thiol group of coenzyme A.
acetyltransferase The last three steps of this four-step sequence are
(thiolase)
catalyzed by either of two sets of enzymes, with the en-
zymes employed depending on the length of the fatty
(C14) R CH2 C S-CoA  CH3 C S-CoA acyl chain. For fatty acyl chains of 12 or more carbons,
O O the reactions are catalyzed by a multienzyme complex
(C14) Acyl-CoA Acetyl -CoA associated with the inner mitochondrial membrane, the
(myristoyl-CoA)
trifunctional protein (TFP). TFP is a heterooctamer
of 44 subunits. Each  subunit contains two activities,
the enoyl-CoA hydratase and the -hydroxyacyl-CoA
dehydrogenase; the  subunits contain the thiolase ac-
(b) tivity. This tight association of three enzymes may allow
C14 Acetyl -CoA
efficient substrate channeling from one active site to the
C12 Acetyl -CoA
C10 Acetyl -CoA FIGURE 178 The -oxidation pathway. (a) In each pass through this
C8 Acetyl -CoA four-step sequence, one acetyl residue (shaded in pink) is removed in
the form of acetyl-CoA from the carboxyl end of the fatty acyl chain
C6 Acetyl -CoA in this example palmitate (C16), which enters as palmitoyl-CoA. (b) Six
C4 Acetyl -CoA more passes through the pathway yield seven more molecules of
acetyl-CoA, the seventh arising from the last two carbon atoms of the
Acetyl -CoA 16-carbon chain. Eight molecules of acetyl-CoA are formed in all.
17.2 Oxidation of Fatty Acids 639

next, without diffusion of the intermediates away from process. Transfer of electrons from NADH or FADH2 to
the enzyme surface. When TFP has shortened the fatty O2 yields one H2O per electron pair. Reduction of O2 by
acyl chain to 12 or fewer carbons, further oxidations are NADH also consumes one H per NADH molecule:
catalyzed by a set of four soluble enzymes in the matrix. NADH  H 
12
O2 On NAD  H2O. In hibernating
As noted earlier, the single bond between methyl- animals, fatty acid oxidation provides metabolic energy,
ene (OCH2O) groups in fatty acids is relatively stable. heat, and waterall essential for survival of an animal
The -oxidation sequence is an elegant mechanism for that neither eats nor drinks for long periods (Box 171).
destabilizing and breaking these bonds. The first three Camels obtain water to supplement the meager supply
reactions of  oxidation create a much less stable COC available in their natural environment by oxidation of
bond, in which the  carbon (C-2) is bonded to two car- fats stored in their hump.
bonyl carbons (the -ketoacyl-CoA intermediate). The The overall equation for the oxidation of palmitoyl-
ketone function on the  carbon (C-3) makes it a good CoA to eight molecules of acetyl-CoA, including the
target for nucleophilic attack by the OSH of coenzyme electron transfers and oxidative phosphorylations, is
A, catalyzed by thiolase. The acidity of the  hydrogen
Palmitoyl-CoA  7CoA  7O2  28Pi  28ADP On
and the resonance stabilization of the carbanion gener-
8 acetyl-CoA  28ATP  7H2O (174)
ated by the departure of this hydrogen make the termi-
nal OCH2OCOOS-CoA a good leaving group, facilitating
Acetyl-CoA Can Be Further Oxidized
breakage of the  bond.
in the Citric Acid Cycle
The Four -Oxidation Steps Are Repeated to Yield The acetyl-CoA produced from the oxidation of fatty
Acetyl-CoA and ATP acids can be oxidized to CO2 and H2O by the citric acid
cycle. The following equation represents the balance
In one pass through the -oxidation sequence, one mol- sheet for the second stage in the oxidation of palmitoyl-
ecule of acetyl-CoA, two pairs of electrons, and four pro- CoA, together with the coupled phosphorylations of the
tons (H) are removed from the long-chain fatty third stage:
acylCoA, shortening it by two carbon atoms. The equa-
tion for one pass, beginning with the coenzyme A ester 8 Acetyl-CoA  16O2  80Pi  80ADP On
of our example, palmitate, is 8CoA  80ATP  16CO2  16H2O (175)

Palmitoyl-CoA  CoA  FAD  NAD  H2O On Combining Equations 174 and 175, we obtain the
myristoyl-CoA  acetyl-CoA FADH2  NADH  H overall equation for the complete oxidation of palmitoyl-
(172) CoA to carbon dioxide and water:

Following removal of one acetyl-CoA unit from palmitoyl- Palmitoyl-CoA  23O2  108Pi  108ADP On
CoA, the coenzyme A thioester of the shortened fatty CoA  108ATP  16CO2  23H2O (176)
acid (now the 14-carbon myristate) remains. The Table 171 summarizes the yields of NADH, FADH2,
myristoyl-CoA can now go through another set of four and ATP in the successive steps of palmitoyl-CoA oxida-
-oxidation reactions, exactly analogous to the first, to tion. Note that because the activation of palmitate to
yield a second molecule of acetyl-CoA and lauroyl-CoA, palmitoyl-CoA breaks both phosphoanhydride bonds in
the coenzyme A thioester of the 12-carbon laurate. ATP (Fig. 175), the energetic cost of activating a fatty
Altogether, seven passes through the -oxidation acid is equivalent to two ATP, and the net gain per mol-
sequence are required to oxidize one molecule of ecule of palmitate is 106 ATP. The standard free-energy
palmitoyl-CoA to eight molecules of acetyl-CoA (Fig. change for the oxidation of palmitate to CO2 and H2O
178b). The overall equation is is about 9,800 kJ/mol. Under standard conditions, the
Palmitoyl-CoA  7CoA  7FAD  7NAD  7H2O On energy recovered as the phosphate bond energy of ATP
8 acetyl-CoA  7FADH2  7NADH  7H (173) is 106 30.5 kJ/mol  3,230 kJ/mol, about 33% of the
theoretical maximum. However, when the free-energy
Each molecule of FADH2 formed during oxidation of the changes are calculated from actual concentrations of re-
fatty acid donates a pair of electrons to ETF of the res- actants and products under intracellular conditions (see
piratory chain, and about 1.5 molecules of ATP are gen- Box 131), the free-energy recovery is more than 60%;
erated during the ensuing transfer of each electron pair the energy conservation is remarkably efficient.
to O2. Similarly, each molecule of NADH formed deliv-
ers a pair of electrons to the mitochondrial NADH de- Oxidation of Unsaturated Fatty Acids Requires
hydrogenase, and the subsequent transfer of each pair
Two Additional Reactions
of electrons to O2 results in formation of about 2.5 mol-
ecules of ATP. Thus four molecules of ATP are formed The fatty acid oxidation sequence just described is typ-
for each two-carbon unit removed in one pass through ical when the incoming fatty acid is saturated (that is,
the sequence. Note that water is also produced in this has only single bonds in its carbon chain). However,
640 Chapter 17 Fatty Acid Catabolism

BOX 171 THE WORLD OF BIOCHEMISTRY

Fat Bears Carry Out  Oxidation in Their Sleep nating) level. Although expending about 25,000 kJ/day
Many animals depend on fat stores for energy during (6,000 kcal/day), the bear does not eat, drink, urinate,
hibernation, during migratory periods, and in other sit- or defecate for months at a time.
uations involving radical metabolic adjustments. One Experimental studies have shown that hibernat-
of the most pronounced adjustments of fat metabo- ing grizzly bears use body fat as their sole fuel. Fat
lism occurs in hibernating grizzly bears. These animals oxidation yields sufficient energy for maintenance of
remain in a continuous state of dormancy for periods body temperature, active synthesis of amino acids
as long as seven months. Unlike most hibernating and proteins, and other energy-requiring activities,
species, the bear maintains a body temperature of be- such as membrane transport. Fat oxidation also re-
tween 32 and 35 C, close to the normal (nonhiber- leases large amounts of water, as described in the text,
which replenishes water lost in breathing. The glyc-
erol released by degradation of triacylglycerols is con-
verted into blood glucose by gluconeogenesis. Urea
formed during breakdown of amino acids is reab-
sorbed in the kidneys and recycled, the amino groups
reused to make new amino acids for maintaining body
proteins.
Bears store an enormous amount of body fat in
preparation for their long sleep. An adult grizzly con-
sumes about 38,000 kJ/day during the late spring and
summer, but as winter approaches it feeds 20 hours a
day, consuming up to 84,000 kJ daily. This change in
feeding is a response to a seasonal change in hormone
secretion. Large amounts of triacylglycerols are
formed from the huge intake of carbohydrates during
the fattening-up period. Other hibernating species, in-
A grizzly bear prepares its hibernation nest, near the McNeil River cluding the tiny dormouse, also accumulate large
in Canada. amounts of body fat.

most of the fatty acids in the triacylglycerols and phos- the trans double bond of the 2-enoyl-CoA generated
pholipids of animals and plants are unsaturated, having during  oxidation. Two auxiliary enzymes are needed
one or more double bonds. These bonds are in the cis for  oxidation of the common unsaturated fatty acids:
configuration and cannot be acted upon by enoyl-CoA an isomerase and a reductase. We illustrate these aux-
hydratase, the enzyme catalyzing the addition of H2O to iliary reactions with two examples.

TABLE 171 Yield of ATP during Oxidation of One Molecule of Palmitoyl-CoA to CO2 and H2O
Number of NADH Number of ATP
Enzyme catalyzing the oxidation step or FADH2 formed ultimately formed*
Acyl-CoA dehydrogenase 7 FADH2 10.5
-Hydroxyacyl-CoA dehydrogenase 7 NADH 17.5
Isocitrate dehydrogenase 8 NADH 20
-Ketoglutarate dehydrogenase 8 NADH 20
Succinyl-CoA synthetase 8
Succinate dehydrogenase 8 FADH2 12
Malate dehydrogenase 8 NADH 20
Total 108

*
These calculations assume that mitochondrial oxidative phosphorylation produces 1.5 ATP per FADH2 oxidized and 2.5 ATP per NADH oxidized.

GTP produced directly in this step yields ATP in the reaction catalyzed by nucleoside diphosphate kinase (p. XXX).
17.2 Oxidation of Fatty Acids 641

Oleate is an abundant 18-carbon monounsaturated 12 9 O


1
fatty acid with a cis double bond between C-9 and C-10 C
(denoted 9). In the first step of oxidation, oleate is con- 18 S-CoA
verted to oleoyl-CoA and, like the saturated fatty acids, b oxidation Linoleoyl-CoA
(three cycles) 3 Acetyl-CoA cis-9,cis-12
enters the mitochondrial matrix via the carnitine shut-
tle (Fig. 176). Oleoyl-CoA then undergoes three passes 6 4 3(b) O
through the fatty acid oxidation cycle to yield three mol- C
ecules of acetyl-CoA and the coenzyme A ester of a 3, 12 5 2(a) S-CoA cis-3,cis-6
12-carbon unsaturated fatty acid, cis-3-dodecenoyl- 3, 2-enoyl-CoA
CoA (Fig. 179). This product cannot serve as a sub- isomerase
strate for enoyl-CoA hydratase, which acts only on trans 6
4 2(a) S-CoA
double bonds. The auxiliary enzyme 3,2-enoyl-CoA
C
isomerase isomerizes the cis-3-enoyl-CoA to the 12 5 3(b)
O trans-2, cis-6
trans-2-enoyl-CoA, which is converted by enoyl-CoA
b oxidation
hydratase into the corresponding L--hydroxyacyl-CoA (one cycle, and
(trans-2-dodecenoyl-CoA). This intermediate is now first oxidation
Acetyl-CoA
acted upon by the remaining enzymes of  oxidation of second cycle)

to yield acetyl-CoA and the coenzyme A ester of a 10- 5 4


2 S-CoA
carbon saturated fatty acid, decanoyl-CoA. The latter 1
C
undergoes four more passes through the pathway to 10 3
O trans-2, cis-4
yield five more molecules of acetyl-CoA. Altogether,
nine acetyl-CoAs are produced from one molecule of the NADPH  H
2,4-dienoyl-CoA
reductase
18-carbon oleate. NADP
The other auxiliary enzyme (a reductase) is re-
quired for oxidation of polyunsaturated fatty acidsfor O
5 3 1
C
10 4 2 S-CoA trans-3

O enoyl-CoA
9 isomerase
18 1
C
S-CoA O
3 1
Oleoyl-CoA C
10 4 2 S-CoA trans-2
 oxidation
(three cycles)
3 Acetyl-CoA b oxidation
(four cycles)

H H O 5 Acetyl-CoA
12
C FIGURE 1710 Oxidation of a polyunsaturated fatty acid. The
S-CoA
example here is linoleic acid, as linoleoyl-CoA (9,12). Oxidation re-
cis-3- quires a second auxiliary enzyme in addition to enoyl-CoA isomerase:
Dodecenoyl-CoA NADPH-dependent 2,4-dienoyl-CoA reductase. The combined action
3, 2-enoyl-CoA isomerase of these two enzymes converts a trans-2,cis-4-dienoyl-CoA inter-
mediate to the trans-2-enoyl-CoA substrate necessary for  oxidation.

H O
C
S-CoA
12
H
example, the 18-carbon linoleate, which has a cis-9,cis-
2
trans- - 12 configuration (Fig. 1710). Linoleoyl-CoA under-
Dodecenoyl-CoA
b oxidation
goes three passes through the -oxidation sequence to
(five cycles) yield three molecules of acetyl-CoA and the coenzyme
A ester of a 12-carbon unsaturated fatty acid with a cis-
3,cis-6 configuration. This intermediate cannot be
6 Acetyl-CoA
used by the enzymes of the -oxidation pathway; its
FIGURE 179 Oxidation of a monounsaturated fatty acid. Oleic acid, double bonds are in the wrong position and have the
as oleoyl-CoA (9), is the example used here. Oxidation requires an wrong configuration (cis, not trans). However, the com-
additional enzyme, enoyl-CoA isomerase, to reposition the double bined action of enoyl-CoA isomerase and 2,4-dienoyl-
bond, converting the cis isomer to a trans isomer, a normal interme- CoA reductase, as shown in Figure 1710, allows reen-
diate in  oxidation. try of this intermediate into the -oxidation pathway
642 Chapter 17 Fatty Acid Catabolism

and its degradation to six acetyl-CoAs. The overall re- H


sult is conversion of linoleate to nine molecules of H C H
acetyl-CoA.
H C H
Propionyl-CoA
Complete Oxidation of Odd-Number Fatty Acids C
Requires Three Extra Reactions CoA-S O

Although most naturally occurring lipids contain fatty HCO


3

acids with an even number of carbon atoms, fatty acids propionyl-CoA ATP
with an odd number of carbons are common in the lipids carboxylase biotin

of many plants and some marine organisms. Cattle and ADP  Pi


other ruminant animals form large amounts of the three-
carbon propionate (CH3OCH2OCOO) during fer- H
mentation of carbohydrates in the rumen. The propi-  H C H
O
onate is absorbed into the blood and oxidized by the
C C H
liver and other tissues. And small quantities of propi- D-Methylmalonyl-CoA
O C
onate are added as a mold inhibitor to some breads and
cereals, thus entering the human diet. CoA-S O
Long-chain odd-number fatty acids are oxidized in
the same pathway as the even-number acids, beginning methylmalonyl-CoA
at the carboxyl end of the chain. However, the substrate epimerase

for the last pass through the -oxidation sequence is a


fatty acylCoA with a five-carbon fatty acid. When this
is oxidized and cleaved, the products are acetyl-CoA and H H
O
propionyl-CoA. The acetyl-CoA can be oxidized in the H C H coenzyme
C C H
B12
citric acid cycle, of course, but propionyl-CoA enters a O CoA-S
different pathway involving three enzymes. methyl-
C C H malonyl-CoA H C H
Propionyl-CoA is first carboxylated to form the D CoA-S mutase
C C
stereoisomer of methylmalonyl-CoA (Fig. 1711) by  
O O O O
propionyl-CoA carboxylase, which contains the co-
factor biotin. In this enzymatic reaction, as in the pyru- L-Methylmalonyl-CoA Succinyl-CoA
vate carboxylase reaction (see Fig. 1616), CO2 (or its
hydrated ion, HCO 3 ) is activated by attachment to bi- FIGURE 1711 Oxidation of propionyl-CoA produced by  oxida-
otin before its transfer to the substrate, in this case the tion of odd-number fatty acids. The sequence involves the carboxy-
propionate moiety. Formation of the carboxybiotin in- lation of propionyl-CoA to D-methylmalonyl-CoA and conversion of
termediate requires energy, which is provided by the the latter to succinyl-CoA. This conversion requires epimerization of
cleavage of ATP to ADP and Pi. The D-methylmalonyl- D- to L-methylmalonyl-CoA, followed by a remarkable reaction in

CoA thus formed is enzymatically epimerized to its L which substituents on adjacent carbon atoms exchange positions (see
stereoisomer by methylmalonyl-CoA epimerase (Fig. Box 172).
1711). The L-methylmalonyl-CoA then undergoes an
intramolecular rearrangement to form succinyl-CoA,
transfer of long-chain fatty acylCoA into mitochondria.
which can enter the citric acid cycle. This rearrange-
The three-step process (carnitine shuttle) by which
ment is catalyzed by methylmalonyl-CoA mutase,
fatty acyl groups are carried from cytosolic fatty
which requires as its coenzyme 5-deoxyadenosyl-
acylCoA into the mitochondrial matrix (Fig. 176) is
cobalamin, or coenzyme B12, which is derived from
rate-limiting for fatty acid oxidation and is an important
vitamin B12 (cobalamin). Box 172 describes the role of
point of regulation. Once fatty acyl groups have entered
coenzyme B12 in this remarkable exchange reaction.
the mitochondrion, they are committed to oxidation to
acetyl-CoA.
Fatty Acid Oxidation Is Tightly Regulated
Malonyl-CoA, the first intermediate in the cytoso-
Oxidation of fatty acids consumes a precious fuel, and lic biosynthesis of long-chain fatty acids from acetyl-CoA
it is regulated so as to occur only when the need for en- (see Fig. 211), increases in concentration whenever
ergy requires it. In the liver, fatty acylCoA formed in the animal is well supplied with carbohydrate; excess
the cytosol has two major pathways open to it: (1)  ox- glucose that cannot be oxidized or stored as glycogen
idation by enzymes in mitochondria or (2) conversion is converted in the cytosol into fatty acids for storage
into triacylglycerols and phospholipids by enzymes in as triacylglycerol. The inhibition of carnitine acyltrans-
the cytosol. The pathway taken depends on the rate of ferase I by malonyl-CoA ensures that the oxidation of
17.2 Oxidation of Fatty Acids 643

fatty acids is inhibited whenever the liver is amply sup- frequency of carriers (individuals with this recessive
plied with glucose as fuel and is actively making tria- mutation on one of the two homologous chromosomes)
cylglycerols from excess glucose. is about 1 in 40, and about 1 individual in 10,000 has
the diseasethat is, has two copies of the mutant MCAD
O
allele and is unable to oxidize fatty acids of 6 to 12 car-

OOC CH2 C S-CoA bons. The disease is characterized by recurring episodes
Malonyl-CoA of a syndrome that includes fat accumulation in the liver,
Two of the enzymes of  oxidation are also regu- high blood levels of octanoic acid, low blood glucose
lated by metabolites that signal energy sufficiency. (hypoglycemia), sleepiness, vomiting, and coma. The
When the [NADH]/[NAD] ratio is high, -hydroxyacyl- pattern of organic acids in the urine helps in the diag-
CoA dehydrogenase is inhibited; in addition, high con- nosis of this disease: the urine commonly contains high
centrations of acetyl-CoA inhibit thiolase (Fig. 1712). levels of 6-carbon to 10-carbon dicarboxylic acids (pro-
duced by  oxidation) and low levels of urinary ketone
Genetic Defects in Fatty AcylCoA Dehydrogenases bodies (we discuss  oxidation below and ketone bod-
ies in Section 17.3). Although individuals may have no
Cause Serious Disease
symptoms between episodes, the episodes are very se-
Stored triacylglycerols are typically the chief rious; mortality from this disease is 25% to 60% in early
source of energy for muscle contraction, and an childhood. If the genetic defect is detected shortly
inability to oxidize fatty acids from triacylglycerols has after birth, the infant can be started on a low-fat, high-
serious consequences for health. The most common ge- carbohydrate diet. With early detection and careful man-
netic defect in fatty acid catabolism in U.S. and north- agement of the dietincluding avoiding long intervals
ern European populations is due to a mutation in the between meals, to prevent the body from turning to its
gene encoding the medium-chain acyl-CoA dehy- fat reserves for energythe prognosis for these indi-
drogenase (MCAD). Among northern Europeans, the viduals is good.

Dietary High blood Low blood


carbohydrate glucose glucose
Fatty acyl CoASH
1 Fatty carnitine
acylCoA
Insulin P Glucagon carnitine
2 5 acyl-
Carnitine
ACC transferase I
7
Inactive 4 Fatty
phosphatase 6 PKA Fatty acyl acylCoA
carnitine
Pi ACC FADH

3
8
Glucose AcetylCoA Malonyl-CoA b oxidation
glycolysis,
pyruvate multistep
NADH
dehydrogenase
complex
Fatty acids
Acetyl-CoA

Fatty acid Fatty acid


synthesis b oxidation Mitochondrion

FIGURE 1712 Coordinated regulation of fatty acid synthesis and (the first intermediate of fatty acid synthesis), and 4 malonyl-CoA in-
breakdown. When the diet provides a ready source of carbohydrate hibits carnitine acyltransferase I, thereby preventing fatty acid entry
as fuel,  oxidation of fatty acids is unnecessary and is therefore down- into the mitochondrial matrix.
regulated. Two enzymes are key to the coordination of fatty acid When blood glucose levels drop between meals, 5 glucagon re-
metabolism: acetyl-CoA carboxylase (ACC), the first enzyme in the lease activates cAMP-dependent protein kinase (PKA), which 6 phos-
synthesis of fatty acids (see Fig. 211 ), and carnitine acyl transferase I, phorylates and inactivates ACC. The concentration of malonyl-CoA
which limits the transport of fatty acids into the mitochondrial matrix falls, the inhibition of fatty acid entry into mitochondria is relieved,
for  oxidation (see Fig. 176). Ingestion of a high-carbohydrate meal and 7 fatty acids enter the mitochondrial matrix and 8 become the
raises the blood glucose level and thus 1 triggers the release of in- major fuel. Because glucagon also triggers the mobilization of fatty
sulin. 2 Insulin-dependent protein phosphatase dephosphorylates acids in adipose tissue, a supply of fatty acids begins arriving in the
ACC, activating it. 3 ACC catalyzes the formation of malonyl-CoA blood.
BOX 172 WORKING IN BIOCHEMISTRY

Coenzyme B12: A Radical Solution (a) H H O H H O


coenzyme B12
to a Perplexing Problem H C C C H C C C
In the methylmalonyl-CoA mutase reaction (see Fig. H C O methylmalonyl-CoA C H O
mutase
1711), the group OCOOS-CoA at C-2 of the original O S-CoA O S-CoA
propionate exchanges position with a hydrogen atom
at C-3 of the original propionate (Fig. 1a). Coenzyme L-Methylmalonyl-CoA Succinyl-CoA
B12 is the cofactor for this reaction, as it is for almost
(b) coenzyme B12
all enzymes that catalyze reactions of this general type C C C C
(Fig. 1b). These coenzyme B12dependent processes
H X X H
are among the very few enzymatic reactions in biol-
ogy in which there is an exchange of an alkyl or sub- FIGURE 1
stituted alkyl group (X) with a hydrogen atom on an
adjacent carbon, with no mixing of the transferred syl group (Fig. 2). This is a relatively weak bond; its
hydrogen atom with the hydrogen of the solvent, bond dissociation energy is about 110 kJ/mol, com-
H2O. How can the hydrogen atom move between two pared with 348 kJ/mol for a typical COC bond or 414
carbons without mixing with the enormous excess of kJ/mol for a COH bond. Merely illuminating the com-
hydrogen atoms in the solvent? pound with visible light is enough to break this CoOC
Coenzyme B12 is the cofactor form of vitamin B12, bond. (This extreme photolability probably accounts
which is unique among all the vitamins in that it for the absence of vitamin B12 in plants.) Dissociation
contains not only a complex organic molecule but an produces a 5-deoxyadenosyl radical and the Co2
essential trace element, cobalt. The com- O
H H
plex corrin ring system of vitamin B12
1 4
(colored blue in Fig. 2), to which cobalt OH HO
(as Co3) is coordinated, is chemically re- 2 3
lated to the porphyrin ring system of heme 5-Deoxy- H H
and heme proteins (see Fig. 51). A fifth adenosine N N
5
coordination position of cobalt is filled CH2 O
N
by dimethylbenzimidazole ribonucleotide N
C NH2
(shaded yellow), bound covalently by its NH2
CH2
3-phosphate group to a side chain of the O
O
corrin ring, through aminoisopropanol. CH2 C
The formation of this complex cofactor oc- H2N C H
CH3 CH2 NH2
curs in one of only two known reactions in CH2 CH3
which triphosphate is cleaved from ATP CH3 O Corrin
CH3 N ring
(Fig. 3); the other reaction is the forma- H C system
tion of S-adenosylmethionine from ATP N H CH2 NH2
Co3
and methionine (see Fig. 1818). CH2 N
O CH3 H
Vitamin B12 as usually isolated is called CH 2 N
cyanocobalamin, because it contains a C CH3
NH2 H CH2
cyano group (picked up during purification)
attached to cobalt in the sixth coordination CH 3 CH 3 CH2
CH2
position. In 5-deoxyadenosylcobalamin, O O C
CH2
the cofactor for methylmalonyl-CoA mu-
C NH
tase, the cyano group is replaced by the NH2
5-deoxyadenosyl group (red in Fig. 2), CH2 Amino-
covalently bound through C-5 to the cobalt. isopropanol
CH 3 N HC CH3
The three-dimensional structure of the co-
factor was determined by Dorothy Crowfoot O
CH 3 N
Hodgkin in 1956, using x-ray crystallo- Dimethyl- O P O
graphy. benzimidazole O H
ribonucleotide O
The key to understanding how coen- OH
zyme B12 catalyzes hydrogen exchange lies H CH 2OH
in the properties of the covalent bond be-
H H
tween cobalt and C-5 of the deoxyadeno- FIGURE 2

644
form of the vitamin. The the productlike radical, generating the product and
chemical function of 5-de- regenerating the deoxyadenosyl free radical. 5 The
oxyadenosylcobalamin is to bond re-forms between cobalt and the OCH2 group of
generate free radicals in this the deoxyadenosyl radical, destroying the free radical
way, thus initiating a series and regenerating the cofactor in its Co3 form, ready
of transformations such as to undergo another catalytic cycle. In this postulated
that illustrated in Figure 4 mechanism, the migrating hydrogen atom never exists
a postulated mechanism for as a free species and is thus never free to exchange
the reaction catalyzed by with the hydrogen of surrounding water molecules.
methylmalonyl-CoA mutase Vitamin B12 deficiency results in serious disease.
and a number of other coen- This vitamin is not made by plants or animals
Dorothy Crowfoot Hodgkin,
zyme B12dependent trans- and can be synthesized only by a few species of mi-
19101994
formations. croorganisms. It is required by healthy people in only
1 The enzyme first breaks the CoOC bond in the minute amounts, about 3 g/day. The severe disease
cofactor, leaving the coenzyme in its Co2 form and pernicious anemia results from failure to absorb vi-
producing the 5-deoxyadenosyl free radical. 2 This tamin B12 efficiently from the intestine, where it is
radical now abstracts a hydrogen atom from the sub- synthesized by intestinal bacteria or obtained from di-
strate, converting the substrate to a radical and pro- gestion of meat. Individuals with this disease do not
ducing 5-deoxyadenosine. 3 Rearrangement of the produce sufficient amounts of intrinsic factor, a gly-
substrate radical yields another radical, in which the coprotein essential to vitamin B12 absorption. The
migrating group X (OCOOS-CoA for methylmalonyl- pathology in pernicious anemia includes reduced pro-
CoA mutase) has moved to the adjacent carbon to form duction of erythrocytes, reduced levels of hemoglobin,
a radical that has the carbon skeleton of the eventual and severe, progressive impairment of the central nerv-
product (a four-carbon straight chain). The hydrogen ous system. Administration of large doses of vitamin B12
atom initially abstracted from the substrate is now part alleviates these symptoms in at least some cases.
of the OCH3 group of 5-deoxyadenosine. 4 One of
the hydrogens from this same OCH3 group (it can be Deoxyadenosine
the same one originally abstracted) is now returned to Deoxyadenosine
5-Deoxyadenosyl
H C H
Coenzyme CH2 free radical
B12 N N N N
H O H
Co3 Co2
1 4 1
OH HO N N N N C C
2 3 H X
O O O
H H Substrate
2
N N 5
CH2 O P O P O P O 5
N O O O
N Deoxyadenosine
ATP
NH2 Co Deoxyadenosine H C H C C
Cobalamin
H C H 5-Deoxy- H X
adenosyl
O O O N N free radical N N Substrate

radical
O P O P O P O Co 2
Co2
   N N N N
O O O

H O H 3 radical
4 rearrangement
OH HO
Deoxyadenosine
H H Coenzyme B12 Productlike
N N CH2 C C H C H C C
radical
X H H X
N
N Co Product N N
NH2 Co2
FIGURE 3 MECHANISM FIGURE 4 N N

645
646 Chapter 17 Fatty Acid Catabolism

More than 20 other human genetic defects in fatty Mitochondrion Peroxisome/glyoxysome


acid transport or oxidation have been documented, most
much less common than the defect in MCAD. One of the
most severe disorders results from loss of the long-chain O
R CH2 CH2 C
-hydroxyacyl-CoA dehydrogenase activity of the tri- S-CoA
functional protein, TFP. Other disorders include defects
O2 FAD FAD H2 O2
in the  or  subunits that affect all three activities of Respiratory
chain O2
TFP and cause serious heart disease and abnormal H2 O FADH2 FADH2
skeletal muscle. ATP H2O  12O2
H
O
R C C C
Peroxisomes Also Carry Out  Oxidation S-CoA
H
The mitochondrial matrix is the major site of fatty acid H 2O H2O
oxidation in animal cells, but in certain cells other com-
partments also contain enzymes capable of oxidizing OH
O
fatty acids to acetyl-CoA, by a pathway similar to, but R C CH2 C
S-CoA
not identical with, that in mitochondria. In plant cells, H
the major site of  oxidation is not mitochondria but
peroxisomes. O2 NAD+ NAD+
Respiratory NADH exported
In peroxisomes, membrane-enclosed organelles of chain for reoxidation
H2 O NADH NADH
animal and plant cells, the intermediates for  oxidation ATP O
of fatty acids are coenzyme A derivatives, and the O
process consists of four steps, as in mitochondrial  ox- R C CH2 C
S-CoA
idation (Fig. 1713): (1) dehydrogenation, (2) addition CoASH CoASH
of water to the resulting double bond, (3) oxidation of O
the -hydroxyacyl-CoA to a ketone, and (4) thiolytic R C
cleavage by coenzyme A. (The identical reactions also S-CoA
occur in glyoxysomes, as discussed below.) 
Citric O
Acetyl-CoA
One difference between the peroxisomal and mito- acid CH3 C exported
cycle S-CoA
chondrial pathways is in the chemistry of the first step.
In peroxisomes, the flavoprotein acyl-CoA oxidase that
introduces the double bond passes electrons directly to
O2, producing H2O2 (Fig. 1713). This strong and po-
FIGURE 1713 Comparison of  oxidation in mitochondria and in
tentially damaging oxidant is immediately cleaved to
peroxisomes and glyoxysomes. The peroxisomal/glyoxysomal system
H2O and O2 by catalase. Recall that in mitochondria,
differs from the mitochondrial system in two respects: (1) in the first
the electrons removed in the first oxidation step pass
oxidative step electrons pass directly to O2, generating H2O2, and
through the respiratory chain to O2 to produce H2O, and
(2) the NADH formed in the second oxidative step cannot be reoxi-
this process is accompanied by ATP synthesis. In per- dized in the peroxisome or glyoxysome, so reducing equivalents are
oxisomes, the energy released in the first oxidative step exported to the cytosol, eventually entering mitochondria. The acetyl-
of fatty acid breakdown is not conserved as ATP, but is CoA produced by peroxisomes and glyoxysomes is also exported; the
dissipated as heat. acetate from glyoxysomes (organelles found only in germinating seeds)
A second important difference between mito- serves as a biosynthetic precursor (see Fig. 1714). Acetyl-CoA pro-
chondrial and peroxisomal  oxidation in mam- duced in mitochondria is further oxidized in the citric acid cycle.
mals is in the specificity for fatty acylCoAs; the
peroxisomal system is much more active on very-long-
chain fatty acids such as hexacosanoic acid (26:0) and oxidize very-long-chain fatty acids, apparently for lack
on branched-chain fatty acids such as phytanic acid and of a functional transporter for these fatty acids in the
pristanic acid (see Fig. 1717). These less-common fatty peroxisomal membrane. Both defects lead to accumu-
acids are obtained in the diet from dairy products, the lation in the blood of very-long-chain fatty acids, espe-
fat of ruminant animals, meat, and fish. Their catabo- cially 26:0. XALD affects young boys before the age of
lism in the peroxisome involves several auxiliary en- 10 years, causing loss of vision, behavioral disturbances,
zymes unique to this organelle. The inability to oxidize and death within a few years.
these compounds is responsible for several serious hu- In mammals, high concentrations of fats in the diet
man diseases. Individuals with Zellweger syndrome result in increased synthesis of the enzymes of peroxi-
are unable to make peroxisomes and therefore lack all somal  oxidation in the liver. Liver peroxisomes do not
the metabolism unique to that organelle. In X-linked contain the enzymes of the citric acid cycle and cannot
adrenoleukodystrophy (XALD), peroxisomes fail to catalyze the oxidation of acetyl-CoA to CO2. Instead,
17.2 Oxidation of Fatty Acids 647

long-chain or branched fatty acids are catabolized to Seed triacylglycerols


shorter-chain products, such as hexanoyl-CoA, which lipases
are exported to mitochondria and completely oxidized.
Fatty acids
Plant Peroxisomes and Glyoxysomes Use Acetyl-CoA
 oxidation
from  Oxidation as a Biosynthetic Precursor
In plants, fatty acid oxidation does not occur primarily Acetyl-CoA
in mitochondria (as noted earlier) but in the peroxi- glyoxylate cycle
somes of leaf tissue and in the glyoxysomes of germi-
nating seeds. Plant peroxisomes and glyoxysomes are Oxaloacetate
similar in structure and function; glyoxysomes, which
occur only in germinating seeds, may be considered spe- gluconeogenesis

cialized peroxisomes. The biological role of  oxidation


in these organelles is to use stored lipids primarily to Glucose
Sucrose, Metabolic
provide but biosynthetic precursors, not energy. polysaccharides intermediates
During seed germination, stored triacylglycerols are
Amino acids Nucleotides
converted into glucose, sucrose, and a wide variety of
essential metabolites (Fig. 1714). Fatty acids released Energy
from the triacylglycerols are first activated to their coen-
zyme A derivatives and oxidized in glyoxysomes by the FIGURE 1714 Triacylglycerols as glucose source in seeds.  Oxi-
same four-step process that takes place in peroxisomes dation is one stage in a pathway that converts stored triacylglycerols
(Fig. 1713). The acetyl-CoA produced is converted via to glucose in germinating seeds. For more detail, see Figure 1622.
the glyoxylate cycle (see Fig. 1620) to four-carbon
precursors for gluconeogenesis (see Fig. 1418). Gly-
fatty acids (D-3-hydroxyacyl-CoA epimerase and 3,2-
oxysomes, like peroxisomes, contain high concentra-
enoyl-CoA isomerase); the fourth enzyme, thiolase, is a
tions of catalase, which converts the H2O2 produced by
separate, soluble polypeptide.
 oxidation to H2O and O2.
It is interesting that the enzymes that catalyze es-
sentially the reversal of  oxidation in the synthesis of
The -Oxidation Enzymes of Different Organelles
fatty acids are also organized differently in prokaryotes
Have Diverged during Evolution and eukaryotes; in bacteria, the seven enzymes needed
Although the -oxidation reactions in mitochondria are for fatty acid synthesis are separate polypeptides, but
essentially the same as those in peroxisomes and gly- in mammals, all seven activities are part of a single, huge
oxysomes, the enzymes (isozymes) differ significantly polypeptide chain (see Fig. 217). One advantage to the
between the two types of organelles. The differences cell in having several enzymes of the same pathway en-
apparently reflect an evolutionary divergence that oc- coded in a single polypeptide chain is that this solves
curred very early, with the separation of gram-positive the problem of regulating the synthesis of enzymes that
and gram-negative bacteria (see Fig. 16). must interact functionally; regulation of the expression
In mitochondria, the four -oxidation enzymes that of one gene ensures production of the same number of
act on short-chain fatty acylCoAs are separate, soluble active sites for all enzymes in the path. When each en-
proteins (as noted earlier), similar in structure to the zyme activity is on a separate polypeptide, some mech-
analogous enzymes of gram-positive bacteria (Fig. anism is required to coordinate the synthesis of all the
1715a). The gram-negative bacteria have four activities gene products. The disadvantage of having several ac-
in three soluable subunits (Fig. 1715b), and the eukary- tivities on the same polypeptide is that the longer the
otic enzyme system that acts on long-chain fatty acids polypeptide chain, the greater is the probability of a mis-
the trifunctional protein, TFPhas three enzyme activ- take in its synthesis: a single incorrect amino acid in the
ities in two subunits that are membrane-associated (Fig. chain may make all the enzyme activities in that chain
1715c). The -oxidation enzymes of plant peroxisomes useless. Comparison of the gene structures for these pro-
and glyoxysomes, however, form a complex of proteins, teins in many species may shed light on the reasons for
one of which contains four enzymatic activities in a sin- the selection of one or the other strategy in evolution.
gle polypeptide chain (Fig. 1715d). The first enzyme,
acyl-CoA oxidase, is a single polypeptide chain; the mul- The  Oxidation of Fatty Acids Occurs
tifunctional protein (MFP) contains the second and
in the Endoplasmic Reticulum
third enzyme activities (enoyl-CoA hydratase and
hydroxyacyl-CoA dehydrogenase) as well as two auxil- Although mitochondrial  oxidation, in which enzymes
iary activities needed for the oxidation of unsaturated act at the carboxyl end of a fatty acid, is by far the most
648 Chapter 17 Fatty Acid Catabolism

(a) Gram-positive bacteria and mitochondrial (b) Gram-negative bacteria


short-chain-specific system

Substrate
Enz1
Intermediate Substrate
Product
Product
Enz1
Enz2
Enz4 Enz4 Enz2

Intermediate
Enz3
Intermediate
Enz3

(c) Mitochondrial very-long- (d) Peroxisomal and glyoxysomal


chain-specific system system of plants

Substrate
Substrate
Product Enz1
Enz1
Matrix
Enz2
Enz3 MFP
Enz4 Enz2
Product
Enz4
Enz6
Enz3

Inner Enz5
membrane

FIGURE 1715 The enzymes of  oxidation. Shown here are the dif- activities reside in three polypeptides; enzymes 2 and 3 are parts of a
ferent subunit structures of the enzymes of  oxidation in gram-positive single polypeptide chain. (c) The very-long-chain-specific system of
and gram-negative bacteria, mitochondria, and plant peroxisomes and mitochondria is also composed of three polypeptides, one of which
glyoxysomes. Enz1 is acyl-CoA dehydrogenase; Enz2, enoyl-CoA hy- includes the activities of enzymes 2 and 3; in this case, the system is
dratase; Enz3, L--hydroxyacyl-CoA dehydrogenase; Enz4, thiolase; bound to the inner mitochondrial membrane. (d) In the peroxisomal
Enz5, D-3-hydroxyacyl-CoA epimerase, and Enz6, 3,2-enoyl-CoA iso- and glyoxysomal -oxidation systems of plants, enzymes 1 and 4 are
merase. (a) The four enzymes of  oxidation in gram-positive bacteria separate polypeptides, but enzymes 2 and 3, as well as two auxiliary
are separate, soluble entities, as are those of the short-chain-specific enzymes, are part of a single polypeptide chain, the multifunctional pro-
system of mitochondria. (b) In gram-negative bacteria, the four enzyme tein, MFP.

important catabolic fate for fatty acids in animal cells, The first step introduces a hydroxyl group onto the
there is another pathway in some species, including ver-  carbon (Fig. 1716). The oxygen for this group comes
tebrates, that involves oxidation of the  (omega) car- from molecular oxygen (O2) in a complex reaction that
bonthe carbon most distant from the carboxyl group. involves cytochrome P450 and the electron donor
The enzymes unique to  oxidation are located (in NADPH. Reactions of this type are catalyzed by mixed-
vertebrates) in the endoplasmic reticulum of liver and function oxidases, described in Box 211. Two more
kidney, and the preferred substrates are fatty acids of enzymes now act on the  carbon: alcohol dehydro-
10 or 12 carbon atoms. In mammals  oxidation is nor- genase oxidizes the hydroxyl group to an aldehyde, and
mally a minor pathway for fatty acid degradation, but aldehyde dehydrogenase oxidizes the aldehyde group
when  oxidation is defective (because of mutation or to a carboxylic acid, producing a fatty acid with a car-
a carnitine deficiency, for example) it becomes more boxyl group at each end. At this point, either end can
important. be attached to coenzyme A, and the molecule can en-
17.2 Oxidation of Fatty Acids 649

O
 COOH Phytanic acid
CH3 (CH2)10 C 

O
ATP, CoA-SH
NADPH, O2 phytanoil-CoA
mixed-function synthetase AMP, PPi
oxidase

NADP

O
CO S-CoA Phytanoyl-CoA
HO CH2 (CH2)10 C
O -Ketoglutarate, Ascorbate
2
 phytanoyl-CoA Fe
NAD
alcohol
hydroxylase CO2 , Succinate
dehydrogenase
NADH

O O CO S-CoA -Hydroxyphytanoyl-
C (CH2)10 C CoA
OH
H O

NAD
aldehyde
dehydrogenase
NADH -hydroxyphytanoyl-
CoA lyase Formyl-CoA Formic acid
O O CO2
C (CH2)10 C
O

O O Pristanal
C
H
 oxidation

O O O O 
NAD(P)
C (CH2)2 C C (CH2)4 C aldehyde

O O 
O O dehydrogenase NAD(P)H
Succinate Adipate (adipic acid)
Pristanic acid
FIGURE 1716 The  oxidation of fatty acids in the endoplasmic COOH
reticulum. This alternative to  oxidation begins with oxidation of the
carbon most distant from the  carbonthe  (omega) carbon. The
substrate is usually a medium-chain fatty acid; shown here is lauric
acid (laurate). This pathway is generally not the major route for ox-  oxidation
idative catabolism of fatty acids.
O
4,8,12-Trimethyltri-
C
decanoyl-CoA
S-CoA
ter the mitochondrion and undergo  oxidation by the
normal route. In each pass through the -oxidation
pathway, the double-ended fatty acid yields dicar- 
boxylic acids such as succinic acid, which can enter the O
citric acid cycle, and adipic acid (Fig. 1716). CH3CH2 C Propionyl-CoA
S-CoA
Phytanic Acid Undergoes  Oxidation in Peroxisomes
FIGURE 1717 The  oxidation of a branched-chain fatty acid (phy-
The presence of a methyl group on the  carbon tanic acid) in peroxisomes. Phytanic acid has a methyl-substituted 
of a fatty acid makes  oxidation impossible, and carbon and therefore cannot undergo  oxidation. The combined
these branched fatty acids are catabolized in peroxi- action of the enzymes shown here removes the carboxyl carbon of
somes of animal cells by  oxidation. In the oxidation phytanic acid, to produce pristanic acid, in which the  carbon is
of phytanic acid, for example (Fig. 1717), phytanoyl- unsubstituted, allowing oxidation. Notice that  oxidation of pristanic
CoA is hydroxylated on its  carbon, in a reaction that acid releases propionyl-CoA, not acetyl-CoA. This is further catabo-
involves molecular oxygen; decarboxylated to form an lized as in Figure 1711. (The details of the reaction that produces
aldehyde one carbon shorter; and then oxidized to the pristanal remain controversial.)
650 Chapter 17 Fatty Acid Catabolism

corresponding carboxylic acid, which now has no sub- specialize in the oxidation of very-long-chain
stituent on the  carbon and can be oxidized further by fatty acids and branched fatty acids. In
 oxidation. Refsums disease, resulting from a genetic glyoxysomes, in germinating seeds,  oxidation
defect in phytanoyl-CoA hydroxylase, leads to very high is one step in the conversion of stored lipids
blood levels of phytanic acid and severe neurological into a variety of intermediates and products.
problems including blindness and deafness. The reactions of  oxidation, occurring in the
endoplasmic reticulum, produce dicarboxylic
SUMMARY 17.2 Oxidation of Fatty Acids fatty acyl intermediates, which can undergo 
oxidation at either end to yield short
In the first stage of  oxidation, four reactions dicarboxylic acids such as succinate.
remove each acetyl-CoA unit from the carboxyl
end of a saturated fatty acylCoA:
(1) dehydrogenation of the  and  carbons 17.3 Ketone Bodies
(C-2 and C-3) by FAD-linked acyl-CoA
dehydrogenases, (2) hydration of the resulting In humans and most other mammals, acetyl-CoA formed
trans-2 double bond by enoyl-CoA hydratase, in the liver during oxidation of fatty acids can either en-
(3) dehydrogenation of the resulting ter the citric acid cycle (stage 2 of Fig. 177) or un-
L--hydroxyacyl-CoA by NAD-linked -
dergo conversion to the ketone bodies, acetone, ace-
hydroxyacyl-CoA dehydrogenase, and toacetate, and D--hydroxybutyrate, for export to
(4) CoA-requiring cleavage of the resulting other tissues. (The term bodies is a historical artifact;
-ketoacyl-CoA by thiolase, to form acetyl-CoA the term is occasionally applied to insoluble particles,
and a fatty acylCoA shortened by two but these compounds are quite soluble in blood and
carbons. The shortened fatty acylCoA then urine.) Acetone, produced in smaller quantities than
reenters the sequence. the other ketone bodies, is exhaled. Acetoacetate and
D--hydroxybutyrate are transported by the blood to tis-
In the second stage of fatty acid oxidation, the sues other than the liver (extrahepatic tissues), where
acetyl-CoA is oxidized to CO2 in the citric acid they are converted to acetyl-CoA and oxidized in the
cycle. A large fraction of the theoretical yield citric acid cycle, providing much of the energy required
of free energy from fatty acid oxidation is by tissues such as skeletal and heart muscle and the
recovered as ATP by oxidative phosphorylation, renal cortex. The brain, which preferentially uses glu-
the final stage of the oxidative pathway. cose as fuel, can adapt to the use of acetoacetate or
Malonyl-CoA, an early intermediate of fatty D--hydroxybutyrate under starvation conditions, when
acid synthesis, inhibits carnitine acyltransferase glucose is unavailable. The production and export of ke-
I, preventing fatty acid entry into mitochondria. tone bodies from the liver to extrahepatic tissues allow
This blocks fatty acid breakdown while continued oxidation of fatty acids in the liver when
synthesis is occurring. acetyl-CoA is not being oxidized in the citric acid cycle.
Genetic defects in the medium-chain acyl-CoA CH3 OCOCH3
dehydrogenase result in serious human disease, B
O
as do mutations in other components of the
-oxidation system. Acetone

Oxidation of unsaturated fatty acids requires O


J
two additional enzymes: enoyl-CoA isomerase CH3 OCOCH2 OC
B G 
and 2,4-dienoyl-CoA reductase. Odd-number O O
fatty acids are oxidized by the -oxidation Acetoacetate
pathway to yield acetyl-CoA and a molecule
of propionyl-CoA. This is carboxylated to OH O
A J
methylmalonyl-CoA, which is isomerized to CH3 OCOCH2 OC
A G 
succinyl-CoA in a reaction catalyzed by H O
methylmalonyl-CoA mutase, an enzyme
D- -Hydroxybutyrate
requiring coenzyme B12.
Peroxisomes of plants and animals, and Ketone Bodies, Formed in the Liver, Are Exported
glyoxysomes of plants, carry out  oxidation in
to Other Organs as Fuel
four steps similar to those of the mitochondrial
pathway in animals. The first oxidation step, The first step in the formation of acetoacetate, occurring
however, transfers electrons directly to O2, in the liver (Fig. 1718), is the enzymatic condensation
generating H2O2. Peroxisomes of animal tissues of two molecules of acetyl-CoA, catalyzed by thiolase;
17.3 Ketone Bodies 651

O O D stereoisomer; it does not act on L--hydroxyacyl-CoAs


J J
CH3 OC  CH3 OC and is not to be confused with L--hydroxyacyl-CoA
G G
S-CoA S-CoA dehydrogenase of the -oxidation pathway.
2 Acetyl-CoA In healthy people, acetone is formed in very small
amounts from acetoacetate, which is easily de-
thiolase carboxylated, either spontaneously or by the action of
CoA-SH
acetoacetate decarboxylase (Fig. 1718). Because
O O individuals with untreated diabetes produce large quan-
B J
CH3 OCOCH2 OC tities of acetoacetate, their blood contains significant
G
S-CoA amounts of acetone, which is toxic. Acetone is volatile
Acetoacetyl-CoA and imparts a characteristic odor to the breath, which
is sometimes useful in diagnosing diabetes.
Acetyl-CoA  H2O
HMG-CoA In extrahepatic tissues, D--hydroxybutyrate is ox-
synthase
CoA-SH idized to acetoacetate by D--hydroxybutyrate dehy-
drogenase (Fig. 1719). The acetoacetate is activated
O OH O
M A J to its coenzyme A ester by transfer of CoA from suc-
COCH2 OCOCH2 OC cinyl-CoA, an intermediate of the citric acid cycle (see

D A G
O CH3 S-CoA Fig. 167), in a reaction catalyzed by -ketoacyl-CoA
 -Hydroxy- -methylglutaryl-CoA transferase. The acetoacetyl-CoA is then cleaved by
(HMG-CoA) thiolase to yield two acetyl-CoAs, which enter the citric
acid cycle. Thus the ketone bodies are used as fuels.
HMG-CoA
lyase
The production and export of ketone bodies by the
Acetyl-CoA liver allow continued oxidation of fatty acids with only
O
minimal oxidation of acetyl-CoA. When intermediates of
O B the citric acid cycle are being siphoned off for glucose
M
COCH2 OCOCH3

D
O
Acetoacetate OH O
CH3 C CH2 C D- -Hydroxybutyrate
D- -hydroxybutyrate 
acetoacetate NADH H O
dehydrogenase
decarboxylase  H 
NAD
CO2 D- -hydroxybutyrate
NAD dehydrogenase
NADH  H
O O OH
B M A
O O
CH3 OCOCH3 COCH2 OCHOCH3

D
O CH3 C CH2 C Acetoacetate
Acetone D- -Hydroxybutyrate O
Succinyl-CoA
FIGURE 1718 Formation of ketone bodies from acetyl-CoA.  -ketoacyl-CoA
transferase
Healthy, well-nourished individuals produce ketone bodies at a rela- Succinate
tively low rate. When acetyl-CoA accumulates (as in starvation or un-
treated diabetes, for example), thiolase catalyzes the condensation of
O O
two acetyl-CoA molecules to acetoacetyl-CoA, the parent compound CH3 C CH2 C Acetoacetyl-CoA
of the three ketone bodies. The reactions of ketone body formation S-CoA
occur in the matrix of liver mitochondria. The six-carbon compound
-hydroxy--methylglutaryl-CoA (HMG-CoA) is also an intermediate CoA-SH
thiolase
of sterol biosynthesis, but the enzyme that forms HMG-CoA in that
pathway is cytosolic. HMG-CoA lyase is present only in the mito-
O O
chondrial matrix.
CH3 C  CH3 C
S-CoA S-CoA
2 Acetyl-CoA
this is simply the reversal of the last step of  oxidation.
The acetoacetyl-CoA then condenses with acetyl-CoA to FIGURE 1719 D--Hydroxybutyrate as a fuel. D--Hydroxybutyrate,
form -hydroxy--methylglutaryl-CoA (HMG-CoA), synthesized in the liver, passes into the blood and thus to other tis-
which is cleaved to free acetoacetate and acetyl-CoA. sues, where it is converted in three steps to acetyl-CoA. It is first ox-
The acetoacetate is reversibly reduced by D--hydroxy- idized to acetoacetate, which is activated with coenzyme A donated
butyrate dehydrogenase, a mitochondrial enzyme, to from succinyl-CoA, then split by thiolase. The acetyl-CoA thus formed
D--hydroxybutyrate. This enzyme is specific for the is used for energy production.
652 Chapter 17 Fatty Acid Catabolism

synthesis by gluconeogenesis, for example, oxidation of


cycle intermediates slowsand so does acetyl-CoA oxi-
Lipid droplets
dation. Moreover, the liver contains only a limited amount
of coenzyme A, and when most of it is tied up in acetyl-
CoA,  oxidation slows for want of the free coenzyme. Hepatocyte
The production and export of ketone bodies free coen- Acetoacetate,
D- -hydroxybutyrate,
zyme A, allowing continued fatty acid oxidation. acetone Acetoacetate and
D- -hydroxybutyrate

Ketone Bodies Are Overproduced in Diabetes ketone body exported as energy


CoA formation source for heart,
and during Starvation skeletal muscle,
kidney, and brain
Starvation and untreated diabetes mellitus lead
Fatty
to overproduction of ketone bodies, with several acids Acetyl-CoA
associated medical problems. During starvation, gluco-  oxidation
neogenesis depletes citric acid cycle intermediates, di- Oxaloacetate citric
verting acetyl-CoA to ketone body production (Fig. acid
cycle
1720). In untreated diabetes, when the insulin level is
gluconeogenesis
insufficient, extrahepatic tissues cannot take up glucose
Glucose exported
efficiently from the blood, either for fuel or for conver- Glucose as fuel for brain
sion to fat. Under these conditions, levels of malonyl- and other tissues
CoA (the starting material for fatty acid synthesis) fall,
inhibition of carnitine acyltransferase I is relieved, and
fatty acids enter mitochondria to be degraded to acetyl-
CoAwhich cannot pass through the citric acid cycle
because cycle intermediates have been drawn off for use FIGURE 1720 Ketone body formation and export from the liver.
as substrates in gluconeogenesis. The resulting accu- Conditions that promote gluconeogenesis (untreated diabetes, severely
mulation of acetyl-CoA accelerates the formation of ke- reduced food intake) slow the citric acid cycle (by drawing off ox-
tone bodies beyond the capacity of extrahepatic tissues aloacetate) and enhance the conversion of acetyl-CoA to acetoacetate.
to oxidize them. The increased blood levels of acetoac- The released coenzyme A allows continued  oxidation of fatty acids.
etate and D--hydroxybutyrate lower the blood pH,
causing the condition known as acidosis. Extreme
acidosis can lead to coma and in some cases death.
Ketone bodies in the blood and urine of untreated SUMMARY 17.3 Ketone Bodies
diabetics can reach extraordinary levelsa blood con-
centration of 90 mg/100 mL (compared with a normal The ketone bodiesacetone, acetoacetate, and
level of 3 mg/100 mL) and urinary excretion of 5,000 D--hydroxybutyrateare formed in the liver.

mg/24 hr (compared with a normal rate of 125 mg/ The latter two compounds serve as fuel
24 hr). This condition is called ketosis. molecules in extrahepatic tissues, through
Individuals on very low-calorie diets, using the fats oxidation to acetyl-CoA and entry into the
stored in adipose tissue as their major energy source, citric acid cycle.
also have increased levels of ketone bodies in their blood Overproduction of ketone bodies in
and urine. These levels must be monitored to avoid the uncontrolled diabetes or severely reduced
dangers of acidosis and ketosis (ketoacidosis). calorie intake can lead to acidosis or ketosis.

Key Terms
Terms in bold are defined in the glossary.
 oxidation XXX carnitine shuttle XXX methylmalonyl-CoA multifunctional protein
chylomicron XXX carnitine acyltransferase mutase XXX (MFP) XXX
apolipoprotein XXX I XXX coenzyme B12 XXX  oxidation XXX
lipoprotein XXX acyl-carnitine/carnitine pernicious anemia XXX mixed-function
perilipins XXX transporter XXX intrinsic factor XXX oxidases XXX
hormone-sensitive carnitine acyltransferase malonyl-CoA XXX  oxidation XXX
lipase XXX II XXX medium-chain acyl-CoA acidosis XXX
free fatty acids XXX trifunctional protein dehydrogenase ketosis XXX
serum albumin XXX (TFP) XXX (MCAD) XXX
Chapter 17 Problems 653

Further Reading
General metabolism and their nutritional management. Annu. Rev. Nutr.
Boyer, P.D. (1983) The Enzymes, 3rd edn, Vol. 16: Lipid 18, 179206.
Enzymology, Academic Press, Inc., San Diego, CA. Kerner, J. & Hoppel, C. (2000) Fatty acid import into mitochon-
Ferry, G. (1998) Dorothy Hodgkin: A Life, Cold Spring Harbor dria. Biochim. Biophys. Acta 1486, 117.
Laboratory Press, Cold Spring Harbor, NY. Kunau, W.H., Dommes, V., & Schulz, H. (1995) -Oxidation of
Fascinating biography of an amazing woman. fatty acids in mitochondria, peroxisomes, and bacteria: a century of
Gurr, M.I., Harwood, J.L., & Frayn; K.N. (2002) Lipid Biochem- continued progress. Prog. Lipid Res. 34, 267342.
istry: An Introduction, 5th edn, Blackwell Science, Oxford, UK. A good historical account and a useful comparison of  oxida-
tion in different systems.
Langin, D., Holm, C., & Lafontan, M. (1996) Adipocyte
hormone-sensitive lipase: a major regulator of lipid metabolism. Rinaldo, P., Matern, D., & Bennett, M.J. (2002) Fatty acid
Proc. Nutr. Soc. 55, 93109. oxidation disorders. Annu. Rev. Physiol. 64, 477502.
Advanced review of metabolic defects in fat oxidation, including
Ramsay, T.G. (1996) Fat cells. Endocrinol. Metab. Clin. N. Am.
MCAD mutations.
25, 847870.
A review of all aspects of fat storage and mobilization in Sherratt, H.S. (1994) Introduction: the regulation of fatty acid
adipocytes. oxidation in cells. Biochem. Soc. Trans. 22, 421422.
Introduction to reviews (in this journal issue) of various aspects
Scheffler, I.E. (1999) Mitochondria, Wiley-Liss, New York.
of fatty acid oxidation and its regulation.
An excellent book on mitochondrial structure and function.
Thorpe, C. & Kim, J.J. (1995) Structure and mechanism of
Wang, C.S., Hartsuck, J., & McConathy, W.J. (1992) Structure
action of the acyl-CoA dehydrogenases. FASEB J. 9, 718725.
and functional properties of lipoprotein lipase. Biochim. Biophys.
Short, clear description of the three-dimensional structure and
Acta 1123, 117.
catalytic mechanism of these enzymes.
Advanced-level discussion of the enzyme that releases fatty
acids from lipoproteins in the capillaries of muscle and adipose Peroxisomal  Oxidation
tissue. Graham, I.A. & Eastmond, P.J. (2002) Pathways of straight and
branched chain fatty acid catabolism in higher plants. Prog. Lipid
Mitochondrial  Oxidation Res. 41, 156181.
Bannerjee, R. (1997) The yin-yang of cobalamin biochemistry.
Chem. Biol. 4, 175186. Hashimoto, T. (1996) Peroxisomal -oxidation: enzymology and
A review of the biochemistry of coenzyme B12 reactions, includ- molecular biology. Ann. N. Y. Acad. Sci. 804, 8698.
ing the methylmalonyl-CoA mutase reaction. Mannaerts, G.P. & van Veldhoven, P.P. (1996) Functions and
Eaton, S., Bartlett, K., & Pourfarzam, M. (1996) Mammalian organization of peroxisomal -oxidation. Ann. N. Y. Acad. Sci.
mitochondrial -oxidation. Biochem. J. 320, 345357. 804, 99115.
A review of the enzymology of  oxidation, inherited defects in Wanders, R.J.A., van Grunsven, E.G., & Jansen, G.A. (2000)
this pathway, and regulation of the process in mitochondria. Lipid metabolism in peroxisomes: enzymology, functions and dys-
Eaton, S., Bursby, T., Middleton, B., Pourfarzam, M., Mills, functions of the fatty acid - and -oxidation systems in humans.
K., Johnson, A.W., & Bartlett, K. (2000) The mitochondrial Biochem. Soc. Trans. 28, 141148.
trifunctional protein: centre of a -oxidation metabolon? Biochem.
Ketone Bodies
Soc. Trans. 28, 177182.
Foster, D.W. & McGarry, J.D. (1983) The metabolic derange-
Short, intermediate-level review.
ments and treatment of diabetic ketoacidosis. N. Engl. J. Med.
Harwood, J.L. (1988) Fatty acid metabolism. Annu. Rev. Plant 309, 159169.
Physiol. Plant Mol. Biol. 39, 101138.
McGarry, J.D. & Foster, D.W. (1980) Regulation of hepatic fatty
Jeukendrup, A.E., Saris, W.H., & Wagenmakers, A.J. (1998) acid oxidation and ketone body production. Annu. Rev. Biochem.
Fat metabolism during exercise: a review. Part III: effects of nutri- 49, 395420.
tional interventions. Int. J. Sports Med. 19, 371379.
Robinson, A.M. & Williamson, D.H. (1980) Physiological roles
This paper is one of a series that reviews the factors that influ-
of ketone bodies as substrates and signals in mammalian tissues.
ence fat mobilization and utilization during exercise.
Physiol. Rev. 60, 143187.
Kerner, J. & Hoppel, C. (1998) Genetic disorders of carnitine

Problems
1. Energy in Triacylglycerols On a per-carbon basis, 2. Fuel Reserves in Adipose Tissue Triacylglycerols,
where does the largest amount of biologically available en- with their hydrocarbon-like fatty acids, have the highest en-
ergy in triacylglycerols reside: in the fatty acid portions or ergy content of the major nutrients.
the glycerol portion? Indicate how knowledge of the chemi- (a) If 15% of the body mass of a 70.0 kg adult consists
cal structure of triacylglycerols provides the answer. of triacylglycerols, what is the total available fuel reserve, in
654 Chapter 17 Fatty Acid Catabolism

both kilojoules and kilocalories, in the form of triacylglyc- tance of various ATP-producing pathways is the Vmax of cer-
erols? Recall that 1.00 kcal  4.18 kJ. tain enzymes of these pathways. The values of Vmax of sev-
(b) If the basal energy requirement is approximately eral enzymes from the pectoral muscles (chest muscles used
8,400 kJ/day (2,000 kcal/day), how long could this person sur- for flying) of pigeon and pheasant are listed below.
vive if the oxidation of fatty acids stored as triacylglycerols
Vmax
were the only source of energy?
(mol substrate/min/g tissue)
(c) What would be the weight loss in pounds per day
under such starvation conditions (1 lb  0.454 kg)? Enzyme Pigeon Pheasant
3. Common Reaction Steps in the Fatty Acid Oxida- Hexokinase 3.0 2.3
tion Cycle and Citric Acid Cycle Cells often use the Glycogen phosphorylase 18.0 120.0
same enzyme reaction pattern for analogous metabolic con- Phosphofructokinase-1 24.0 143.0
versions. For example, the steps in the oxidation of pyruvate Citrate synthase 100.0 15.0
to acetyl-CoA and of -ketoglutarate to succinyl-CoA, al- Triacylglycerol lipase 0.07 0.01
though catalyzed by different enzymes, are very similar. The
first stage of fatty acid oxidation follows a reaction sequence
closely resembling a sequence in the citric acid cycle. Use (a) Discuss the relative importance of glycogen metab-
equations to show the analogous reaction sequences in the olism and fat metabolism in generating ATP in the pectoral
two pathways. muscles of these birds.
(b) Compare oxygen consumption in the two birds.
4. Chemistry of the Acyl-CoA Synthetase Reaction (c) Judging from the data in the table, which bird is the
Fatty acids are converted to their coenzyme A esters in a re- long-distance flyer? Justify your answer.
versible reaction catalyzed by acyl-CoA synthetase: (d) Why were these particular enzymes selected for
comparison? Would the activities of triose phosphate iso-
R COO  ATP  CoA
merase and malate dehydrogenase be equally good bases for
O
comparison? Explain.
R C CoA  AMP  PPi
8. Effect of Carnitine Deficiency An individual
(a) The enzyme-bound intermediate in this reaction has developed a condition characterized by progressive
been identified as the mixed anhydride of the fatty acid and muscular weakness and aching muscle cramps. The symp-
adenosine monophosphate (AMP), acyl-AMP: toms were aggravated by fasting, exercise, and a high-fat diet.
O O The homogenate of a skeletal muscle specimen from the
Adenine patient oxidized added oleate more slowly than did control
R C O P O CH2 O
homogenates, consisting of muscle specimens from healthy
O H H individuals. When carnitine was added to the patients mus-
H H cle homogenate, the rate of oleate oxidation equaled that in
OH OH the control homogenates. The patient was diagnosed as hav-
ing a carnitine deficiency.
Write two equations corresponding to the two steps of (a) Why did added carnitine increase the rate of oleate
the reaction catalyzed by acyl-CoA synthetase. oxidation in the patients muscle homogenate?
(b) The acyl-CoA synthetase reaction is readily re- (b) Why were the patients symptoms aggravated by
versible, with an equilibrium constant near 1. How can this fasting, exercise, and a high-fat diet?
reaction be made to favor formation of fatty acylCoA? (c) Suggest two possible reasons for the deficiency of
5. Oxidation of Tritiated Palmitate Palmitate uniformly muscle carnitine in this individual.
labeled with tritium (3H) to a specific activity of 2.48 108 9. Fatty Acids as a Source of Water Contrary to legend,
counts per minute (cpm) per micromole of palmitate is added camels do not store water in their humps, which actually con-
to a mitochondrial preparation that oxidizes it to acetyl-CoA. sist of large fat deposits. How can these fat deposits serve as
The acetyl-CoA is isolated and hydrolyzed to acetate. The a source of water? Calculate the amount of water (in liters)
specific activity of the isolated acetate is 1.00 107 that a camel can produce from 1.0 kg of fat. Assume for sim-
cpm/mol. Is this result consistent with the -oxidation path- plicity that the fat consists entirely of tripalmitoylglycerol.
way? Explain. What is the final fate of the removed tritium?
10. Petroleum as a Microbial Food Source Some mi-
6. Compartmentation in  Oxidation Free palmitate croorganisms of the genera Nocardia and Pseudomonas can
is activated to its coenzyme A derivative (palmitoyl-CoA) in grow in an environment where hydrocarbons are the only food
the cytosol before it can be oxidized in the mitochondrion. If source. These bacteria oxidize straight-chain aliphatic hy-
palmitate and [14C]coenzyme A are added to a liver ho- drocarbons, such as octane, to their corresponding carboxylic
mogenate, palmitoyl-CoA isolated from the cytosolic fraction acids:
is radioactive, but that isolated from the mitochondrial frac-
tion is not. Explain. CH3(CH2)6CH3  NAD  O2 y
z
CH3(CH2)6COOH  NADH  H
7. Comparative Biochemistry: Energy-Generating
Pathways in Birds One indication of the relative impor- How could these bacteria be used to clean up oil spills? What
Chapter 17 Problems 655

would be some of the limiting factors to the efficiency of this 15. Role of FAD as Electron Acceptor Acyl-CoA dehy-
process? drogenase uses enzyme-bound FAD as a prosthetic group to
dehydrogenate the  and  carbons of fatty acylCoA. What
11. Metabolism of a Straight-Chain Phenylated Fatty
is the advantage of using FAD as an electron acceptor rather
Acid A crystalline metabolite was isolated from the urine
than NAD? Explain in terms of the standard reduction po-
of a rabbit that had been fed a straight-chain fatty acid con-
tentials for the Enz-FAD/FADH2 (E  0.219 V) and
taining a terminal phenyl group:
NAD/NADH (E  0.320 V) half-reactions.

CH2 (CH2)n COO 16.  Oxidation of Arachidic Acid How many turns of
the fatty acid oxidation cycle are required for complete oxi-
dation of arachidic acid (see Table 101) to acetyl-CoA?
A 302 mg sample of the metabolite in aqueous solution was
completely neutralized by 22.2 mL of 0.100 M NaOH. 17. Fate of Labeled Propionate If [3-14C]propionate
(a) What is the probable molecular weight and structure (14C in the methyl group) is added to a liver homogenate,
of the metabolite? 14
C-labeled oxaloacetate is rapidly produced. Draw a flow
(b) Did the straight-chain fatty acid contain an even or chart for the pathway by which propionate is transformed
an odd number of methylene (OCH2O) groups (i.e., is n even to oxaloacetate, and indicate the location of the 14C in
or odd)? Explain. oxaloacetate.
12. Fatty Acid Oxidation in Uncontrolled Dia- 18. Sources of H2O Produced in  Oxidation The com-
betes When the acetyl-CoA produced during  ox- plete oxidation of palmitoyl-CoA to carbon dioxide and wa-
idation in the liver exceeds the capacity of the citric acid ter is represented by the overall equation
cycle, the excess acetyl-CoA forms ketone bodiesacetone, Palmitoyl-CoA  23O2  108Pi  108ADP On
acetoacetate, and D--hydroxybutyrate. This occurs in CoA  16CO2  108ATP  23H2O
severe, uncontrolled diabetes: because the tissues cannot use
glucose, they oxidize large amounts of fatty acids instead. Water is also produced in the reaction
Although acetyl-CoA is not toxic, the mitochondrion must ADP  Pi On ATP  H2O
divert the acetyl-CoA to ketone bodies. What problem would
but is not included as a product in the overall equation. Why?
arise if acetyl-CoA were not converted to ketone bodies? How
does the diversion to ketone bodies solve the problem? 19. Biological Importance of Cobalt In cattle, deer,
sheep, and other ruminant animals, large amounts of pro-
13. Consequences of a High-Fat Diet with No Carbo-
pionate are produced in the rumen through the bacterial
hydrates Suppose you had to subsist on a diet of whale
fermentation of ingested plant matter. Propionate is the
blubber and seal blubber, with little or no carbohydrate.
principal source of glucose for these animals, via the route
(a) What would be the effect of carbohydrate depriva-
propionate n oxaloacetate n glucose. In some areas of
tion on the utilization of fats for energy?
the world, notably Australia, ruminant animals sometimes
(b) If your diet were totally devoid of carbohydrate,
show symptoms of anemia with concomitant loss of appetite
would it be better to consume odd- or even-numbered fatty
and retarded growth, resulting from an inability to trans-
acids? Explain.
form propionate to oxaloacetate. This condition is due to a
14. Metabolic Consequences of Ingesting -Fluoro- cobalt deficiency caused by very low cobalt levels in the
oleate The shrub Dichapetalum toxicarium, native to soil and thus in plant matter. Explain.
Sierra Leone, produces -fluorooleate, which is highly toxic
20. Fat Loss during Hibernation Bears expend about
to warm-blooded animals.
25 106 J/day during periods of hibernation, which may last
H H as long as seven months. The energy required to sustain life
is obtained from fatty acid oxidation. How much weight loss
F CH2 (CH2)7 C C (CH2)7 COO
(in kilograms) has occurred after seven months? How might
-Fluorooleate
ketosis be minimized during hibernation? (Assume the oxi-
This substance has been used as an arrow poison, and pow- dation of fat yields 38 kJ/g.)
dered fruit from the plant is sometimes used as a rat poison
(hence the plants common name, ratsbane). Why is this sub-
stance so toxic? (Hint: Review Chapter 16, Problem 13.)

Das könnte Ihnen auch gefallen