Sei sulla pagina 1di 30

LIPD

METABOLISM
Biologically Important Lipids
The Carnitine Shuttle

31

FattyAcid B-Oxidation

34
37
38

Ketone Bodies
FatryAcid Synthesis
Insulin Signaling in Lipid Metabolism
Metabolism of Glycerolipids and Sphingolipids
Lysosomal Storage Disorders - The Sphingolipidoses
Eicosanoids
Metabolic Changes During Fasting
Cholesterol
Lipid Transport
Disorders of Lipid Transport
Important Lipid Modulating Drugs
Important Cholesterol D erivatives
Vitamin D in Calcium Homeostasis
Important Diseases of Lipid Metabolism
Review Qrestions

33

4T

4L

43
45
47
47
48
52
53
54
56
57
58

Atherosclerotic

BiologicallylmportantLipids

BIOLO GICALLY IMP O RTA\IT LIPID

31

FattyAcids are hydrocarbon chains with a terminal carboxyl group. Saturated FattyAcids are fatLy acids that contain
no double bonds (all single bonds). Saturated fatty acids tend to have relatively high melting points and are generally
solid at room temperature. The most common saturated fatty acid is palmitic acid, which contains 16 carbons, shown
in figure 2.1. Monounsaturated FattyAcids are fatty acids that contain a single double bond (generally cis), while
polyunsaturated Fatty Acids are fatty acids that contain
multiple double bonds (generally cis).
There are multiple classification systems for unsaturated
fatty acids, and you should be familiar with them because
Figure 2.1. Palmitic Acid.
they occur repeatedly in various sources. The simplest, and
most common, is the ro system. The ar-carbon of a fatfy acid is the terminal carbon. This is also known as ar-l. The carbons are then numbered towards the carbory-terminus as <o-2, or3, a-4, etc., as shown in figure 2.2 in the black numbering scheme. The fatty acids are classified based on the first carbon at which a double bond occurs. For example, all
c.r-3 fatfy acids have a double bond between the o-3 and co-4 carbons. o-linolenic acid, an example of an o-3 fatty acid
is shown in figure 2.2.The other classification system relies of specifying the length of the carbon chain, the number of
double bonds, and the carbons at which the double bonds occur. This system counts the carbon of the carboxyl group
as carbon I and then counts toward the end of the chain (grey numbering system in figur e 2.2) .For example, a-linolenic
acid would be named 183;9,12,75 because it has 18 carbons, 3 double bonds, and the double bonds occur at carbon 9,
12, and I 5 of the grey numbering system. Both numbering systems exist and you should be familiar with them, although
you do not have to memorize the specific nomenclatures for particular fatty acids. It is, however, worthwhile to familiarize yourselves with a number of important a-3, o16, and ar-9 fatty acids that are of biological importance.
c)-1

(l}-3

CI-6

Essential Fatty Acids: There are three fatty acids that


are considered essential fatty acids because the human
body cannot synthesize them. These are Iinoleic acid,
c-linolenic acid and arachidonic acid. Arachidonic
acid is technically not an essential fatty acid because the
body can synthesize it from linolenic acid; however, deficiencies can occur, and arachidonic acid becomes essential when dietary intake of linoleic acid is insufficient.

Figure 2.2. a-linolenic Acid.


A Trans Fafty Acid is an an unsaturated fatty acid that
contains one or more trans double bond. These are rare in nature, and are usually the product of partial hydrogenation, an industrial process that serves to solidify liquid fats.
Simple Fatty Acids and Nutrition: Monounsaturated and Polyunsaturated fatty acids appear to have a number of
health benefits. These include lowering the risk of atherosclerosis and coronary heart disease. On the other hand., saturated fatty acids and trans fatty acids appear to have a number of detrimental effects. These include increased risk of
atherosclerosis and cbronary heart disease (by increasing plasma LDL levels, reducing plasma HDL levels (only trans
fats), and overall increasing the LDL/HDLratio). Eicosapentaenoic acid (Ufe) is an ar-3 fatty acid present in fish oils
that has been promoted as having anti-inflammatory effects. This will be described subsequently in the discussion of
eicosanoids. Docosahexaenoic acid (DHA) is another c.r-3 fatty acids present in fish oils that has been promoted as
necessary for proper brain and retina function. There is evidence that patients with retinitis pigmentosa are defi-

cientinDHA.

Simple Lipids are esters of fatty acids with various alcohols. Fats and Oils are esters of fatty acids with 6[pl, and
include monoacylglycerols (monoglycerides), diacylglycerols (diglycerides), and triacylglycerols (triglycerides).Waxes are esters of fatty acid with high molecular weight alcohols. Interesterified Fat are unnatural fats that have some demonstrated negative health consequences and are produced through hydrogenation of unsaturated fats in industry.
The glycerol backbone of simple fats has a nomenclature system in
which the carbons are labeled snl, sn2, and sn3 as shown in figure
2.3. Because enzymes are chiral, they will usually act upon one of
the three faffy acid chains of a triacylglycerol. This is especially important within the context of pancreatic lipases that digest lipids by
selectively cleaving particular fatty acids from the glycerol backbone,
a topic that is generally covered in more detail in gastrointestinal
physiology courses.

1,r,-e-t-*,,

o
il=
ll

Rz-C-O!>-v<n

e;

ll

xF-o-C-o*R3
Figure 2.3. A Simple Fat.

Complex Lipids are modified esters of fatty acids.


Glycerophospholipids (phospholipids) consist of two fatty acids attached to a glycerol backbone at the snl and sn2
positions and a phosphate molecule attached at the sn3 position. The glycerophospholipid is named according to the
group (R) attached to the phosphate. A typical glycerophospholipid is shown in figure 2.4.
PhosphatidicAcid (Phosphatidate) - R= H
o
Phosphatidylcholines (lecithins) - R = choline CHTCHTN(CH.)3.
Dipalmitoyl lecithin - a phosphatidylcholine with two palmitic acid
o
cH2-o-c-Rr
side chains; this is pulmonary surfactant, the degradation of which
results in Acute Respiratory Distress Syndrome (anOS)
,l
R2_C_O_9H
O
Phosphatidylethanolamines - R = ethanolamine CHTCHTNHT*
Phosphatidylserines - R = serine
Phosphatidylinositols - R= myoinositol (a sugar)
Cardiolipins - a diphosphatidylglycerol (R = another phospholip6e
id); these are the major lipids of mitochondrial membranes
Plasmalogens - a modified glycerophospholipid-like molecule that
Figure 2.4. The General Phospholipid.
has a fatty ether linkage at the snl position of the glycerol. This is
shown in figure 2.5 in bold. Plasmalogens constitute as much as 10% of the phospholipids of the brain and muscle. Plasmalogens may also contain choline, ethanolamine, serine , or inositol, as the R group on the phosphate and are named

ll

1ll

Jll

Hzc-o-?-o-*

accordingly.

Phosphatidalethanolamine - largelyfound in the brain and nervous tissues


Phosphatidalcholine - largelyfound in the heart

Multiple Sclerosis is a demyelinating disorder of the brain, the


symptoms ofwhich result from the loss ofphospholipids and ethan6[amins plasmalogen (phosphatidalethanolamine) from the
myelin of the white matter of the brain.

H,C

il

2l

R2-C-O

-cJ

HzC

HH
-o-c:c-Rr

HO

il

-o-P-o-R
I

5o
Sphingolipids are esters of fatty acids that contain a molecule of
sphingosine in place of the glycerol backbone of fats and glyceroFigure 2.5. The Plasmalogen.
phospholipids. To the sphingosine backbone is attached one fatry
acid and one variable group (n). fhe sphingosine backbone and
fatty acid together are known as ceramide. The general structure of sphingolipids is shown infigarc2.6.
Sphingomyelins - R= phosphorylcholine; these are present in the myelin sheaths of neurons and a structure is shown
infigure2,7

_-

The Carnitine
Glycolipids (Glycosphingolipids)

Shuttle

33

all glycolipids have

a ceramide backbone and are hence more properly classified as glycosphingolipids. These are sphingolipids in
which the Rgroup is a monosaccharide or oligosaccharide. An example of a glycolipid is galactosylceramide,
where R - galactose; it is found in abundance in the
brain and other nervous tissues and shown in figure

Figure 2.6. The Sphingotipid.

2.8.

Gangliosides - R = an oligosaccharide that


containi a sialic acid (such as neuraminidic
acid - NeuAc). Gangliosides are found in
neurons in high concentrations and are also
found in various other tissues, where they
serve as cell surface recognition molecules

and as cell surface receptors. Gangliosides


are basically complex glycolipids.
Other sphingolipids include sulfosphingolipids and aminosphingolipids.

Figure

2.7 .

The Sphingomyelin.

Ceramide

Sphingosine

THE CARI\TITINE SHUTTLE

(e.9. cerebronic acid)

r-

A(*,,4[#(cH3]6-cH,
-t

The first step in fatfy acid catabolism is the transport ofthe fatty acid to the appropriate sub-cellular
localization. F"tty acid oxidatiory as we will learry
takes place in the mitochondrial matrix. However,
fatty acids are generally found in the cytosol. The
purpose of the carnitine shuttle, schematized in
figure 2.9, is to transport fatty acids into the mitochondrial matrix.

First, long chain fatty acids are activated by CoA in


the cytosol byAcyl-CoA synthetase, an enzyme of
the outer mitochondrial membrane, in an ATP dependent fashion. This is the one step in fatty acid
degradation that expends AIP. This step requires
the equivalent of two AIPs. This is because ATP is
Figure 2.8. The Gtycolipid.
converted to AMP and pyrophosphate during this
process, with pyrophosphate further degradated by
pyrophosphatase into two inorganic phosphates. I do not discuss the details of this process here.

1""'"".''"

Acyl-CoAs are freelypermeable through the outermitochondrial membrane. However, they cannotpenetrate the inner
mitochondrial membrane. This is the role of the carnitine shuttle, shown in figure 2.10. Carnitine acts as a shuttle protein that transfers the fatty acid across the inner mitochondrial membrane. Carnitine palnitoyl transferase I serves to
exchange CoA and carnitine to form an acyl-carnitine from the acyl-CoA within the intermembrane space. Carnitine

34

Lipid Metabolism

Acylcarnitine

AfP

CoASH

B-oxidation

Figure 2.9.The Carnitine Shuttle allows for the transport of activated fatty acids in the form of acylCoA into the mitochondrial matrix, where oxidation occurs. CPTI is Carnitine Palmitoyl Transferase
I, and is the rate limiting step of the shuttle. CPTII is Carnitine Palmitoyl Transferase II.
Acylcarnitine Translocase then allows the acylcarnitine to enter the mitochondrial matrix in exchange for a free carnitine molecule. Within the matrix, carnitine palmitoyl-transferase II then serves to convert acyl-carnitine back to acylCoA. The acyl-CoA is then metabolized within the mitochondrial matrix by a process known as p-oxidation. Carnitine
Palmitoyl-transferase I is generally regarded as the rate limiting enzyme of fatty acid oxidation. As such, it is highty regulated. Malonyl-CoA, a key intermediate in fatty acid synthesis, inhibits CPTI, thus inhibiting oxidation of fatty acids
while synthesis is actively taking place. lnsulin also influences the activity of CPTI, as described subsequently.
Carnitine deficiencies can result in a syndrome called fastinghypoketotic hypoglycemia. The syndrome is associated
with muscle weakness and myoglobinuria after prolonged exercise (as muscle breaks down, releasing myoglobin). The
same symptoms are seen with genetic defects in the carnitine palmitoyl transferase I.
Short and medium chain fatty acids (less than 12 carbons) freelypenetrate the inner mitochondrial membrane in their
nonionized forms and are simply activated by an acyltransferase to acyl-CoAwithin the mitochondrial matrix.

FATIYACTD B-OXTDATTON
Within the mitochondrial matrix, fatty acids are oxidized two carbons at a time from the carboxyl end to form units of
acetyl-CoA. This acetyl-CoA then enters the TCA cycle. The bond between the a and the p carbons is cleaved hence
the name p-oxidation. The general scheme of p-oxidation is shown in figure 2.10.

Congenital defects in the medium chain acyl-CoADehydrogenase (UCAO) lead to the accumulation of medium
chain fatfy acids in the mitochondria. Because medium chain fatty acids are an important source of acetyl-CoA that
supplies the energy required for gluconeogenesis and ketone body synthesis (as we will later learn), this disorder is also
associated with fasting hypoketotic hypoglycemia. Symptoms can include vomiting, coma, and death due to insufficient energ-y delivery to the brain.

.to
ta
(l

E
x

o
I

fo
U

*.
c,

tI'

CYTOSOL

Mg2*

ATP

Fatty

Electron
Transport Chain

Electron
Transport Chain

AMP + PPi

o\/o
ll --)-z-+

nMoH

MATRIX

Add CoA*SH

ll

nYS-coA -

Acyl-CoA

A2-trans-Enoyl-CoA L-(+)-3-Hydroryacyl{oA 3-Ketoacyl-CoA

*Au-coe
Acyl-CoA

Figure 2.10. B-Oxidation of FattyAcids.

36

Lipid Metabolism

As you mryht imagine, the oxidation of an even chain fatty acid of n carbons yields n/2 acetyl-CoA molecules. The
oxidation of an odd chain fatty acid of n carbons yields (n3) /Z aceryl-CoA molecules plus one molecule ofpropionylCoA. This is because the final3-carbon chain cannot be further metabolized through B-oxidation.

n/2acetylCo-A
("4)/Z acetyl Co-A+ propionyl-CoA

Even Chain FatryAcid (n carbons)


Odd Chain FattyAcid (n carbons)

The metabolism of proionyl-CoA is shown in figure 2.11. This is a pathway with several key points to remember.
This pathway is one of two pathways in human metabolism that requires vitamin 812 (the other is the conversion of
the amino acid methionine to the amino acid cysteine, which we will cover in chapter 3). Propionyl-CoA is converted
to D-Methylmalonyl-CoA by Propionyl-CoA Carboxylase, an enzyme that requires biotin as a cofactor. D-Methylmalonyl-CoA is converted to its enantiomer, L-Methylmalonyl-CoA, by Methylmalonyl-CoA Racemase. Then LMethylmalonyl-CoA is converted to Succinyl-CoA by Methylmalonyl-CoA Isomerase, the enzyme which requires
vitarnin Bl2. Succinyl-CoA then enters the TCA.
Because propionate can yield Succinyl-CoA, it is eventually metabolized to oxaloacetate in the TCA. Because oxaloacetate is an intermediate in gluconeogenesis, propionate can be converted to glucose! This is the only means by which

fatty acid oxidation can be glucogenic. Otherwise, the carbon atoms in acetyl-CoA produced by all other fatty acid
synthesis cannot form glucose and can onlybe converted into ketone bodies.

Inactivating mutations in propionyl-CoA carboxylase result in propionic acidemiq while mutations in methylmalonyl-CoA isomerase (methylmalonyl-CoA mutase) result in methylmalonic acidemia. Both of these conditions are associated with hyperammonemia, which can lead to encephalopathy and severe mental disorders. Hyperammonemia is discussed in chapter 3.

EnergyAccounting: Each two carbon fragment of an even chain fatty acid yields one NADH and one FADH2 in the
process ofbeing converted to acetyl-CoA. This is the equivalent of a net 5 AIP. Additionally, each acetyl-CoAproduces

CoA

rc
re
-sH

HscJooh
Propionate

ATp

AMp * ppi

CO2 +

grg

toA-Ar-coA

HrC-Ar-CoA
Propionyl-CoA

oo

exa

ATP

AMP + PPi

D-Methylmalonyl-CoA

ctTRtc
ACID
CYCLE

Succinyl-Col\

L-Methyl-

malonyloA
Figure 2.11. Propionic Acid is Metabolized to Succinyl-CoA, an intermediate of the citric acid cycle,
by a series of enzymes that require biotin and vitamin 812 as cofactors.

Ketone Bodies 37
NADH, 1 FADH2,

AIP

(or GTP) through the TCA, yielding an additional 12 AIP. Hence, each two carbon
fragment of a fatty acid yields 17 AIP. Fatty acid metabolism requires activation at the beginning, which expends two
3

and one

AIPs. Hence:
Number ofAIP Produced = l7*(n/2)

-2

n = number of carbons

for even chain fatty acids

Oxidation ofUnsaturated FattyAcids: Unsaturated fatty acids are metabolized just like saturated fatty acids until the
first unsaturated carbon comes to the p-position. At this point, the cis double bond is isomerized to a trans double bond
by A3-cis + A2-trans-enoyl-CoA isomerase. As you imagine, the resulting substrate now looks exactly like a substrate
for A2-trans-enoyl-CoAhydratase and continues through the normal p oxidation pathway for saturated fatty acids.
Peroxisomal FattyAcid Metabolism: Very long chain fatty acids (greater than 20 carbons) cannot be oxidized directly
in the mitochondrion. Very long chain fatry acids (WCfas) are oxidized first in peroxisomes to octanoyl-CoA (an
eight carbon fatty acid), which can then be metabolized in the mitochondrion. WCFAs enter the peroxisome through
a transporter known as AIDP. Once inside the peroxisome, VLCFAs are metabolizedby p-oxidation by an FAD dependent enzyme called acyl-coA oxidase that leads to the formation of aceryl-CoA andH2O2. Notice the distinction
here between mitochondrial fatty acid oxidation that requires FAD and NAD+ us. peroxisomal fatty acid oxidation, which
only requires FAD. Octanoyl-CoA then exits the peroxisome through AIDP to be further metabolised in mitochondria.
Inactivating mutations in ALDP lead to a disorder known as X-Iinked Adrenoleukodystrophy, in which VLCFAs
accumulate in various tissues, leading to progressive brain damage, adrenal gland dysfunctiory and eventually death.
Inactivating mutations in genes involved in the formation of peroxisomes, knovrn as peroxins (nE,Xt, PE){L,
PEX3, PEXS, PEX6, and others), result in Zellweger Syndrome (cerebrohepatorenal syndrome), which is characterized by liver failure, mental retardation, and seizures.

KETONE BODIES
While fatty acid oxidation yields large quantities of ATP that can be used for gluconeogenesis in the liver, another
important metabolic end product of fauy acid oxidation are ketone bodies. Because gluconeogenic substrates, such
as amino acids, are depleted over time, ketone bodies are another energy source that can be shuttled into the blood
stream by the liver for use by other tissues, including the brain. The major utilizable ketone bodies are acetoacetate and

p-hydroxybutyrate. Additionally, acetoacetate may spontaneously decompose to acetone, releasing carbon dioxide.
Acetoacetate can be converted to acetyl-CoA, while p-hydroxybutyrate is first converted to acetoacetate and then metabolized to acetyl-CoA. Hence, these provide important alternative energy sources in the absence of glucose.
As the serum fatty acid concentration increases, the fatty acid metabolism in the liver exceeds the capacity of the TCA
and electron transport chain. Thus, acetyl-CoA is diverted into ketone body formation by the pathway shown in figure
2.LZ.IlIl0,dG-CoA Synthase is the rate limiting enzyme in the process of ketone body formation. The balance between acetoacetate and p-hydroxybutyrate is determined by the NADH:NAD+ ratio within the tissues, especially the
liver, just as is the balance between pyruvate and lactate. High NADH levels result in the formation of p-hydrorybutyrate,
while low NADH levels result in the formation of acetoacetate. Both are usable by extrahepatic tissues. Acetoacetate
and p-hydrorybutyrate are interconverted within the mitochondrial matrix by p-hydroxybutyrate dehydrogenase.
The liver releases ketone bodies (acetoacetate and p-hydroxybutyrate) into the circulation. Extrahepatic tissues possess

CoA Transferase (thiophorase), which converts acetoacetate to acetoacetyl-CoA, which can then be transformed to
acetyl-CoA by thiolase and used to generate NADH and FADH2 in the TCA. This process is shown in figure 2.13.
The liver does not possess thiophorase and thus cannot metabolize ketone bodies because it cannot make the activated
acetoacetyl-CoA from acetoacetate. The acetoacetyl-CoA generated in the liver to produce ketone bodies is made by
thiolase from acetyl-CoA, which is primarily generated from fatty acid oxidation.
l

38 Lipid Metabolism

oHo

/I
NADH

x.cAs-coA
Acetyl-GoA

ffi

I
CoA-SH

H3cuoo
NAD*

B-Hydrorybutyrate

oo
HscAAr'coA
Acetoacetyl-CoA

Figure 2.12. Ketone Bodies are generated from Acetyl-CoA in the liver as alternative sources of energy when glucose stores are low.

FAITYACID SYNTHESIS

L',ER

While fatty acid oxidation occurs within the


mitochondrial matri:r, fatty acid synthesis (1ipogenesis) occurs within the cytosol. This
physical separation offatty acid oxidation and
synthesis allows for the independent control
ofboth processes according to the needs ofthe
tissue. Acetyl-CoA serves as the starting material andpalmitic acid is the finalproduct of
cytosolic fatty acid synthesis.
l

I
I
a

'I

The Citrate Shuttle: Remember that acetylCoA generated by pyruvate dehydrogenase is


trapped within the mitochondrial matrix and is
not permeable across the inner mitochondrial

Acetyt-coA
I

re

t-

Acetoacetate

'l

Acetoacetyl-CoA

-tr

.l
p-hydroxybutyrate

,[fi::r^

Acetoacetate

1l

-------+ B-hydroxybutyrate

Figure 2.13. Ketone Body Cycling.

membrane. Hence, another shuttle, the citrate shuttle, is necessary to transfer acetyl-CoA equivalents across the inner
mitochondrial membrane. Here, aceryl-CoA and oxaloacetate are transformed into citrate by citrate synthase, and citrate diffuses into the cytosol. Citrate lyase then catalyzes the formation of acetyl-CoA and oxaloacetate in the cytosol,
expending a molecule of AIP. This acetyl-CoA is then used for fatty acid synthesis, first being transformed into malo-

nyl-CoA by acetyl-CoA carboxylase in an ATP dependent manner, before being polymerizedby frtty acid synthase.
AcetyI-CoA Carboxylase is the rate limiting step of fatty acid synthesis and is thus the most regulated step of the
Process. This process is shown in figure 2.l4.Itthe presence of insulin, acetyl-CoA carboxylase is dephosphorylated
and thus activated leading to increased fatty acid synthesis. Glucagon inhibits fatty acid synthesis by inducing phosphorylation of acetyl-CoA carboxylase through PKA (protein kinase A).

FattyAcid Synthesis 39
Fatly acid synthase is a multienzyme complex that transforms two carbon building
blocks into the 16 carbon
saturated fatty acid palmitate.
Fatty acid synthesis is in many
ways an imperfect mirror image of p-oxidation. While the
enzymes that catalyze the two
pathways are completely different, the biochemical reactions are very similar - the
long chain ofpalmitate is built
MArRlx
acetVl*oA;(r-CoA
up in units of two carbons just
as fatty acids are oxidized by
breaking off units of two carbons. Of note, the first two
carbons in palmitate are de-o
L-Matate NAd* Ndox 6x"ur"a"t" Hio cLAsH Ho"
rived from a molecule of acetyl-CoA, while all subsequent
Figure 2.l4.The Citrate Shuttle mediates transport ofAcetyl-CoA from
two-carbon fragments come
the mitochondrial matrix to the cytosol for fatty acid synthesis.
from malonyl-CoA, with decarboxylation of malonylCoAyielding the energetic driving force that propels fatty acid synthesis.

.,Hry"i8-(rw."kK,

Of important note, while p-oxidation produces NADH from NAD+, f^W acid synthesis uses NADPH and yields
NADP+. One of the major uses of NADPH, as discussed in the carbohydrate metabolism review sheet, is fatty acid
synthesis. Without a functional pentose phosphate pathway (Ultn Shunt), fatty acid synthesis is impaired. Fatty acid
synthesis primarily occurs in the liver, while fat storage primarily occurs in adipose tissue. As we will learn in the section
on lipid transport, the primary function of VLDL is the transport of lipids produced in the liver to the rest of the body.
I present the pathway of fatty acid synthesis in figure 2.15, not because you are expected to memorize the steps of this
pathway, but because it is worthwhile to familiarize yourself with the process. What you should k ro*, however, is that
the principal product of cytosolic fatty acid synthesis is palmitic acid (the 16 carbon saturated fatty acid). Notice here
once again that this is a cyclical process, just like p-oxidation. The whole process is catalyzed by a single multi-domain
enzyme, fatty acid synthase.

FaftyAcidElongation: The bodyhas a need to produce fattyacids that are longer than 16 carbons, of course.Additionally, it has a need to elongate preexisting fatf acids. Elongation is one important pathway by which essential dietary
fatty acids are transformed into other physiologically important fatty acids. This elongation takes place in the smooth
endoplasmic reticulum. The microsomal fatty acid elongase system elongates saturated and unsaturated fatty acids that
are at least l0 carbons long in two carbon segments using malonyl-CoA and NADPH in a manner similar to that used
by fatty acid synthase. This system produces the very long chain fatty acids (C22 and C24) that are necessary for myelin
formation and other processes.
Synthesis of Monounsaturated and Polyunsaturated FattyAcids: The liver is capable of synthesizing several nonessential monounsaturated and polyunsaturated faffy acids. The liver uses desaturases, enzymes that utilize molecular
02 and NADH to desaturate a preformed saturated fatty acid. These enzymes are also found within the endoplasmic

40

Lipid Metabolism

aceur$$rlr.""o o,i--,[r:"i
malonYloA
t Co, irt

$t",-t*

*r"",-r)a-,
Ioo

__--=

)-cvs*sn

D-cys-sx

ftr"r, r)ft.r,

j=G

COz*/

}_cys*sH

fttrn-s\r,^\..cH3

t6

-Y
}-cye-sx

$rrn-sy-^\,,cHg
o
Figure 2.15. FattyAcid Synthesis

Insulin Signaling in Lipid Metabolism 4l


reticulum microsomal system. Using the desaturase and elongase system in conjunction, the liver can synthesize
of monounsaturated and polyunsaturated fatty acids.

variety

Regulation of FattyAcid Synthesis: The majority of regulation occurs at the step of aceryl-CoA Carboxylase. Phosphorylation of this enzyme leads to its inactivation, while dephosphorylation leads to its activation. As expected glucagon activates PKA and other kinases such as AMPK, which in turn phosphorylate and inactivate this enzyme. Conversely,
insulin activates various protein phosphatases that dephosphorylate this en4rme and lead to its activation. Thus, insulin
glucagon inhibits lipogenesis, while insulin promotes lipogenesis.

INSULIN SIGNALING IN LIPID METABOLISM


lnsulin stimulates fatfy acid synthesis (lipogenesis) by
a. Increasing acetyl-CoA carboxylase activityby causing its dephosphorylation
b. Increasing the transport of glucose into adipose tissue and thus increasing the availability of sub
strates for lipogenesis
c. Increasing pyruvate dehydrogenase activity in adipose tissue by increasing the activity of PDH
phosphatase and thus increasing the levels of dephosphorylated active PDH. This leads to the
increased production of aceryl-CoA, the major substrate for lipogenesis.
lnsulin inhibits fatf acid catabolism (lipolysis) by
a. Decreasing hormone sensitive lipase activity. HSL is an enzyme in adipose tissue that releases free fatfy
acids from triglycerides, allowing them to diffuse to the liver for metabolism.
b. Regulating in the liver the activity of carnitine palmitoyltransferase I (CPTI), a key enzyme that allows
fatty acids to enter the mitochondrion.
i. Indirectly by increasing fatty acid synthesis and thus the levels of malonyl-CoA, an important al
losteric inhibitor of CPTI
ii. Directly through poorly understood mechanisms
Glucagon and Epinephrine largelyhave the opposite effects in all of the above pathways.

Diabetic Ketoacidosis (Ofe) is a condition associated an absolute lack of insulin, which results both in uncontrolled
gluconeogenesis and in increased lipolysis and release of fatfy acids from adipose tissue and increased fatty oxidation in
the liver. This uncontrolled fatty acid metabolism leads to the accumulation of ketone bodies in the blood. Because these
are acids, an acidosis develops that can be fatal if not treated immediately by insulin administration. Thus, while ketone
bodyproduction normally occurs in hypoglycemic states as a means to provide other sources of fuel to the body's tissues,
in DKA, ketone bodies are pathologically produced in the setting of elevated glucose levels because of the absence of
insulin mediated suppression of fatty acid metabolism. Ketoacidosis can be diagnosed by examining urine ketone levels.
Acetone and Acetoacetate are detected by urine dipstick. However, p-hydroxybutyrate is not detected by urine dipstick!
This is important because, as you administer insulin, the patient begins to recover and fewer ketone bodies are produced.
This means that less fatty acid oxidation is occurring and thus less NADH accumulates in the liver. This leads to the
reoxidation of remaining p-hydroxybutyrate back to acetoacetate. Thus, in the early stages of recovery, blood and urine

p-hydrorybutyrate levels drop and acetoacetate levels stay steady or rise. Thus, patients may paradoxically appear to be
doing worse if you follow urine dipstick only! Therefore, blood ketones should be monitored simultaneously.
Comparing and Contrasting FattyAcid Synthesis and Oxidation: Fatty acid sfnthesis and oxidation are compared and
contrasted in table

2.l,inwhich the key

METAB OLISM

OF

aspects

ofboth processes are also summarized.

GLYCERO LIPID

Metabolism ofGlycerolipids: Once fattyacids

AI\ID SPHING OLIPTD

are synthesized or absorbed in the intestine,

theymustbe transformed into

Triacylglycerol (three fatty acids attached to


glycerol)

Full Oxidation: 9 kcal/g of fat or 131 ATP from


one molecule of palmityl CoA (16 carbons)
Ketone Bodies: generated by the liver to be
converted to ATP in other tissues
Glycerol is reused to produce glucose through

,ll

Table 2.1. Comparing and Contrasting Fatty Acid Synthesis and Oxidation.
the various simple lipids. The acylglycerols (fats and oils) require three fatty acids and

glycerol backbone. The glycerol


backbone is derived from glycerol-3-phosphate, which is formed from dihydroxyacetone phosphate (DFIAP), a glycolysis intermediate, by glycerol-3-phosphate dehydrogenase. Fatty acids are activated by acyl-CoA synthetase to acyl-CoAs.
a

Two acyl-CoAs can then combine with the glycerol-3-phosphate to form a phosphatidate. This is shown in figure 2.16.
Phosphatidic acid is the intermediate that leads to the synthesis of triacylglycerols (triglycerides), the ultimate storage
form of fatty acids in adipose tissue. This is achieved by phosphatidate phosphohydrolase and diacylglycerol acyltransferase. Phosphatidic acid also serves as the intermediate for the synthesis ofthe phospholipids (glycerophospholipids).
Choline and ethanolamine are activated to CDP-choline and CDP-ethanolamine and these are conjugated to phosphatidic acid to yield the
o
respective phospholipids.
il
This is a common theme
CoAS-C-R1
o
that we have encountered
I
R2-C-SGoA
before in glycogen synthe-

sis

activation through
nucleotidation. Phosphatidylserine is formed from

H2c-oH
-r\I-ril-l NADH

N.AD*

grg-gg

:f,
:S--.4-HO-CH
:S---+HO-CH
O
O:0
O
9tlrll-lll
H2C-O-P-OH
H2C-O-P-OH

fl

H2c-o-b-Rl

R-=-C-O-CH
O
R2-C-O-CH
I
H2C-O-P-OH

phosphatidylethanolamine
by a replacement reaction
Phosphatidic Acid
Dihydroxyacetone Phosphate Glycerol-3fhosphate
in which free serine replaces
->
(oHAP)
(G3P)
ethanolamine. Phospholipids are degraded by phos- Figure 2.16. Synthesis of Phosphatidic Acid, the Backbone of Glycerolipids.
pholipases, which catalyze
the hydrolysis of the fatty
acid residue from the glycerol backbone. There are a number of phospholipases involved in cellular signaling cascades
and others that are involved in digestion.

ao

lo

lo

Metabolism ofSphingolipids: Ceramide is synthesizedwithin the endoplasmic reticulum. Sphingomyelins are formed
when the choline from aphosphatidylcholine is transferred to ceramide,leavingbehind a diacylglycerol. Glycolipids are
formed as UDP-glucose and UDP-galactose are added to ceramide, releasing UDP in the process. Gangliosides are just
complex glycolipids that also contain sialic acids such as neuraminidic acid.

--i!-

Lysosomal Storage Disorders - The Sphingolipidoses 43

LYS OS OI\{AL STORAGE DIS

ORDERS

THE SPHTNGOLIPIDOSES
Sphingolipids are largely degraded within lysosomes by sphingolipases and ceramidases, while glycolipid side chains
are first specifically degraded by a variety of saccharidases, including p-galactosidases and hexosaminidases. Avariety
of sphingolipidoses result from the improper targeting of lysosomal enzymes involved in the degradation of sphingolipids. A11 of these disorders are characteizedby the accumulation of complex lipids containing ceramide within the
cells (particularly neurons), Ieading to cell death. Many of these lysosomal storage disorders are particularly prevalent in
AshkenaziJews. Lipids are constantlybeing degraded and resynthesized in the body. This process is essential to ensure
that oxidized and otherwise chemically altered lipids are removed from membranes and replaced with intact lipids. The
sphingolipidoses and sulfatidoses are disorders of lipid degradation and are all recessive lysosomal storage disorders.
Essential lysosomal enzlrrnes involved in the degradation of particular lipids are mutated and individuals homozygous
for these inactivating mutations accumulate those lipids within the lysosomes ofparticular organs of the body, resulting
in the pathology of the disease. As these lipids accumulate, the cells in which they accumulate become dysfunctional.
The sphingolipidoses are summarized in table2.2.
Tay-Sachs Disease: Hexosaminadase A, a lysosomal enz)rrne involved in the degradation of Ganglioside GM2, is
mutated in this disease. GM2 ganglioside is normally present in the myelin sheaths of neurons, in the sarcolemma of
cardiomyocytes, and in the membranes of hepatocytes. The myelin sheaths of neurons, formed by cells called oligodendroglia in the brairy are constantly being degraded and reformed. Without hexosaminadase A, damaged GM2 ganglioside accumulates in oligodendroglia, resulting in their death and eventually demyelination of neurons, resulting in
their degeneration. Symptoms of the disease develop by 6 months of age and are caused by this neurodegeneration. A
normally developing infant -6 months begins to deteriorate and lose both motor and intellectual capacity. The cherryred spot on the macula (part of the retina) is a classic feature and is caused by neurodegeneration within the macula.

Gaucher Disease: Glucocerebroside accumulates in phagocytic cells throughout the body (spleen, liver, lungs, bone
marrow) due to deficiency of the lysosomal enzyme glucocerebrosidase. Glucocerebroside is a component of the cell
membranes of erythrocytes and leukocytes. All of these cells have a finite life span and are eventually phagocytosed by
macrophages of the reticuloendothelial system, the bodyt housekeeping system involving the liver, spleen, and bone
marrow. These macrophages normally degrade all of the components of the red and white blood cells, but without
glucocerebrosidase, glucocerebroside accumulates within the lysosomes of these macrophages, leading to macrophage
dysfunction. These abnormal lipid-filled macrophages resemble crumpled tissue paper histologically, leading to the
pathognemonic catch phrase for the disease, tissue paper macrophages. These macrophages accumulate in bone marrow, spleen, and liver. Patients have anemia from bone marrow dysfunctiory hepatosplenomegaly, and lung and kidney
dysfunction.
Niemann-PickDisease: Sphingomyelin is an essential membrane lipid in the myelin sheaths of neurons and also within the plasma membranes of erythrocytes. Sphingomyelinase deficiency results in the accumulation of sphingomyelin
in oligodendroglia within the brain and macrophages of the reticuloendothelial system. From this, you could expect
that the symptoms of the disease may be a combination of those observed in Tay Sachs Disease and Gaucher Disease.
lndeed, this is the case, with infants developing neurological deficits similar to those present in Tay Sachs diseas eby 6
months of age and later developing hepatosplenomegaly. As with Tay Sachs disease, Niemann Pick disease is associated
with the cherryred spot on the macula.
Krabbe Disease: Deficiency ofgalactocerebrosidase leads to the accumulation ofgalactosylceramide, an essential component of myelin, within lysosomes in oligodendroglia, resulting in neurodegeneration in a mechanism similar to that
observed in Tay Sachs disease. A feature of this disease is that accumulating galactosylceramide also induces macro-

Lipid Metabolism

Table 2.2. The Sphingolipidoses


phages of the brain, called microglia, to form multinucleated giant cells, called globoid cells. This is the histological

pathognemonic of t'his disease. Early symptoms of the disease result from neurodegeneration with developmental
delay, hypotonia, microcephaly and absent reflexes. Eventually seizures develop, with death ensuing.

Fabry Disease: cr-galactosidase A deficiency results in the accumulation of ceramide trihexoside primarily within
endothelial cells, leading to compromised blood flow and the formation of abnormal capillaries called angiokeratomas.
Compromised blood flowwithin the capillarybeds of the kidneylead to renal failure, while angiokeratomas form in
the skin and corneas, leading to corneal clouding.
Some of these diseases can be treated with enzyme replacement therapy, as listed in table 2.2. Because cells are constantly endocytosing material from the extracellular space, it is possible to simply intravenously infuse a patient with
functional enzyme. Because these lysosomal enzymes are normally functional only at the acidic pH of the lysosome,

they are harmless in the blood and extracellular fluid. Once they are endocytose4 th"y end up in lysosomes of the cell,
where they can be activated by the acidic pH and perform their degradative function.
There are two other major lysosomal storage disorders that are not defects in sphingolipid metabolism but rather defects in glycosaminoglyca, metabolism, called mucopolysaccharidoses. These are Hurler Syndrome and Hunter Syndrome. Hurler Syndrome results from a deficiency in a-L-iduronidase. The disease is associated with corneal clouding
and mental retardation. Hunter Syndrome results from a deficiency in iduronate sulfatase and results in a milder form
of disease associatedwith some mental retardationbut no corneal clouding. Hunter Syndrome is X-linked recessive.

Eicosanoids 45

EICOSA\IOtDS
Eicosanoids are important signaling molecules derived from fatty acids. They act as autacoids, locally acting signaling
mediators that generally act within the cell in which they are formed or rarely on adjacent cells. They are generally short

lived. Many important drugs, including acetaminophen (Tylenol), aspirin, and non-steroidal anti-inflarnmatory
drugs (USefOS) work by modulating enzymes involved in the production of eicosanoids. Eicosanoids found in the
human body are derived primarily from eicosatetraenoic acid (arachidonic acid), but also eicosapentaenoic acid (Efe)
and rarely eicosatrienoic acid (usually during states of deficiency in the essential fatty acids). Important autacoids and
their functions are presented in table 2.3, andtheir synthesis is presented in figure 2.17.

Arachidonic acid is found esterified to the sn2 position of phospholipids in the plasma membranes. Phospholipase
A2 frees arachidonic acid by cleaving the ester linkage. This is the rate limiting step for the synthesis of all eicosaniods! Arachidonic acid is then rapidly converted to one of a number of eicosanoids depending on the enzyme expression profile of the cell. The activity ofphospholipase A2 is upregulated by a number of inflammatory mediators, including TNF-a, IFN-1, and others, while glucocorticoids, as we will learry are potent inhbitors of Phospholipase A2 activity.
PROSTANOIDS

PG&
Linoleic Acid
tdiet)

PROSTAITIqDS

TXA{

PGE2

PGF2

PGEl
PGFl

PGl2 (prostacyclin)
TXAa

LtPOX|NS

LX&

/-.t.#coox

LXBa

LXCI

EicosalrienoicnciA-

LEUKOTRIENES
LTA3
LTC3
LTD3

{Eicosatctraenoic Acid)

LEUKOTRIENES
LTA4
LTBa

LXDa

LXEl

LTCa

LTDI

LTEl
PROSTANODS

o-Linolenic Acid
{diet)

re

PGD3
PGE3
PGF3
TXA3

;ffiLm-

(diet)

RESOLVINS

re=

RVEl

LEUKOTRIENES
Eicoeapentaenoic Acid

cl-Linolenic Acld

LTA5

RVEz

PRorEcrtNs

Docosahexaenoic Acid

LTBs
LTC5

Figure 2.l7.The Eicosanoids and their Parent FattyAcids.


Prostanoids (prostaglandins, prostacyclins, and thromboxanes) are formed by the cyclooxygenases (COXf and
COX2). COX1 is expressed in most tissues. It is described as a "housekeeping" enzyme, whose products regulate normal cellular processes such as gastric cytoprotectior5 vascular homeostasis, platelet aggregation, and kidney function,
and COXI is stimulated by various hormones and growth factors. COX2 is usually undetectable in most tissues; its
expression is increased during states of inflammation, or experimentally in response to mitogenic stimuli. As an example, growth factors, phorbol esters, and interleukin-l stimulate the expression of COX2 in fibroblasts, while endotoxin
serves the same function in monocytes/macrophages. COX2 is constitutively expressed in the brain, kidney, bone,
and probably in the female reproductive system. Prostaglandins are largelyvasodilators and bronchoconstricors,
generally pro-inflammatory by initiating various cytokine cascades, inflammatory mediators, enchanced p"i.r, and fever. Prostaglandin E2 (PGE}) is an important housekeeping prostaglanding involved in vasodilation, maintaining

46

Lipid Metabolism

mast cells, macrophages,


muscle, brain, kidney

vascularsmooth gastroprotection
diuresis
painlhyperalgesia
vasodilator

immunomodulator
fever

Table 2.3. The Eicosanoids


blood flow to the gastric mucosa (gastric protection) and renal cortex. Importantly, it is also a potent inflammatory
mediator, leading to hyperalgesia (heightened sensation of pain) and fever. A true double-edged sword! Prostacyclins (Prostaglandin 12 -PGl2) have the important and distinct effect ofbeing anti-thrombotic. Thromboxanes are
largely vasoconstrictory and prothrombotic.
The lipoxygenases synthesize leukotrienes, Iipoxins, resolvins, and protectins. Leukotrienes are typically proinflammatory mediators. The common precursors to the leukotrienes are the hydroperoryeicosatetraenoic acids
(Hffffs). Leukotriene 84 (LTB4) primarily serves an immunomodulatory function, while the Cysteinyl Leukotrienes (WC+,1:ID4,UfF.4) are important mediators of bronchoconstriction and allergic reactions such as asthma
and anaphylaxis and are their receptor is the target of anti-asthma drugs (montelukast, zafirlukast). Lipoxygenase
inhibitors such as zileuton also work in the treatment of asthma by inhibiting the synthesis of the cysteinyl leukotri
enes. Lipoxins are typically anti-inflammatory mediators. Resolvins are compounds derived from eicosapentaenoic
acid (fne) that act as potent anti-inflammatory molecules, while Protectins are compounds derived from docosahexaenoic acid (DHa) that serve as immunomodulators and are important in normal brain function.
There are many other important lipid autacoids. Epoxins are formed by the epoxygenase pathway and are important
for regulation of blood flow and vascular tone in the kidney and other olgans. Isoprostanes are formed as part of oxidative stress (hypoxia, ischemia-reperfusion injury), during which phospholipids containing arachidonic acid that can be
peroxidized by oxygen free radicals. When this oxidized phospholipid containing arachidonic acid is cleaved by phospholipase A2 and processed in by cyclooxygenases, isoprostanes result. During oxidative stress, levels of isoprostanes

Cholesterol 47
in the blood are elevated. Isoprostanes appear to activate a variety of oxidative stress response pathways. Anandamide
(N-arachidonylethanolamine) is synthesized byphospholipases and is involved in analgesia an{pleasure sensing pathways. It is an endocannabinoid and thus binds to the CBl cannabinoid receptor. Cannabinoids contained in marijuana
(Cannibis sativa) seem to co-opt the normal anandamide signaling pathways.

Essential Difierences Among the Eicosanoid Families: Why does it matter that different fatty acid precursors can
give rise to different classes of eicosanoids? Depending on the number of double bonds, eicosanoids have very different activities. For example, the prothrombotic activity of TXA2 is greater than the antithrombotic activity of PGI2.
Thus, when arachidonic acid is the major cellular precursor for prostaglandin synthesis, there is a tendency towards
a prothrombotic state. On the other hand, the prothrombotic activity of TXA3 is much less than the antithrombotic
activity of PGI3. Thus, when eicosapentaenoic acid (fne) is the major precursor for prostaglandin synthesis, there is a
tendency towards an antithrombotic state. This may be the major mechanism by which fish oils (of which eicosapentaenoic
acid and docosahexaenoic acid are the major constituents) improue cardiouascular health. We are only now beginning to
understand the functional differences between the various classes of eicosanoids. Additionally, eicosapentaenoic acid
(gpe) and docosahexaenoic acid (DHA) gi"" rise to resolvins and protectins respectively through the lipoxygenase
pathway. These are the major co-3 components of fish oils, and any commercial fish oil preparation will predominantly
contain these two fatty acids.
The Dangers of COX2 Selective Inhibition (The VIOXX Story) : The cardiovascular risks of COX2 inhibition seem
largely to result from an ensuing imbalance between PROSTACYCLIN and THROMBOXANE synthesis. COX2 spe-

cifically initiates prostacyclin synthesis in the vascular endothelium. When COX2 is inhibited, prostacyclin production
drops dramatically. However, COX(Z seems less important in thromboxane production. Thus, the balance tips in the favor of thrombosis when COX2 is inhibited leading to a prothrombotic state and vasoconstriction. The prothrombotic
state leads to platelet aggregation and endothelial injury, while the vasoconstriction can lead to hypertension. Both of
these significantly increase the risk of adverse cardiovascular events. On the other hand the primary effect of NSAIDs
(nonselective COX blockers) is to inhibit both COX1 and COX2, thereby impairing the ultimate transformation of
arachidonic acid to prostaglandins, prostacyclin, and thromboxane. Thus, both thromboxanes and prostacyclins are
downregulated simultaneously.

METABOLIC CHAAIGES DURING FASTING


The immediate source of energy during the earliest stages of fasting is blood glucose. These stores are maintained up
to approximately 12-18 hours by hepatic glycogen reserves. After approximately 12-18 hours, hepatic glycogen stores
are depleted. At this point, the liver undertakes gluconeogenesis. Specifically, fatty acid oxidation increases, producing
acetyl-CoA. This serves as the primary source of AIP for gluconeogenesis. Amino acids derived from muscle breakdown serve as the primary substrates for gluconeogenesis, as will be described in the amino acid metabolism review.
Ketogenesis occurs simultaneouslywith gluconeogenesis in the earlydays offasting. Some ofthe acetyl-CoAgenerated
from fatty acid oxidation is metabolized in the TCA to yield AIP for gluconeogenesis, while the rest is used to form
ketone bodies. Over time, as protein reserves are depleted, usuallywithin the first week offastingr ketogenesis overtakes
gluconeogenesis
a

as

the primary fuel generating pathway. However, some glucose is needed for proper brain functior5 so

basal level of gluconeogenesis is also maintained.

CHOLESTEROL
Cholesterol is a multifunctional lipid that not onlyplays an important role in regulating the rigidity of the plasma memas a metabolic precursor to ( I ) bile acids; (2) steroid horrnones, including mineralocorticoids, glucocorticoids, and sex hormones; and (3) vitamin D. Its structure is given in figure 2.18.
brane of all cells but also serves

48

Lipid Metabolism

Cholesterol Biosynthesis: Cholesterol is derived both from the diet and from endogenous synthesis within the liver.
The liver excretes approximately 1.2 g of cholesterol per day into the bile. In additioq the average American diet contains approximately 0.4 g of cholesterol. Thus, the major source of cholesterol in the intestine is the liver!
The general schema of cholesterol biosynthesis is given in figure 2.19. Acetyl-CoA is the maior starting material for
cholesterol biosynthesis. The synthesis of cholesterol can be divided into five phases: (t) the synthesis of mevalonate
from aceryl-CoA; (2) the conversion of mevalonate to dimethylallyl
pyrophosphate (DMAPP) and isopentenyl pyrophosphate (IPP),
(g) the conversion of DMAPP and IPP into squalene (a 30 carbon
chain); (+) the cyclization of squalene to produce a 30 carbon steroid known as lanosterol; and (S) the conversion of lanosterol to
cholesterol (a27 carbonsteroid) through the removal of three carbons. The first four phases occur in the cytosol, while the last step
occurs in the endoplasmic reticulum. Hence, cholesterol synthesis also
relies on the citrate shuttle to produce cytosolic acetyl-CoA.

Figure 2.18. Cholesterol


The first phase of cholesterol synthesis is the most important in
terms of regulation and pharmacological targeting and therefore
worthwhile knowing in detail as presented in figure 2.L9. The first step is the formation of acetoaceryl-CoA from two
acetyl-CoA molecules catalyzed by thiolase. The second step is the formation of 3-hydrory-3-methylglutaryl-CoA
(ffUC-Coe) from acetoacetyl-CoA and acetyl-CoA cat alyzedbyEMG-CoA synthase. The final step is the formation
of mevalonate from HMG-CoA. This step requires NADPH and is catalyzedby HMG-CoAreductase.
Regulation of HMG-CoA Reductase: This is the rate limiting step and the most regulated step of cholesterol syna large degee ofproduct inhibition. Cholesterol and bile acids inhibit HMG-CoA reductase transcription. In the absence of cholesterol and bile salts, sterol regulatory element binding protein (Sngnp) translocates
to the nucleus and activates HMG-CoAreductase transcription. ln the presence of cholesterol and bile salts, SREBP is
prevented from translocating to the nucleus, thus reducing HMG-CoA reductase transcription. Mevalonate can also inhibit the enzyme allosterically. Insulin, glucagon, and other hormones also have important regulatory roles here. lnsulin
and thyroid hormone increase HMG-CoA reductase activity and thus stimulate cholesterol synthesis, while glucagon
and glucocorticoids inhibit HMG-CoA reductase activity and thus inhibit cholesterol synthesis. The powerful cholesterol reducing drugs known as statins are HMG-CoA reductase inhibitors.

thesis. Again, there is

Notice at this point that insulin and glucagon regulate carbolrydrate metabolkm, fatty acid metabolism, and cholesterol metaboIt should now become apparent that these are hormones that regulate metabolism in response to the ouerall nutritional

lism.

state ol the body (Jed w. Jasting) and regulate all the metabolic processes in conjunction. Thus, d.iseases such as iliabetes mellitus are truly pan-metabolic diseases, not just iliseases of carbohyilrate metabolkm!

The other steps of cholesterol biosynthesis are worthwhile knowing in as much detail as I have given in figure z.Lg, although there is much more available in the largerliterature (fascinating chemistry) for those who are interested.

LIPID TRA\ISPORI
Lipids are relatively insoluble in the serum and thus require highly sophisticated transport mechanisms that prevent
their precipitation within blood vessels. The lipoprotein system transports triglycerides and cholesterol throughout
the body for use by the tissues and/or storage. Lipoproteins classically have two components, a hydrophilic shell
containing apolipoproteins, phospholipids, unesterified cholesterol and a hydrophobic core containing triacylglycerol
and cholesterly esters.

Lipid Transport 49

ffi
-7-\-

"oo..-[5$f-o.
3-Hydroxyl-3-methyl-

,o.srf,
Ho,,,#o'

Mevalonate

NADH/H+ NAD+

glutaryl-CoA (HMG-CoA)

isopentenyl pyrophosphate
I

t
geranylgeranyl

prenylation of membrane
associated proteins

ffi
"-

oo
x."AA"'coATr,"Ar-coa
Acetoacetyl-CoA

coi_sn

Acetyl4oA

farnesyl pyrophosphate

ubiquinone
heme

squalene

------> lanosterol ----+

CHOLESTEROL

Figure 2.19. Cholesterol Biosynthesis.


Important Apolipoproteins
AI - activates LCAI
B-48 - catalyzes the formation of chylomicrons
B-100 - catalyzes the formation ofVLDL particles
C-II - activates the enzyme lipoprotein lipase
E - induces lipoprotein uptake by the liver
Chylomicrons: Chylomicrons are the first system that is important is the transport of intestinally absorbed long chain
fatty acids and cholesterol. Medium chain fatty acids are generally soluble and enter portal blood directly to be metabolized at the liver. Cholesterol is absorbed through a channel known as NPCILI in enterocytes. This cholesterol is
then largely esterified to cholesteryl esters by acyl-Coa:cholesterol acyltransferase (ACAI). Long chain fatty acids
are generally absorbed directly through the plasma membrane in their uncharged form. In the intestinal epithelial cells

(enterocytes), these fatty acids are then converted back to triglycerides by diacylglycerol acyltransferase

(OCet).

In the enterocytes, the ApoB gene is transcribed. The mRNA forApoB is then edited byApoB Editing Complex I
(anOnECf ), a protein which converts a glutamate codon to a stop codon. This results in the synthesis ofApo B-48 in
the enterocyte, a protein that is 48o/o of the length of the firll length protein. Within the smooth endoplasmic reticuIum, Apo B-48 is translipidated by microsomal transfer protein (Utn), which basically adds triglycerides and choIesterol to Apo B-48. Thus, the nascent chylomicron is formed. Nascent chylomicrons are secreted into the interstitial
fluid from the basolateral membrane of enterocytes. They are too large to enter the capillaries, so instead they are taken
up by lymphatics and delivered to the blood through the thoracic duct. Once in the blood, the main function of chylomicrons is to deliver triglycerides to tissue. This process is shown in figure 2.19. Atthis point nascent chylomicrons are
inactive. They must be activated by gaining a molecule ofApo CII from circulating HDL particles, which act as reservoirs of this apolipoprotein. Once they gain Apo CII, chylomicrons are activated and can deliver their lipids to tissues.
Muscle and adipose tissue produce a protein called lipoprotein lipase (Lnl). They secrete this protein, which binds to
glycoproteins on endothelial cells. Thus, endothelial cells lining capillaries in muscle and adipose tissue selectivelypossess this protein. LPL hydrolyzes triglycerides in the core of chylomicrons to produce free fatty acids. These free fatty acids then get absorbed directly through the plasma membrane of the local cells. These chylomicrons shrink as their fatty

50

Lipid Metabolism
Dietary

Small

lntestine

Figure 2.20. The Chylomicron Cycle.


HDL once agairy now exchangingApo CII and getting
Apo E, becoming chylomicron remnants. Chylomicron remnants are important in the process of reverse cholesterol
transport, which will be described subsequently. Apo E binds to the LDL receptor or the LDl-receptor Related Protein
(I-np), and these chylomicron remnants are then endocytosed by the liver, thus completing the chylomicron life cycle.
acids are slowly removed. These chylomicrons then interact with

Summary of Chylomicrons
. Least dense of lipoproteins
. Formed in smooth ER of intestinal mucosa and secreted into lymphatics to transport dietary lipids
. Triacylglycerol in the chylomicrons is hydrolyzed byLPL in vasculature of cardiac, skeletal muscle and adipose tissue
. Apo B4B is unique to chylomicrons
. Apo C-II activates LPL and results in fatty acid release to the heart, skeletal muscle, and mammary glands
. Apo E allows clearance of chylomicron remnants by the liver by binding to the LDL receptor on hepatocytes
VLDLT IDL, andLDL: Asyou remember, there are two sources of cholesterol and triglycerides - the diet and the liver!
Hence there are two modes of delivery. As the chylomicron is the delivery system for lipids absorbed through the diet,
the WDL system is the mode of systemic delivery for lipids synthesized by the liver. The pathways for the two systems
are remarkably similar, but different in some very critical ways! The majority of the cholesterol in the body is synthesized in the liver - this far outweighs the amount of cholesterol received through the diet!
In the liver, hepatocytes do notpossess theAPOBECI editingmachinery and thus synthesizeApo B-100, the full length
Apo B protein. This is then translipidated by MTP in the liver to form VLDL. The liver then secretes VLDL particles
i

Lipid Transport 5l
into the blood for peripheral delivery of triglycerides. This may have evolved as a system to ensure sufficient peripheral
delivery of fatty acids when there is insufficient dietary intake. Once in the periphery VLDL must be activated just like
chylomicrons. This process is shown in figure 2.20.VLDL particles get Apo CII from HDL particles, after which they
can be metabolized by peripheral LPL. Eventually, as these particles shrink, about 50% of these particles exchange Apo
CII and get Apo E from HDL, thus becoming VLDL remnants. These VLDL remnants also participate in reverse cholesterol transport and eventually are taken up by the LDL receptor and metabolized by the liver just like chylomicrons.

ofWDL

continues to be metabolized by LPL before exhanging Apo CII for Apo E and becomes intermediate-densrtylipoproteins (tUl). Aportion of these IDL particles are then taken up by the LDL receptor, which
binds the Apo E on ID! and thus are metabolized just like VLDL remnants and chylomicron remnants. The remaining
portion of this IDL is metabolized byhepatic lipase to low-densitylipoproteins (LDL). Importantly,LDLparticles
lose their affinity for Apo E and thus lose it to HDL particles. Thus, LDL particles do not have Apo E. HENCE THEY
DO NOT BIND WELL TO THE LDL RECEPTOR! THIS IS A WRY IMPORThNT POINT - LDL PARTICLES DO
NOT BIND WELL TO THE LDL KECEPTOR! Thus, they cannot be taken up by the liver with any great efficiency.
Weak interactions ofApo 8-100 with the LDL receptor allow its clearance after an extended circulatorylifetime.
The other

Thus,

50%o

LDL

t ) is very rich in cholesterol because most of the fatty acids have been removed by the lipases and

(2) can-

not be efficiently cleared from the blood. Thus, these cholesterol rich particles float in the blood for extended periods
of time, during which time they can be oxidized, glycosylated, or otherwise altered. This modified LDL binds scavenger
receptors on macrophages in the blood vessels and is then internalized. These macrophages are converted to foam cells
as they continue to take up modified LDL particles. These foam cells secrete a variety of cytokines and chemokines that
cause localized inflammation and thus contribute to atherogenesis and atherosclerosis.

VLDL sumrnary
. Made in the liver to transport triglycerides and choles-

LDLSummaly
. Generated from IDL by the action

teryl esters
. More enriched in cholesterol than chylomicrons
. Also metabolized by lipoprotein lipase to produce IDL

. Have a larger proportion of cholesteryl esters in the core


. Retain only apo B 100, which interacts with LDL receptor
on hepatocytes

of hepatic lipase

poorly

HDL and Reverse Cholesterol Transport: HDL serves two major functions: ( 1) to act as an apolipoprotein reservoir,
holding the exchangeable apopipoproteins Apo CII, Apo E, and others, as you have seen in both the chylomicron cycle
and the VLDL cycle; and (Z) to transport excess cholesterol from the periphery back to the liver for excretion. This
second process is referred to as reverse cholesterol transport.

In HDL formation, the first step is the secretion of Apo AI. This apolipoprotein is then translipidated byABCAI, a
protein located in the sinusoids of the liver (hence not in the ER), and thus the nascent HDL particle is generated.
This particle then becomes mature HDL. HDL accepts cholesterol from peripheral tissues, which temporarily reside
on its surface. The Apo AI activates lecithin:cholesterol acyltransferase (LCAT), which esterifies this cholesterol to
cholesteryl esters so that cholesterol does not accumulate on the surface of HDL. This is essentially a flux generating
reaction that allows for more cholesterol to then be added to the surface of the HDL particle, another example of Le
Chatliert Principle at play. Phospholipid Transfer Protein (nlff') transfers phospholipids to HD! which then act
as the source of faffy acids for cholesterol esterification. Thus, the HDL particle accumulates cholesterol primarily in
the form of cholesteryl esters in the core. This is where chylomicron remnants and VLDL remnants come into play.
Remember that these particles are traveling back to the liver for uptake by hepatocytes. Cholesteryl Ester Transfer
Protein (Cmn) transfers these cholesteryl esters from HDL to the remnant particles for deliverybackto the liver.
Two important points about CETP: ( t ) By transferring cholesteryl esters from HDL to remnants, it is reducing HDL
cholesterol (i.e., it is reducing the total amount of blood cholesterol that is in the form of HDL); (Z) BV removing
cholesteryl esters from HDL, it is extending the life of the HDL particle such that it can absorb more cholesterol from

s2

Lipid Metabolism

Figure 2.2l.The VLDL Cycle.


peripheral tissues. Thus, CETP is actually extending the usable life of the HDL particle by allowing for cholesterol to
transported back in particles that will anyway be metabolized.

Once the HDL particle has enlarged to a critical size, it binds to hepatic scavenger receptors and is taken up to be mr
tabolized in the liver, thus completing the cycle of reverse cholesterol transport.

HDL Summary
. Synthesized by the liver
. Approximately

protein
. Its core region is relatively empty when it is secreted by the liver but fills up with cholesteryl esters in the periphery
. Serves as a circulating reservoir for apoproteins
. It moves cholesterol from extrahepatic tissue to the liver by the process of reverse cholesterol transport
50o/o

Other Lipoproteins of Note: Lipoprotein (a) (I-p(a) - "lp little a") and Lipoprotein X appear to have significant
diovascular risk, although their exact functions remain obscure.

car

DISORDERS OF LIPID TRANSPORT

Both hyperapolipoprotenemias and hypolipoproteinemias cause disease in humans, as shown in table 2.4. Many <
these lead to premature atherosclerosis. Atherosclerosis is primarily the result of the accumulation of oxidized choler
terol within the vascular tunica intima. This is usually caused by deranged LDL metabolism, and the LDL:HDL rati

Important Lipid Modulating Drugs 53


LDL in the blood stream
is first modified (oxidized, glycosylated). lhit LDL accumulates in the sub-intima of blood vessels, where it binds to
scavenger receptors on macrophages and is endocytosed. These macrophages become lipid laden foam cells that release
a number ofpro-inflammatory mediators. Inflammation then leads to smooth muscle proliferation and the formation of
an atterosclerotic plaque. These plaques can signfficantly occlude a blood vessel and can also rupture and embolize,
completely occluding smaller distal blood vessels, and leading to acute ischemia and infarction.
is a good clinical predictive parameter of cardiovascular risk. The process begins when excess

Managing and Mitigating Risk: Dietary influences such as increased intake of polyunsaturated fatty acids and monounsaturated fatty acids and reduced intake of saturated fatty acids and trans fatfy acids seem to reduce the LDL:HDL
ratio and result in reduced cardiovascular risk. Lifesryle influences including smoking, obesity, and hypertension seem
to generally increase serum cholesterol levels, while exercise and weight loss may reduce these levels.

IMPORTA\IT LIPID MODULATING DRUGS


Statins (atorvastatin, simvastatir5 rosuvastatirl lovastatir5 pravastatin) are competitive inhibitors of HMG-CoA Reductase, the rate limiting step in cholesterol synthesis. This ultimately reduces the production of cholesterol in the
Iiver. The liver in furn 'tenses" low cholesterol since there is decreased production and a compensatory increase in LDL
receptor trafficking to liver surface resultsr leading to increasedWDL, IDI and LDL uptake and decreased circulating
LDL. Thus, the key point to understand here k that statins increase LDL uptake by the lfuer by decreasing endogenous
hepatic cholesterol synthesis. This is the key mechanism bywhich statins work. Statins also target Klf-2, which maybe
another atheroprotective target.

Table 2.4. Common (and Commonly Tested) Disorders of Lipid Tfansport

s4

Lipid Metabolism

BiIe acid resins (cholestyramine, colesevelam, colestipol) block enterohepatic circulation by binding up bile acids
in the intestine. This increases the liver's metabolism of cholesterol to bile acids, thus reducing the overall cholesterol
levels. They primarily reduce LDL levels but have little to no effect on HDL levels.
Fibrates (gemfibrozil, clofibrate) activate PPARc, causing increased expression of lipoprotein lipase. This in turn increases triacylglycerol removal from circulation. How exactly this reduces cholesterol levels is still uncertairy although
as we have discussed, circulating fats, especially saturated fats, have potent effects in increasing serum cholesterol levels.

Very High Dose Niacin (1SOO - 3000 mg/day) decreases LDL by inhibiting VLDL production and also increases
HDL, possibly by increasing the half-life ofApo AI. This is the only existing drug that increases HDL by any appreciable
amount.

Cholesterol absorption inhibitors (Ezetimibe) blocks cholesterol absorption in intestine through the NPCl-Ll
channel in enterocytes, thus increasing the excretion ofcholesterol in the stool.

CETP inhibitors (Torcetrapib) were tried because CETP deficiency leads to increased HDL, as summarized in table
2.4.However, they actually increased mortality and were abandoned. Theoretically, while they may have increased the
total cholesterol found in HD! they actuallyinhibited the overall process of reverse cholesterol transport!

IMP O RIANT CH
Cholesterol is converted to

LESTERO L DERIVATIVES

number of metabolites, includingbile acids, steroid hormones, and vitamin D.

Bile Acids: Bile acids are emulsifying agents that aid in the absorption of fats in the intestine and are synthesized from
cholesterol through hydrorylation. Importantly, once bile acids are formed from cholesterol, cholesterol cannot be regenerated. The key enzyme in this process is 7c-hydroxylase, the rate limiting step in bile acid formation. This process
relies on NADPH, Vitamin C, and molecular 02. Once formed, bile acids are released inbile to emulsify dietarylipids.
Bile acids are then reabsorbed in the ileum and recycled in a process known as enterohepatic circulation, which reduces
the need for constant hepatic synthesis of bile acids.
Steroids: A hormone is a substance which exerts its effect at a location &stant from its synthesis. Steroid hormones
are by definition lipids and thus readily cross the cell membrane. The main action of steroid hormones is to induce or
repress the expression of specific genes whiclr, in turn, alter cell function. Once steroids cross the cell membrane, they
bind to protein receptors in the cytosol and activate these receptors. Steroid-bound activated receptors then translocate
to the nucleus, where theybind to DNA and alter gene expression.
Steroid hormones are synthesized primarily in the adrenal corte& although the ovaries are important sources of estrogens and progesterone and the testes are important sources of testosterone. The adrenal gland is divided into three
zones, the zona glomerulosa, the zona fasciculata, and the zona reticularis, respectively layered from the surface of the
cortex to the center. The core of the adrenal gland is the medulla, which is involved in catecholamine (epinephrine)
synthesis, which is covered in chapter 3.
The zona glomerulosa is responsible for the synthesis of aldosterone, a mineralocorticoid involved in sodium and water homeostasis that acts primarily on the kidney. The zona fasciculata is responsible for the synthesis of cortisol, a

glucocorticoid responsible for the regulation of stress responses, including increasing blood glucose concentrations
and modulation immune response. The zona reticularis is responsible for synthesizing adrenal androgens. While the
functions of each of these hormones is generally taught in endocrine pathophysiology, the synthesis of these hormones
is schematized in figure 2.22. Nlof the adrenocortical enzymes are part of the P450 cytochrome oxidase family of en-

Important Cholesterol Derivatives 55

cHoLEsrERorK, Pregnenolonem

17-hydroxy
Pregnenolone

PROGESTERONE

-ff

ffil

Deoxycorticosterone

+
Corticosterone

7-hydroxy

Androstenedione

Progesterone

Deoxycortisol

DehydroepiAndrosterone

I
I

T.

TESTOSTERONK

NI

coRTtsoL

+
ESTRADIOL

ffr
-

ALDOSTERONE

Figure 2.22.Enzymes Involved in the Synthesis of Steroid Hormones.


zymes and also have corresponding P450 classification names. Desmolase is considered the rate

limiting enzyme of

steroid synthesis.

Congenital Adrenal Hyperplasia: Enzyme deficiencies can cause an inability to produce certain hormones and accumulation of precursor molecules or shunting to other pathways. The adrenal gland is over-stimulated by pituitaryderived ACTH and thus hlryertrophies due to lack of the normal negative feedback cycle. There are several types:

Lipoid congenital adrenal hyperplasia results from the failure to metabolize cholesterol resulting from desmolase
deficiency. The adrenals fill with cholesterol globules, hence the name lipoid CAH. There is a severe deficiency of all
steroid hormones, and males are severely undervirilized being raised as infertile females in most cases. There is also
1)

marked salt wasting due to the lack of mineralocorticoids.


2) 3p-hydroxysteroid dehydrogenase deficienc, as expected from the above pathway, also results

in severe defi-

ciency of all steroid hormones. The symptoms are essentially identical to those of lipoid CAH.
a) f Zc-hydroxytase deficiency results in an inability to produce the glucocorticoids and sex steroids and shunting
of precursors to the mineralocorticoid pathway, leading to salt retention, which causes hypertension. Agairy males are
severely undervirilized.

+) Zt-hydroxylase deficiency results in

an inability to produce glucocorticoids or mineralocorticoids, leading to salt

wasting. Shunting to the androgen pathway leads to overproduction of adrenal androgens and virilization in females.
5) I f p-hydroxylase deficiency, as with 21-hydroxylase deficiency, causes virilization in females.

Androgens: Testosterone is synthesized in Leydig cells of testes in a process similar to that of adrenal steroid synthesis.,
using the enzymes shown in figure 2.Zl.Dihydroxytestosterone (DHf), a more potent androgen, is produced from
testosterone by So-reductase, an enzyme found in peripheral tissues.
Female SexHormones: Progesterone is produced by the corpus luteum within the ovary and the placenta from pregnenololne as shown in figure 2.22. Estradiol is converted from testosterone by the enzyme aromatase, found in the
granulosa cells of the oyary. Testosterone in turn is produced by the theca cells of the oyary. These reasctions are also
shown infigare2.2l.

56

Lipid Metabolism

Vitamin D: Vitamin D biosynthesis involves four distinct

steps, depicted in figure

2.8. (I)

The liver converts choles-

terol to 7-dehydrocholesterol. (2) Then ultraviolet radiation incident on the skin catalyzes an electrocyclic ring opening
to produce Pre-vitamin D3. This is then isomerized to vitamin D3 (cholecalciferol) at body temperature spontaneously.
(S) Vitamin D3 is converted to 2S-hydroxyvitamin D3 in the liver by 2S-hydrorylase. (+) 2S-hydroxyvitamin D3
is then converted in the kidney by f -c-hydroxylase to lr2S-dihydroxyvitamin D3 (calcitriol), the active form of
vitamin D. Vitamin D2 (ergocalciferol) can be derived from the diet from plant sources and can also be converted to
2S-hydroxyvitamin D2 in the liver and then to l,2S-dihydroxyvitamin D2 in the kidney.

Vitamin D deficiencyleads to rickets in children and osteomalacia in adults. Both ofthese diseases are characterizedby
continued formation of the bony collagen matrix, but insufficient mineralization, ultimately leading to soft bones. Primary causes ofvitamin D deficiency include nutritional deficiency (extremely common in the US - up to 80-90% of the
population currently) and decreased sun exposure. Deficiencies in cortisol, the active form, can result from renal failure.

VITAIVIIN D IN CALCIUM HOMEOSTASIS


Calcium is essential for numerous biological processes, from signal transduction, vesicle trafficking, and apoptosis to
electrical conduction and myocyte contraction. Thus, the proper maintenance of calcium homeostasis is essential. Hypercalcemia and hypocalcemia have serious systemic manifestations, including altered cardiac rhythm and disruption
of neural function.

(nfH) is a hormone that acutely regulates

calcium. When blood levels of calcium fluctuate,


PTH levels will quickly follow suit. The parathyroid gland can quickly modulate its secretion of preformed PTH in
response to calcium levels in the blood, which it senses through the Calcium Sensing Receptor, a G protein coupled receptor (GPCR). PTH itselfbinds to a cell surface GPCR, thus allowing it to signal quickly and thus acutely altering calcium levels. Vitamin D is a hormone that maintains general calcium homeostasis over time. As a steroid-like hormone,
it acts through nuclear hormone receptors (here the Vitamin D receptor, VDR). lhis changes gene transcription, which
is a slow process. Thus, vitamin D makes slow changes to maintain general calcium homeostasis. ln general, peptide
hormones (like nlft, insulin, etc.) are fast acting and have a short duration of action before they are degraded. On the
other hand, steroid hormones (including steroid like Vitamin D) are slow acting and have a long duration of action as
they generally change gene transcription intracellularly.
Parathyroid Hormone

Vitamin D: Vitamin Dt main effect is at the intestine. It specifically upregulates the gene expression of a number of
genes involved in calcium absorption, including (t) the apical calcium pump, (2) calbindln, a protein that transports
calcium within the enterocyte, and (3) a basolateral calcium pump. Thus, it maintains calcium absorption in the gut to
maintain blood calcium levels chronically. At the kidney, vitamin D increases the absorption ofboth calcium and phosphate, both minerals needed for bone formation. At the bone, vitamin D increases osteoclast activity. Osteoclasts break

important and essential part of bone remodeling that is occuring constantly and is important to the
maintenance of bone. Without remodeling, bone is unable to respond to varying stress and grow or shrink in response
to physiological loading. Thus, vitamin D allows for the absorption of essential minerals (calcium and phosphate) for
bone formation while simultaneously stimulating osteoclasts. The net effect is the maintenance of bone in a dynamic
and responsive state that can respond to physiological need for growth. Of course, within the bone, there are numerous
other factors that are also controlling this interplaybetween growth and resorption. Thus, vitamin Dt ultimate purpose
is to maintain calcium levels to allow for all physiological processes that require calcium.
down bone

as an

PTH: PTH is secreted when calcium levels are low, as sensed by the calcium sensing receptor in the parathyroid gland.
PTH acutely acts on all three organs to raise blood calcium levels. At the bone, PTH causes acute resorption. It activates PTH receptors on osteoblasts, causing them to express RANK (receptor activator of NFKB) ligand. This in turn
binds to RANK on osteoclast precursor cells. Activation of RANK leads to osteoclast activation through NFKB path-

--'jil

Important Diseases of LipidMetabolism 57

G
{,2Sdihydroxyvitamin-D3
(cholecalciferol)

<-

KIDNEY

25-hydroxyvitamin-D3

Figure 2.23.Yitamin D Synthesis.


ways. These osteoclasts then resorb bone, increasing calcium and phosphate levels in the blood. At the kidney, PTH
causes calcium resorption as well. However, it causes phosphate excretion. This is because calcium phosphate is a very

insoluble compound. As it is causing bone resorption, which is increasing both calcium and phosphate levels, if phosphate is not quickly excreted then calcium phosphate will precipitate in the bloodvessels, leading to a condition called
calcinosis. Thus, in order to quickly increase blood calcium levels, PTH is bringing out calcium from the bone, but to
prevent it from simplyprecipitating out, it must induce the excretion of the phosphate that comes with it. The effects of
PTH on the intestine are indirect. This is because absorption through the intestine is not a way to quickly increase blood
calcium, especially when the person may not have eaten a meal or even if the person has eaten a meal, it may not be
particularly calcium rich. Thus, teleologically, PTH cannot exert significant acute alterations at the level of the intestine.
lnstead it increases the production of active vitamin D by the kidney. This increased vitamin D then increases over time
the absorption of calcium. This calcium will then restore over time the bone stores that were acutely called into action.

IMPORTA\TT DISEASES OF LIPID METABOLISM


Maior Metabolic Diseases
Atherosclerosis - LDL accumulation in macrophages within blood vessel walls leading to the formation of atherosclerotic plaques that compromise blood flow through (1) constriction of flow; (2) acute thrombotic occlusion; and (3)
distal embolization with occlusiory leading to end organ hypoperfusion, causing myocardial infarctions and strokes.
Diabetic Ketoacidosis - absolute insulin deficiency results in the unabated oxidation of fatty acids in the liver and
production of ketones, leading to acidosis. Concomittant unabated gluconeogenesis results in hyperglycemia. This is in
effect the opposite of the fasting hypoketotic hypoglycemia state.
Disorders oflipid Transport (fable Z.+)

58

Lipid Metabolism

Some Important Enzyme Deficiencies in Carbohydrate Metabolism


Fasting Hypoketotic Hypoglycemia (Carnitine Deficiency, Carnitine Palmitoyl Transferase I Mutations, Medium
Chain Acyl-CoA Dehydrogenase (tUCeO) Deficiency) - inability to produce ketone bodies in hypoglycemic states
PropionicAcidemia - results in hyperammonemia and encephalopathy
Methylmalonic Acidemia - results in hyperammonemia and encephalopathy
X-LinkedAdrenoleukodystrophy - results in progressive brain damage and adrenal gland dysfunction
Zellweger Syndrome - results in liver failure, mental retardatiory and seizures
The Sphingolipidoses (fable z.t)
The Congenital Adrenal Hlperplasias

REVTEWqUESTTONS
Choose the correct answer(s) for the following
questions (each question mayhave multiple correct answers).
Which of the following vitamins is not required for the
conversion of propionate to succinate?
A. Biotin
B. Niacin (Vitamin83)
C. Cobalamin (Vitamin B12)
D. Pantothenate (Vitamin 85)
E. FolicAcid
1.

7.ApoE
8-12. Match each enzyme with its functions.
A. Translipidates ApoB isoforms to form chylo
microns oTVLDL
B. Transfers cholesteryl esters from HDL to remnants
C. Hy dr oly zes triglycerides o n chylomicro ns and VLD L
D. Transfers phospholipids from remnants to HDL
E. Forms cholesteryl esters from free cholesterol in HDL
8. Lipoprotein Lipase

LCAI (PCAT)

2. Site-directed mutagenesis is used to produce a mutation in the gene for apolipoprotein B100 in a mouse.

e.

Because of this mutation:

rO. CETP

A. Hormone-sensitive lipase activity decreases


B. LDL binding to its receptor will be abnormal
C. LCAT activitywillbe reduced
D. Lipoprotein lipase activitywill be reduced
E. Remnant uptake by the liver will be abnormal
3-7. Match each apolipoprotein with its function.
A. Activates lipoprotein lipase
B. Binds to the LDL receptor with high affinity
C. Catalyzes the formation of chylomicrons
D. Catalyzes the formation ofVLDL particles
E. Catalyzes the formation of HDL particles
F. Binds to the LDL receptor with low affinity

TT.

MTP

12. PLTP

13-16. Match each lipoprotein with the apolipoproteins


that are permanently or transiently associated with it.
A.Apo B-48
B.Apo B-10013.

C.Apo E
D.Apo CII
E. Apo

13.

AI

HDL:

3.ApoAI
14.LDL
4.Apo B-48
15.

wDL

5.Apo B-100
16. Chylomicron

6.Apo CII

Potrebbero piacerti anche