You are on page 1of 22

Atherosclerosis

Jordan B. Strom
Peter Libby

VASCULAR BIOLOGY OF ATHEROSCLEROSIS


CH 5 APTER

ATHEROSCLEROSIS RISK FACTORS


Normal Arterial Wall Traditional Risk Factors
Atherosclerotic Arterial Wall Biomarkers of Cardiovascular
Complications of Atherosclerosis Risk Outlook

A therosclerosis is the leading cause of


mortality and morbidity in the developed world.
lumen and therefore most “intimate” with the
blood; the media, which is the middle layer; and
Through its major manifestations of the outer layer, the adventitia. The intimal
cardiovascular disease and stroke, it likely will surface consists of a single layer of endothelial
become the leading global killer by the year
2020. Commonly known as “hardening of the cells that acts as a metaboli-cally active barrier
arteries,” atherosclerosis derives its name from between circulating blood and the vessel wall.
the Greek roots athere-, meaning “gruel,” and The media is the thickest layer of the normal
-skleros, meaning “hardness.” artery. Boundaries of elas-tin, known as the
Recent evidence has demonstrated that ath- internal and external elastic laminae, separate
erosclerosis is a chronic inflammatory condi-tion this middle layer from the intima and adventitia,
and that its pathogenesis involves lipids, respectively. The media consists of smooth
thrombosis, elements of the vascular wall, and
muscle cells and extracel-lular matrix, and
immune cells. The process of atherogenesis
subserves the contractile and elastic functions
can smolder throughout adulthood, punctu-ated
of the vessel. The elastic component, more
by acute cardiovascular events.
prominent in large arter-ies (e.g., the aorta and
This chapter consists of two sections. The
its primary branches), stretches during the high
first part describes the normal arterial wall, the
pressure of systole and then recoils during
pathogenesis of atherosclerotic plaque forma-
diastole, propelling blood forward. The muscular
tion, and pathologic complications that lead to
component, more prominent in smaller arteries
clinical symptoms. The second section relates such as arterioles, constricts or relaxes to alter
findings from population studies to attributes vessel resistance and therefore luminal blood
that lead to this condition, thereby offering op- flow (fl ow ! pressure/resistance; see Chapter
portunities for prevention and treatment. 6). The adventitia contains the nerves, lymphat-
ics, and blood vessels (vasa vasorum) that
VASCULAR BIOLOGY OF nourish the cells of the arterial wall.
ATHEROSCLEROSIS
Normal Arterial Wall Far from an inert conduit, the living arte-rial
wall is a scene of dynamic interchange between
The arterial wall consists of three layers (Fig. its cellular components—most impor-tantly,
5.1): the intima, closest to the arterial endothelial cells, vascular smooth mus-cle cells,
and their surrounding extracellular

113
Chapter 5

Lumen

Endothelial cells Intima


Internal elastic lamina

Smooth muscle cells Media

External elastic lamina


Adventitia

Figure 5.1. Schematic diagram of the arterial wall. The intima, the innermost layer, overlies the
muscular media demarcated by the internal elastic lamina. The external elastic lamina separates
the media from the outer layer, the adventitia.

matrix. An understanding of the dysfunc-tion Endothelial cells also secrete substances


that leads to atherosclerosis requires that modulate contraction of smooth muscle
knowledge of the normal function of these cells in the underlying medial layer. These
components. substances include vasodilators (e.g., NO
and prostacyclin) and vasoconstrictors (e.g.,
endothelin) that alter the resistance of the
Endothelial Cells
vessel and therefore luminal blood flow. In a
In a healthy artery, the endothelium performs normal artery, the predominance of vasodila-
structural, metabolic, and signaling functions tor substances results in net smooth muscle
that maintain homeostasis of the vessel wall. relaxation. Several of the aforementioned
The tightly adjoined endothelial cells form a endothelial products also function within the
barrier that contains blood within the lumen of vessel wall to inhibit proliferation of smooth
the vessel and limits the passage of large muscle cells into the intima, thus enforcing
molecules from the circulation into the sub- their normal residence within the media.
endothelial space. Endothelial cells can also modulate the
As blood traverses the vascular tree, it en- immune response. In the absence of patho-
counters antithrombotic molecules produced by logic stimulation, healthy arterial endothelial
the normal endothelium that prevent it from cells resist leukocyte adhesion and thereby
clotting. Some of these molecules reside on the oppose local infl ammation. However, endo-
endothelial surface (e.g., heparan sulfate, thelial cells in postcapillary venules respond
thrombomodulin, and plasminogen activators; to local injury or infection by secreting
see Chapter 7), while other antithrombotic chemokines—chemicals that attract white
products of the endothelium enter the circula- blood cells to the area. Such stimulation also
tion (e.g., prostacyclin and nitric oxide [NO]; see causes endothelial cells to produce cell
Chapter 6). Although a net anticoagulant state surface adhesion molecules, which anchor
normally prevails, the endothelium can also mononuclear cells to the endothelium and
produce prothrombotic molecules when facilitate their migration to the site of injury.
subjected to various stressors. These effects may be mediated in part via

114
Atherosclerosis

NORMAL ACTIVATED

ENDOTHELIAL
CELLS
• Impermeable to large molecules Permeability
• Anti-inflammatory Inflammatory cytokines
• Resist leukocyte adhesion Leukocyte adhesion molecules
• Promote vasodilation Vasodilatory molecules
• Resist thrombosis Antithrombotic molecules

SMOOTH MUSCLE
CELLS

• Normal contractile function Inflammatory cytokines


• Maintain extracellular matrix Extracellular matrix synthesis
• Contained in medial layer Migration and proliferation
into subintima
Figure 5.2. Endothelial and smooth muscle cell activation by inflammation. Normal
endothelial and smooth muscle cells maintain the integrity and elasticity of the normal arterial
wall while limiting immune cell infiltration. Inflammatory activation of these vascular cells corrupts
their normal functions and favors proatherogenic mechanisms that drive plaque development.

Kruppel-like factor 2 (KLF2), a gene regula- interleukin-6 (IL-6) and tumor necrosis
tor in endothelial cells. As described later, factor-" (TNF-"), which promote leukocyte
under the adverse infl uences present during prolif-eration and induce endothelial
atherogenesis, endothelial cells similarly re- expression of leukocyte adhesion molecules
cruit leukocytes to the vessel wall. (LAM). These synthetic functions become
Thus, the normal endothelium provides a more prominent at sites of atherosclerotic
protective, nonthrombogenic surface with plaque and may con-tribute to their
homeostatic vasodilatory and anti- pathogenesis.
inflammatory properties (Fig. 5.2).
Extracellular Matrix
Vascular Smooth Muscle Cells
In healthy arteries, fibrillar collagen, proteo-
Smooth muscle cells within the vessel wall glycans, and elastin make up most of the extra-
have both contractile and synthetic capabili- cellular matrix in the medial layer. Interstitial
ties. Various vasoactive substances modulate collagen fibrils, constructed from intertwining
the contractile function, resulting in vasocon- helical proteins, possess great biomechani-cal
striction or vasodilation. Such agonists strength, while elastin provides flexibility.
include circulating molecules (e.g., Together these components maintain the struc-
angiotensin II), those released from local tural integrity of the vessel, despite the high
nerve terminals (e.g., acetylcholine), and pressure within the lumen. The extracellular
others originating from the overlying matrix also regulates the growth of its resident
endothelium (e.g., endothelin and NO). cells. Native fibrillar collagen, in particular, can
Normal biosynthetic functions of smooth inhibit smooth muscle cell proliferation in vitro.
muscle cells include production of the colla-gen, Furthermore, the matrix influences cel-lular
elastin, and proteoglycans that form the responses to stimuli—matrix-bound cells
vascular extracellular matrix (see Fig. 5.2). respond in a specific manner to growth factors
Smooth muscle cells can also synthesize vaso- and are less likely to undergo apoptosis (pro-
active and inflammatory mediators, including grammed cell death).

115
Chapter 5

Atherosclerotic Arterial Wall the cells of atherosclerotic lesions continu-


ously interact and compete with each other,
The arterial wall is a dynamic and regulated
shaping the plaque over decades into one of
system, but noxious elements can disturb
many possible profi les. This section catego-
normal homeostasis and pave the way for
rizes these mechanisms into three pathologic
atherogenesis. For example, as described later,
stages: the fatty streak, plaque progression,
vascular endothelial and smooth muscle cells
and plaque disruption (Fig. 5.4).
react readily to inflammatory mediators such as
IL-1 and TNF-". These inflammatory agents can
Fatty Streak
also activate vascular cells to produce IL-1 and
TNF-"—contrary to past dogma stating that only Fatty streaks represent the earliest visible le-
cells of the immune system synthe-size such sions of atherosclerosis. On gross inspection,
cytokines. they appear as areas of yellow discoloration on
Realizing that immune cells were not the the artery’s inner surface, but they nei-ther
only source of proinfl ammatory agents, inves- protrude substantially into the arterial lumen nor
tigations into the role of “activated” endothe-lial impede blood fl ow. Surprisingly, fatty streaks
and smooth muscle cells in atherogenesis exist in the aorta and coronary arteries of most
burgeoned. This fundamental research has people by age 20. They do not cause
identifi ed several key components that con- symptoms, and in some locations in the
tribute to the atherosclerotic infl ammatory vasculature, they may regress over time.
process, including endothelial dysfunction, Although the precise initiation of fatty streak
accumulation of lipids within the intima, re- development is not known, observations in
cruitment of leukocytes and smooth muscle animals suggest that various stressors cause
cells to the vessel wall, formation of foam cells, early endothelial dysfunction, as described in
and deposition of extracellular matrix (Fig. 5.3), the next section. Such dysfunction allows entry
as described in the following sec-tions. Rather and modifi cation of lipids within the
than follow a sequential path, subendothelial space, where they serve as

Foam cell
8
1 Monocytes
Cell apoptosis
LDL 4 Macrophage
Cell
Vascular adhesion Smooth muscle
endothelium molecule mitogens
Scavenger Smooth muscle
Oxidized LDL IL-1 MCP-1
receptor proliferation
5
Internal elastic Smooth muscle
3
2
7

lamina 6 migration

Figure 5.3. Schematic diagram of the evolution of atherosclerotic plaque. (1) Accumulation of lipoprotein
par-ticles in the intima. The darker color depicts modification of the lipoproteins (e.g., by oxidation or glycation).
(2) Oxidative stress, including constituents of mLDL, induces local cytokine elaboration. (3) These cytokines
promote increased expression of adhesion molecules that bind leukocytes and of chemoattractant molecules
(e.g., monocyte chemoattractant protein 1 [MCP-1]) that direct leukocyte migration into the intima. (4) After
entering the artery wall in response to chemoattractants, blood monocytes encounter stimuli such as macrophage
colony–stimulating factor (M-CSF) that augment their expression of scavenger receptors. (5) Scavenger
receptors mediate the uptake of modified lipoprotein particles and promote the development of foam cells.
Macrophage foam cells are a source of additional cytokines and effector molecules such as superoxide anion
(O2−) and matrix metalloproteinases. (6) Smooth muscle cells migrate into the intima from the media. Note the
increasing intimal thickness. (7) Intimal smooth muscle cells divide and elaborate extracellular matrix, promoting
matrix accumulation in the growing atherosclerotic plaque. In this manner, the fatty streak evolves into a fibrofatty
lesion. (8) In later stages, calcification can occur (not depicted) and fibrosis continues, sometimes accompanied
by smooth muscle cell death (including programmed cell death, or apoptosis), yielding a relatively acellular
fibrous capsule surrounding a lipid-rich core that may also contain dying or dead cells. IL-1, interleukin 1; LDL,
low-density lipoprotein. (Modified from Zipes D, Libby P, Bonow RO, et al., eds. Braunwald’s Heart Disease: A
Textbook of Cardiovascular Medicine. 7th ed. Philadelphia, PA: Elsevier Saunders; 2005:925.)

116
Atherosclerosis

A B C

FATTY STREAK PLAQUE PROGRESSION PLAQUE DISRUPTION

• Endothelial dysfunction • Smooth muscle cell migration • Disrupted plaque integrity


• Lipoprotein entry and • Altered matrix synthesis and • Thrombus formation
modification degradation
• Leukocyte recruitment
• Foam cell formation
Figure 5.4. Stages of plaque development. A. The fatty streak develops as a result of endothelial dysfunction,
lipoprotein entry and modification, leukocyte recruitment, and foam cell formation. B. Plaque progression is char-
acterized by migration of smooth muscle cells into the intima, where they divide and elaborate extracellular matrix.
The fibrous cap contains a lipid core. C. Hemodynamic stresses and degradation of extracellular matrix increase
the susceptibility of the fibrous cap to rupture, allowing superimposed thrombus formation. (Modified from Libby P,
Ridker PM, Maseri A. Inflammation and atherosclerosis. Circulation. 2002;105:1136.)

proinfl ammatory mediators that initiate leu- Endothelial Dysfunction


kocyte recruitment and foam cell formation—
Injury to the arterial endothelium represents a
the pathologic hallmarks of the fatty streak
primary event in atherogenesis. Such in-jury
(Fig. 5.5).
can result from exposure to diverse agents,
including physical forces and chemi-cal
Chemical irritants Hemodynamic stress irritants.
The predisposition of certain regions of
arteries (e.g., branch points) to develop
Endothelial Dysfunction
atheromata supports the role of hydro-
dynamic stress. In straight sections of arter-
ies, the normal laminar (i.e., smooth) shear
Lipoprotein entry & Inflammatory forces favor the endothelial production of NO,
modification cytokines
which is an endogenous vasodilator, an
inhibitor of platelet aggregation, and an anti-
infl ammatory substance (see Chapter 6).
Chemokines Leukocyte adhesion
Moreover, laminar fl ow not only activates
molecules KLF-2 as described above, but also accen-
tuates expression of the antioxidant en-zyme
Leukocyte recruitment superoxide dismutase, which protects against
reactive oxygen species produced by
Unregulated uptake chemical irritants or transient ischemia.
of modified LDL Conversely, disturbed fl ow occurs at arterial
branch points, which impairs these locally
Foam Cell Formation
atheroprotective endothelial functions. Ac-
cordingly, arteries with few branches (e.g.,
Figure 5.5. Endothelial dysfunction is the primary event
in plaque initiation. Physical and chemical stressors alter the internal mammary artery) show relative
the normal endothelium, allowing lipid entry into the subintima resistance to atherosclerosis, whereas bifur-
and promoting inflammatory cytokine release. This cytokine- cated vessels (e.g., the common carotid and
and lipid-rich environment promotes recruitment of leukocytes
to the subintima, where they may become foam cells—a
left coronary arteries) are common sites for
prominent inflammatory participant. atheroma formation.

117
Chapter 5

Endothelial dysfunction may also result from Hypertension, a major risk factor for ath-
exposure to a “toxic” chemical environment. For erosclerosis, may promote retention of lipo-
example, tobacco smoking, abnormal cir- proteins in the intima by accentuating the
culating lipid levels, and diabetes—all known production of LDL-binding proteoglycans by
risk factors for atherosclerosis—can promote smooth muscle cells.
endothelial dysfunction. Each of these states in- Oxidation is one type of modifi cation that
creases endothelial production of reactive oxy- befalls LDL trapped in the subendothelial space.
gen species—notably, superoxide anion—which It can result from the local action of reactive
interact with other intracellular molecules to in- oxygen species and pro-oxidant en-zymes
fluence the metabolic and synthetic functions of derived from activated endothelial or smooth
the endothelium. In such an environment, the muscle cells, or from macrophages that
cells promote local inflammation. penetrate the vessel wall. In addition, the
When physical and chemical stressors microenvironment of the subendothelial space
interrupt normal endothelial homeostasis, an can sequester oxidized LDL from an-tioxidants
activated state ensues, manifested by in the plasma. In diabetic patients with
impairment of the endothelium’s role as a sustained hyperglycemia, glycation of LDL can
permeability barrier, release of infl amma-tory occur—a modifi cation that may ultimately
cytokines, increased production of cell render LDL antigenic and proin-flammatory.
surface adhesion molecules that recruit leu- These biochemical modifi cations of LDL act
kocytes, altered release of vasoactive sub- early and contribute to the inflam-matory
stances (e.g., prostacyclin and NO), and mechanisms initiated by endothelial
interference with normal antithrombotic dysfunction, and they may continue to pro-mote
properties. These undesired effects of en- infl ammation throughout the lifespan of the
dothelial dysfunction lay the groundwork for plaque. In the fatty streak, and likely throughout
subsequent events in the development of plaque development, modifi ed LDL (mLDL)
atherosclerosis (see Fig. 5.2). promotes leukocyte recruitment and foam cell
formation.
Lipoprotein Entry and Modification
Leukocyte Recruitment
The activated endothelium no longer serves
as an effective barrier to the passage of cir- Recruitment of leukocytes (primarily monocytes
culating lipoproteins into the arterial wall (see and T lymphocytes) to the vessel wall is a key step
Box 5.1 for a summary of the major in atherogenesis. The process depends on the
lipoprotein pathways). Increased endothe-lial expression of LAM on the normally nonadherent
permeability allows the entry of low-density endothelial luminal surface, and on chemoat-
lipoprotein (LDL) into the intima, a process tractant signals (e.g., monocyte chemotactic
facilitated by an elevated circulating LDL protein 1 [MCP-1], IL-8, interferon-inducible
concentration. In addition to high LDL protein-10) that direct diapedesis (passage of cells
concentrations from excessive dietary intake, through the intact endothelial layer) into the
several monogenic causes of elevated LDL subintimal space. Two major subsets of LAM
exist, including mutations in the LDL recep- persist in the inflamed atherosclerotic plaque: the
tor, apolipoprotein B, and PCSK9, a protease immunoglobulin gene superfam-ily (particularly,
involved in regulation of the LDL receptor. vascular cell adhesion mol-ecule 1 [VCAM-1] and
Once within the intima, LDL accumulates in intercellular adhesion molecule 1 [ICAM-1]) and
the subendothelial space by binding to com- the selectins (par-ticularly, E- and P-selectin).
ponents of the extracellular matrix known as Despite the central role of T lymphocytes in the
proteoglycans. This “trapping” increases the immune system, plaque LAM and chemoattractant
residence time of LDL within the ves-sel wall, signals direct mainly monocytes to the forming
where the lipoprotein may undergo chemical lesion. Re-cent research shows that
modifi cations that appear critical to the hypercholesterolemia favors accumulation in blood
development of atherosclerotic lesions. of a particularly

118
Atherosclerosis

proinflammatory subset of monocytes, charac- Plaque Progression


terized by expression of high levels of proin-
Whereas endothelial cells play a central role in
flammatory cytokines (e.g., IL-1 and TNF-1),
formation of the fatty streak, smooth muscle cell
distinguished in mice by expression of the cell
migration into the intima dominates early plaque
surface marker Ly6c. Although outnumbered by
progression. During decades of devel-opment,
macrophages, T lymphocytes localize within
plaques at all stages, where they likely furnish the typical atherosclerotic plaque ac-quires a
an important additional source of cytokines. distinct thrombogenic lipid core that underlies a
mLDL and proinflammatory cytokines can protective fibrous cap. Not all fatty streaks
induce LAM and chemoattractant cytokine progress into fibrofatty lesions, and it is
(che-mokine) expression independently, but unknown why some evolve and others do not.
mLDL also potently stimulates endothelial
Early plaque growth shows a compensatory
and smooth muscle cells to produce
outward remodeling of the arterial wall that
proinflamma-tory cytokines, thereby
preserves the diameter of the lumen and per-
reinforcing the direct action. This dual ability
mits plaque accumulation without limitation of
of mLDL to promote leukocyte recruitment
blood flow, hence producing no ischemic
and inflammation di-rectly and indirectly
symptoms. This stage can even evade detec-
persists throughout atherogenesis.
tion by angiography. Later plaque growth,
however, can outstrip the compensatory arte-rial
Foam Cell Formation enlargement, restrict the vessel lumen, and
impede perfusion. Such flow-limiting plaques
After monocytes adhere to and penetrate the
can result in tissue ischemia, causing symp-
intima, they differentiate into phago-cytic
toms such as angina pectoris (see Chapter 6) or
macrophages and imbibe lipoproteins to form
intermittent claudication of the extremities (see
foam cells. It is important to note that foam cells
Chapter 15).
do not arise from uptake of LDL by the classic
Most acute coronary syndromes (acute
cell–surface LDL-receptor mechanism
myocardial infarction and unstable angina
described in Box 5.1, because the high
pectoris) result when the fi brous cap of an
cholesterol content within these cells actually
atherosclerotic plaque ruptures, exposing
suppresses expression of the recep-tor.
prothrombotic molecules within the lipid core
Furthermore, the classic LDL receptor does not
and precipitating an acute thrombus that
recognize chemically mLDL. Rather,
suddenly occludes the arterial lumen. As
macrophages rely on a family of “scavenger”
described in this section, the extracellular
receptors that preferentially bind and inter-
matrix plays a pivotal role in fortifying the fi
nalize mLDL. Unlike uptake via the classic LDL
brous cap, isolating the thrombogenic plaque
receptor, mLDL ingestion by scavenger
interior from coagulation substrates in the
receptors evades negative feedback inhibition
circulation.
and permits engorgement of the macrophages
with cholesterol and cholesteryl ester, result-ing
in the typical appearance of foam cells.
Smooth Muscle Cell Migration
Although such uptake initially may be benefi -
cial (by sequestering proinfl ammatory mLDL The transition from fatty streak to fi brous
particles), the impaired effl ux of these cells, as atheromatous plaque involves the migra-tion
compared to the amount of infl ux, leads to local of smooth muscle cells from the arte-rial
accumulation in the plaque, mitigat-ing their media into the intima, proliferation of the
protective role and fueling foam cell apoptosis smooth muscle cells within the intima, and
and release of proinfl ammatory cytokines that secretion of extracellular matrix macro-
promote atherosclerotic plaque progression. molecules by the smooth muscle cells. Foam
The lipid-rich center of a plaque, formed by cells, activated platelets entering through
necrotic foam cells, is often called the necrotic microfi ssures in the plaque surface, and
core. endothelial cells all elaborate substances that

119
Chapter 5

BOX 5.1 The Lipoprotein Transport System

Lipoproteins ferry water-insoluble fats through the bloodstream. These particles consist of a lipid core
surrounded by more hydrophilic phospholipid, free cholesterol, and apolipoproteins (also called apopro-
teins). The apoproteins present on various classes of lipoprotein molecules serve as the “conductors” of
the system, directing the lipoproteins to specific tissue receptors and mediating enzymatic reactions. Five
major classes of lipoproteins exist, distinguished by their densities, lipid constituents, and associated apo-
proteins. In order of increasing density, they are chylomicrons, very low density lipoproteins (VLDL),
intermediate-density lipoproteins (IDL), low-density lipoproteins (LDL), and high-density lipopro-
teins (HDL). The major steps in the lipoprotein pathways are labeled in the accompanying figure and
described as follows. The key apoproteins (apo) at each stage are indicated in the figure in parentheses.

Exogenous Pathway Endogenous Pathway

Dietary fat
5
Bile acids and Nonhepatic
1
cholesterol cells
10
LDL
Liver (Apo B-100)
Intestine LPL
9
4 6 8 HL

Chylomicron
remnants VLDL IDL
Chylomicrons (Apo B-100, C, E) (Apo B-100, E)
(Apo B-48, A,C, E) (Apo B-48, E)

3 7
E Lipoprotein lipase E Lipoprotein lipase
, ,
)
2 C C TP
o
o

p p E
A A C
(
FFA FFA
HDL
HDL

Muscle Adipose tissue Muscle Adipose tissue

Exogenous (Intestinal) Pathway


1. Dietary fats are absorbed by the small intestine and repackaged as chylomicrons,
accompanied by apo B-48. Chylomicrons are large particles, particularly rich in
triglycerides, that enter the circulation via the lymphatic system.
2. Apo E and subtypes of apo C are transferred to chylomicrons from HDL particles in the bloodstream.
3. Apo C (subtype CII) enhances interactions of chylomicrons with lipoprotein lipase (LPL)
on the endothelial surface of adipose and muscle tissue. This reaction hydrolyzes the
triglycerides within chylomicrons into free fatty acids (FFA), which are stored by adipose
tissue or used for energy in cardiac and skeletal muscle.
4. Chylomicron remnants are removed from the circulation by the liver, mediated by apo E.
5. One fate of cholesterol in the liver is incorporation into bile acids, which are exported to
the in-testine, completing the exogenous pathway cycle.
Endogenous (Hepatic) Pathway
Because dietary fat availability is not constant, the endogenous pathway provides a reliable
supply of triglycerides for tissue energy needs:

120
Atherosclerosis

6. The liver packages cholesterol and triglycerides into VLDL particles, accompanied by apo B-
100 and phospholipid. The triglyceride content of VLDL is much higher than that of cholesterol,
but this is the main means by which the liver releases cholesterol into the circulation.
7. VLDL is catabolized by LPL (similar to chylomicrons, as described in step 3), releasing
fatty acids to muscle and adipose tissue. During this process, VLDL also interacts with
HDL, exchanging some of its triglyceride for apo C subtypes, apo E, and cholesteryl ester
from HDL. The latter exchange (important in reverse cholesterol transport, as described in
the next section) is mediated by cho-lesteryl ester transfer protein (CETP).
8. Approximately 50% of the VLDL remnants (termed intermediate-density lipoproteins [IDL])
are then cleared in the liver by hepatic receptors that recognize apo E.
9. The remaining IDL is catabolized further by LPL and hepatic lipase (HL), which remove
additional triglyceride, apo E, and apo C, forming LDL particles.
10. Plasma clearance of LDL occurs primarily via LDL receptor–mediated endocytosis in the
liver and peripheral cells, directed by LDL’s apo B-100 and apo E.

Cholesterol Homeostasis and Reverse Cholesterol Transport


Intracellular cholesterol content is tightly maintained by de novo synthesis, cellular uptake, storage, and
efflux from the cell. The enzyme HMG-CoA reductase is the rate-limiting element of cholesterol biosyn-
thesis, and cellular uptake of cholesterol is controlled by receptor-mediated endocytosis of circulating LDL
(see step 10). When intracellular cholesterol levels are low, the transcription factor sterol regulatory
element binding protein (SREBP) is released from the endoplasmic reticulum. The active fragment of
SREBP enters the nucleus to increase transcription of HMG-CoA reductase and the LDL receptor—which,
through their subsequent actions, tend to normalize the intracellular cholesterol content.

apo AI
Internalized
cholesterol SR-BI Uptake by
ester receptor liver and steroid
Free Nascent hormone-
apo AI
HDL producing tissues
Mature
HDL CETP

Transfer of
ABCAI ABCGI cholesterol to
VLDL, IDL, LDL
Peripheral for transport to liver
cells

Excess
cholesterol

Under conditions of intracellular cholesterol excess (Figure above), peripheral cells increase the tran-scription
of the ATP-binding cassette A1 and G1 genes (ABCA1 and ABCG1, respectively). The ABCA1 gene codes for a
transmembrane protein transporter that initiates efflux of cholesterol from the cell to lipid-poor circulating apo AI
(which is synthesized by the liver and intestine), thus forming nascent (immature) HDL particles. ABCG1
facilitates further efflux of cholesterol to form more mature HDL particles. As free choles-terol is acquired by
circulating HDL, it is esterified by lecithin cholesterol acyltransferase (LCAT), an enzyme activated by apo AI. The
hydrophobic cholesterol esters move into the particle’s core. Most cholesterol esters in HDL can then be
exchanged in the circulation (via the enzyme CETP) with any of the apo B–containing lipoproteins (i.e., VLDL,
IDL, LDL), which deliver the cholesterol back to the liver. HDL can also transport cholesterol to the liver and
steroid hormone–producing tissues via the SR-B1 scavenger receptor.

121
Chapter 5

current evidence suggests that this progres-


Foam Cells in Fatty Streak sion may be punctuated by subclinical events
with bursts of smooth muscle replication. For
Endothelial
Tissue example, morphologic evidence of resolved
Dysfunction
factor intraplaque hemorrhages indicates that small
breaches in plaque integrity can occur with-
NO
out clinical symptoms or signs. At the cellular
PGI2
PDGF Thrombosis level, such breaches expose tissue factor
and platelet from foam cells, which activates coagulation
activation and microthrombus formation. Activated
Heparinase plate-lets within such microthrombi release
Endothelial
heparan
addi-tional potent factors—including PDGF
sulfate and heparinase—that can spur a local wave
of smooth muscle cell migration and prolifera-
Smooth muscle cells migrate to intima, tion. Heparinase degrades heparan sulfate, a
proliferate, and produce extracellular matrix polysaccharide in the extracellular matrix that
normally inhibits smooth muscle cell
Fibrous migration and proliferation. Moreover, other
Plaque forms of lymphocytes—such as Treg and Th2
lymphocytes—may produce factors, includ-
Figure 5.6. Progression from the fatty streak ing TGF-# and IL-10, respectively, which can
involves the migration and proliferation of smooth inhibit smooth muscle cell proliferation and
muscle cells. Substances released from foam cells,
dysfunctional endothelial cells, and platelets thus regulate plaque growth.
contribute to this pro-cess. IL -1, interleukin-1; NO,
nitric oxide; PDGF, platelet-derived growth factor;
PGI2 , prostacyclin; TGF-#, transforming growth Extracellular Matrix Metabolism
factor-#; TNF-", tumor necrosis factor-".
As the predominant collagen-synthesizing cell
signal smooth muscle cell migration and pro- type, smooth muscle cells should, through their
liferation (Fig. 5.6). proliferation, favor fortification of the fibrous cap.
Foam cells produce several factors that con- Net matrix deposition depends on the balance
tribute to smooth muscle cell recruitment. For of synthesis by smooth muscle cells and
example, they release platelet-derived growth degradation, mediated in part by a class of
factor (PDGF)—also produced by platelets and proteolytic enzymes known as matrix metal-
endothelial cells—which likely stimulates the loproteinases (MMP). While PDGF and TGF-#
migration of smooth muscle cells across the stimulate smooth muscle cell production of in-
internal elastic lamina and into the subinti-mal terstitial collagens, the T-lymphocyte–derived
space, where they subsequently replicate. cytokine interferon-$ (IFN-$) inhibits smooth
PDGF additionally stimulates the growth of resi- muscle cell collagen synthesis. Furthermore,
dent smooth muscle cells in the intima. Foam inflammatory cytokines stimulate local foam
cells also release cytokines and growth factors cells to secrete collagen- and elastin-degrading
(e.g., TNF-", IL-1, fibroblast growth factor, and MMP, thereby weakening the fibrous cap and
transforming growth factor-# [TGF-#]) that fur- predisposing it to rupture (Fig. 5.7).
ther incite smooth muscle cell proliferation and
synthesis of extracellular matrix proteins. Fur-
thermore, these stimulatory cytokines induce Plaque Disruption
smooth muscle cell and leukocyte activation, Plaque Integrity
promoting further cytokine release, thus rein-
forcing and maintaining inflammation in the The tug of war between matrix synthesis and
lesion. degradation continues over decades, but not
According to the traditional concept, without consequences. Death of smooth muscle
plaques grow gradually and continuously, but and foam cells, either owing to excess

122
Atherosclerosis

sis De
gr a d ati on
e
th
n
y

Lumen

MMP
Collagen and
elastin Fibrous Cap

Smooth muscle
+ cell Foam +
– + cell
IFN-γ CD40L

Lipid core
T lymphocyte
Figure 5.7. Matrix metabolism underlies fibrous cap integrity. The net deposition of extracellular matrix is
the result of competing synthesis and degradation reactions. Smooth muscle cells synthesize the bulk of the
fibrous cap constituents, such as collagen and elastin. Foam cells elaborate destructive proteolytic enzymes,
such as the collagen-degrading matrix metalloproteinases and the elastolytic cathepsins. T-lymphocyte–
derived factors favor destruction of the fibrous cap. All plaque residents, however, contribute to the cytokine
milieu of the plaque, providing multiple activating and inhibitory stimuli as shown. IFN- $, interferon-$; IL-1,
interleukin-1; MCP-1, monocyte chemoattractant protein 1; PDGF, platelet-derived growth factor; TGF-#,
transforming growth factor-#; TNF-", tumor necrosis factor-". (Modified from Libby P. The molecular bases of
acute coronary syndromes. Circu-lation. 1995;91:2844–2850; Young JL, Libby P, Schönbeck U. Cytokines in
the pathogenesis of atherosclerosis. Thromb Haemost. 2002;88:554–567.)

inflammatory stimulation or by contact acti- the extreme spectrums of integrity as “stable


vation of apoptosis pathways, liberates cellu- plaques” (marked by a thick fi brous cap and
lar contents, contributing imbibed lipids and small lipid core) or “vulnerable plaques”
cellular debris to the growing lipid core. The (marked by a thin fi brous cap, rich lipid core,
size of the lipid core has biomechanical im- extensive macrophage infi ltrate, and a pau-
plications for the stability of the plaque. With city of smooth muscle cells; Fig. 5.8). De-
increasing size and protrusion into the arte- spite the common use of these terms, it is
rial lumen, mechanical stress focuses on the important to recognize that this distinction
plaque border abutting normal tissue, the so- oversimplifi es the heterogeneity of plaques
called shoulder region. In addition to bearing and may overestimate the ability to foresee a
increased stress, local accumulation of foam plaque’s “clinical future” based on structural
cells and T lymphocytes at this site information.
accelerates degradation of extracellular
matrix, making this region the most common
site of plaque rupture.
Thrombogenic Potential
The net deposition and distribution of the fi Rupture of atherosclerotic plaque does not
brous cap is an important determinant of inevitably cause major clinical events such as
overall plaque integrity. Whereas lesions with myocardial infarction and stroke. As de-
thick fi brous caps may cause pronounced scribed in the previous section, small non-
arterial narrowing, they have less propensity occlusive thrombi may reabsorb into the
to rupture. Conversely, plaques that have plaque, stimulating further smooth muscle
thinner caps (and often appear less obstruc- growth and fi brous deposition (see Fig. 5.8).
tive by angiography) tend to be fragile, and It is in large part the balance between the
more likely to rupture and incite thrombo-sis. thrombogenic and fi brinolytic potential of the
Current clinical terminology describes plaque, and the fl uid phase of blood,

123
Chapter 5

Normal artery

Early atheroma

“Stable” plaque “Vulnerable” plaque


• Small lipid pool • Large lipid pool
• Thick fibrous cap • Thin fibrous cap
• Preserved lumen • Many inflammatory cells

Ruptured plaque
with thrombus formation

Healed rupture
• Narrowed lumen Acute
• Fibrous intima myocardial
infarction

Figure 5.8. Stable versus vulnerable plaques. Stable plaque is characterized by a small lipid core and a thick
fibrous cap, whereas vulnerable plaque tends to have a large lipid core and a relatively thin fibrous cap. The latter
is subject to rupture, resulting in thrombosis. A resulting occlusive clot can cause an acute cardiac event, such as
myocardial infarction. A lesser thrombus may resorb, but the wound-healing response stimulates smooth muscle
cell proliferation and collagen production, thereby thickening the fibrous cap and narrowing the vessel lumen
further. (Modified from Libby P. Inflammation in athero-sclerosis. Nature. 2002;420:868–874.)

that determines whether disruption of the fi The probability of a major thrombotic event
brous cap leads to a transient, nonobstruc- refl ects the balance between the com-peting
tive mural thrombus or to a completely oc- processes of coagulation and fi brin-olysis.
clusive clot. Infl ammatory stimuli common in

124
Atherosclerosis

Favor Occlusive Thrombus Resist Thrombus Accumulation


Procoagulant Anticoagulants
•Tissue factor • Thrombomodulin
• Protein S, Protein C
• Heparin-like molecules
Antifibrinolytic Profibrinolytics
• PAI-1 • tPA, uPA

Figure 5.9. Competing factors in thrombosis. The clinical manifestations of


plaque disruption rely not only on the stability of the fibrous cap, but also on the
throm-bogenic potential of the plaque core. The balance of physiologic mediators
dictates the prominence of the thrombus, resulting in either luminal occlusion or
resorption into the plaque. PAI-1, plasminogen activator inhibitor 1; tPA, tissue
plasminogen activator; uPA, urokinase-type plasminogen activator.

the plaque microenvironment (e.g., CD40L) of the abdominal aorta and proximal coronary
elicit tissue factor, the initiator of the extrin-sic arteries, followed by the popliteal arteries, de-
coagulation pathway, from many plaque scending thoracic aorta, internal carotid arter-
components, including smooth muscle cells, ies, and renal arteries. Therefore, the regions
endothelial cells, and macrophage-derived perfused by these vessels most commonly
foam cells. Beyond enhancing expression of suf-fer the consequences of atherosclerosis.
the potent procoagulant tissue factor, infl am- Complications of atherosclerotic plaques—
matory stimuli further support thrombosis by including calcification, rupture, hemorrhage, and
favoring the expression of antifi brinolyt-ics embolization—can have dire clinical con-
(e.g., plasminogen activator inhibitor-1) over sequences due to acute restriction of blood flow
the expression of anticoagulants (e.g., or alterations in vessel wall integrity. These
thrombomodulin, heparin-like molecules, complications, which are discussed in greater
protein S) and profi brinolytic mediators (e.g., detail in later chapters, include the following:
tissue plasminogen activator and urokinase-
• Calcification of atherosclerotic plaque,
type plasminogen activator; Fig. 5.9). More-
which imparts a pipe-like rigidity to the ves-
over, as described earlier, the activated
sel wall and increases its fragility.
endothelium also promotes thrombin forma-
tion, coagulation, and fi brin deposition at the • Rupture or ulceration of atherosclerotic
vascular wall. plaque, which exposes procoagulants
A person’s propensity toward coagulation within the plaque to circulating blood,
may be enhanced by genetics (e.g., the pres- causing a thrombus to form at that site.
ence of a procoagulant prothrombin gene Such throm-bosis can occlude the vessel
mutation), comorbid conditions (e.g., diabe- and result in infarction of the involved
tes), and/or lifestyle factors (e.g., smoking, organ. Alterna-tively, the thrombus material
visceral obesity). Consequently, the concept can incorpo-rate into the lesion and add to
of the “vulnerable plaque” has expanded to the bulk of the plaque.
that of the “vulnerable patient,” to acknowl- • Hemorrhage into the plaque owing to rup-
edge other contributors to a person’s ture of the fibrous cap or of the
vascular risk. microvessels that form within the lesion.
The resulting in-tramural hematoma may
further narrow the vessel lumen.
Complications of Atherosclerosis • Embolization of fragments of disrupted
Atherosclerotic plaques do not distribute atheroma to distal vascular sites.
homogeneously throughout the vasculature. • Weakening of the vessel wall: the fibrous
They usually develop first in the dorsal aspect plaque subjects the neighboring medial

125
Chapter 5

layer to increased pressure, which may to acute thrombosis and myocardial infarction
pro-voke atrophy and loss of elastic tissue (see Chapter 7). Such nonstenotic plaques
with subsequent expansion of the artery, are often numerous and dispersed
forming an aneurysm. throughout the arterial tree, and because they
• Microvessel growth within plaques, provid- do not limit ar-terial flow, they do not produce
ing a source for intraplaque hemorrhage symptoms and often evade detection by
and further leukocyte trafficking. exercise testing or angiography.
The description presented here of athero-
The complications of atherosclerotic plaque genesis and its complications can explain the
may result in specific clinical consequences in limitations of widely employed treat-ments.
different organ systems (Fig. 5.10). In the case For example, percutaneous interven-tion
of coronary plaque, lesions with gradu-ally (angioplasty and stent placement) of
progressive expansion and a thick fibrous cap symptomatic coronary stenoses effectively
tend to narrow the vessel lumen and cause relieves angina pectoris, but does not neces-
intermittent chest discomfort on exertion (an- sarily prevent future myocardial infarction or
gina pectoris). In contrast, plaque that does not prolong life. This disparity likely refl ects the
compromise the vessel lumen but has multiplicity of nonocclusive plaques at risk of
characteristics of vulnerability (a thin fibrous cap precipitating thrombotic events. It follows that
and large lipid core) can rupture, leading lifestyle modifi cations and drug therapies

Stroke
4
• Embolic stroke
• Thrombotic stroke 2 3 Coronary artery disease
• Myocardial ischemia 1
• Unstable angina 2 3
• Myocardial infarction 2 3

Renal Artery Disease


• Atheroembolic renal disease 4
• Renal artery stenosis 1

Aneurysms 5

Peripheral artery disease


• Limb claudication 1
• Limb ischemia 1 4
1 Narrowing of vessel by fibrous plaque
2 Plaque ulceration or rupture
3 Intraplaque hemorrhage
4 Peripheral emboli
5 Weakening of vessel wall

Figure 5.10. Clinical sequelae of atherosclerosis. Complications of atherosclerosis arise from the
mechanisms listed in the figure.

126
Atherosclerosis

that curb the risk factors for plaque forma- Lp(a), and certain markers of inflammation,
tion, and lessen features associated with including the acute-phase reactant C-reactive
“vul-nerability,” furnish a critical foundation for protein (CRP). Furthermore, recent genome-
preventing progression and complications of wide association studies (GWAS) have sought
atherosclerosis. to identify variants in genetic loci associated
with increased cardiovascular risk.
ATHEROSCLEROSIS RISK FACTORS The following sections address these risk
factors and biologic markers.
In the early 20th century, it was widely be-lieved
that atherosclerosis was an inevitable process
of aging. But in 1948, the landmark Traditional Risk Factors
Framingham Heart Study began to examine the
Dyslipidemia
relationship between specific attributes and
cardiovascular disease, establishing the A large and consistent body of evidence es-
concept of atherosclerotic risk factors. Among tablishes abnormal circulating lipid levels as a
later studies, the Multiple Risk Factor Inter- major risk factor for atherosclerosis. Obser-
vention Trial (MRFIT) screened more than vational studies have shown that in the United
325,000 men to correlate risk factors with sub- States and other societies where there is high
sequent cardiovascular disease and mortality. consumption of saturated fat and prevalent hy-
These studies and others have established the percholesterolemia, there is greater mortality
importance of modifiable risk factors for ath- from coronary disease than in countries with
erosclerosis, including aberrant levels of circu- traditionally low saturated fat intake and low
lating lipids (dyslipidemia), tobacco smoking, serum cholesterol levels (e.g., rural Japan and
hypertension, diabetes mellitus, and lack of certain Mediterranean nations). Similarly, data
physical activity and obesity (Table 5.1). Major from the Framingham Heart Study and other
nonmodifiable risk factors include ad-vanced cohorts have shown that the risk of ischemic
age, male sex, and heredity—that is, a history heart disease increases with higher total serum
of coronary heart disease among first-degree cholesterol levels. The coronary risk is
relatives at a young age (before age 55 for a approximately twice as high for a per-son with a
male relative or before age 65 for a female total cholesterol level of 240 mg/dL compared
relative). with a person whose cholesterol level is 200
In addition to these standard predictors, mg/dL.
certain biologic markers associated with the In particular, elevated levels of circulating
development of cardiovascular events have LDL correlate with an increased incidence of
been undergoing rigorous evaluation as atherosclerosis and coronary artery disease.
“novel” risk markers. These include elevated When present in excess, LDL can accumulate in
circulating levels of the amino acid metabolite the subendothelial space and undergo the
homocysteine, the special lipoprotein particle chemical modifications that further damage the
intima, as described earlier, initiating and
perpetuating the development of atheroscle-
Table 5.1. Common Cardiovascular Risk Factors
rotic lesions. Thus, LDL is commonly known as
Modifiable risk factors “bad cholesterol.” Conversely, elevated high-
Dyslipidemia (elevated LDL, decreased HDL) density lipoprotein (HDL) particles ap-pear to
Tobacco smoking protect against atherosclerosis, likely because
Hypertension of HDL’s ability to transport choles-terol away
Diabetes mellitus, metabolic
from the peripheral tissues back to the liver for
syndrome Lack of physical activity
disposal (termed “reverse choles-terol
Nonmodifiable risk factors transport”; see Box 5.1) and because of its
Advanced age
putative antioxidative and anti-inflammatory
Male gender
Heredity properties. Thus, HDL has earned the moniker
“good cholesterol.”

127
Chapter 5

Elevated serum LDL may persist for many syndrome (described later in this chapter), or
reasons, including a high-fat diet or abnor- tobacco smoking.
malities in the LDL-receptor clearance mecha- Diet and exercise comprise two important
nism. Patients with genetic defects in the LDL components of the risk-reduction arsenal. For
receptor, which leads to a condition known as example, the Lyon Diet Heart Study demon-
familial hypercholesterolemia, cannot remove strated that patients with coronary disease who
LDL from the circulation efficiently. Heterozy- were randomized to a Mediterranean-style diet
gotes with this condition have one normal and decreased their risk of recurrent cardiac events.
one defective gene coding for the receptor. The diet implemented in this study included
They display high plasma LDL levels and de- replacement of saturated fats with polyun-
velop premature atherosclerosis. Homozygotes saturated fats (particularly "-linolenic acid, an
who completely lack functional LDL receptors omega-3 fatty acid). In vitro evidence indi-cates
may experience vascular events, such as acute that polyunsaturated fats may activate a
myocardial infarction, as early as the first de- transcription factor (peroxisome proliferator-
cade of life. activated receptor-" [PPAR-"], or its obligate
Increasing evidence also implicates partner, retinoid X receptor), which increases
triglyceride-rich lipoproteins, such as very low expression of the major HDL apoprotein (apo
density lipoprotein (VLDL) and intermediate- AI) and the enzyme lipoprotein lipase, and
density lipoprotein (IDL), in the development inhib-its cytokine-induced expression of LAM on
of atherosclerosis. However, it remains unde- endothelial cells. These actions may oppose
termined whether these particles participate atherogenesis. Physical activity and loss of ex-
directly in atherogenesis or simply keep com- cess weight can also improve the lipid profile,
pany with low levels of HDL cholesterol. Of notably by lowering triglycerides and raising
note, poorly controlled type 2 diabetes mel- HDL.
litus commonly associates with the combi- When lifestyle modifi cations fail to
nation of hypertriglyceridemia and low HDL achieve target values, pharmacologic agents
levels. can im-prove abnormal lipid levels. The major
groups of lipid-altering agents (see Chapter
17) in-clude HMG-CoA reductase inhibitors
Lipid-Altering Therapy
(also known as statins), niacin, fi bric acid
Strategies that improve abnormal lipid levels deriva-tives, cholesterol intestinal absorption
can limit the consequences of atherosclerosis. in-hibitors, and bile acid–binding agents. The
Many large studies of patients with coronary statins have emerged as the most effective
disease show that dietary or pharmacologic re- LDL-lowering drugs. They inhibit the rate-
duction of serum cholesterol can slow the pro- limiting enzyme responsible for cholesterol
gression of atherosclerotic plaque. These trials biosynthesis. The resulting reduction in intra-
form the basis of the screening guidelines de- cellular cholesterol concentration promotes
vised by the National Cholesterol Education increased LDL-receptor expression and thus
Program panel, which recommend a fasting augmented clearance of LDL particles from
lipid profile every five years for all adults. The the bloodstream. Statins also lower the rate
guidelines specify an “optimal” LDL choles-terol of VLDL synthesis by the liver (thus lowering
level as % 100 mg/dL. Patients with es- circulating triglyceride levels), and raise HDL
tablished atherosclerosis, or those who have by an unknown mechanism.
equivalent risk (e.g., diabetes), should receive Major clinical trials evaluating statin ther-apy
treatment to attain this goal. An even lower goal have demonstrated reductions in ischemic
of % 70 mg/dL is recommended for pa-tients cardiac events, the occurrence of strokes, and
with atherosclerotic disease at the high-est risk (in many cases) mortality rates (Fig. 5.11). The
of future vascular events—those who have benefits documented in these studies have
recently sustained an acute coronary syn- extended to people with wide ranges of LDL,
drome (see Chapter 7) and those with multiple with or without known preexisting ath-
risk factors, especially diabetes, the metabolic erosclerotic disease. Furthermore, the effect

128
Atherosclerosis

Included patients Excluded patients with


with history of MI or CHD known CHD
Baseline LDL
or total High Normal Normal Normal High Normal Normal
cholesterol to High to High (low HDL)
35

AFCAPS / TexCAPS
30
in(%)
25
ReductionCHDevents

20

WOSCOPS
15

ASCOT
CARE

LIPID
10

HPS
4S

5
0

4S: Scandinavian Simvastatin Survival Study. Lancet. 1994;344:1383–1389.


AFCAPS/TexCAPS: The Air Force/Texas Coronary Atherosclerosis Prevention Study.
JAMA. 1998;279:1615–1622.
ASCOT: Anglo-Scandinavian Cardiac Outcomes Trial—Lipid Lowering Arm. Lancet.
2003;361:1149–1158.
CARE: Cholesterol And Recurrent Events. N Engl J Med. 1996;335:1001–1009.
HPS: Heart Protection Study. Lancet. 2004;363:757–767.
LIPID: The Long-term Intervention with Pravastatin in Ischaemic Disease. N Engl
J Med. 1998;339:1349–1357.
WOSCOPS: West of Scotland Coronary Primary Prevention Study. N Engl J Med.
1995;333:1301–1307.
Figure 5.11. Statins reduce cardiovascular risk. Several major clinical studies have
sup-ported the beneficial role of statin-induced lipid lowering in reducing coronary
heart disease (CHD) events. Regardless of baseline serum cholesterol levels, the
benefits of statins extend to patients with an established history of coronary disease
and to patients with significant risk factors who do not have such a history. HDL, high-
density lipoprotein; LDL, low-density lipoprotein.

may depend on the degree of LDL lowering the macrophage cytokines TNF-", IL-1, and
achieved, as demonstrated by the PROVE IT- IL-6, thereby reducing endothelial expres-
TIMI 22 and TNT studies, which showed that sion of LAM and macrophage tissue factor
intensive statin therapy (yielding lower LDL production. Some of these pleiotropic effects
levels) improves cardiovascular outcomes in may result from activation of the transcrip-tion
acute and chronic coronary heart disease more factor KLF2 and interference with pre-nylation
than moderate-dose statins. of small G proteins implicated in the
The clinical benefi ts of statins likely derive regulation of infl ammatory functions of
from several mechanisms. The combination vascular cells and leukocytes. Clinical trials
of lowering LDL and raising HDL may reduce have supported an anti-infl ammatory action
the lipid content of atherosclerotic plaques of statins because they reduce plasma levels
and thus favorably affect their biologic activ- of CRP, a serum marker of infl ammation de-
ity. Other potentially benefi cial actions (so- scribed later. It should be noted, however,
called “pleiotropic effects”) include increased that it is experimentally diffi cult to separate
NO synthesis, enhanced fi brinolytic activity, the LDL-lowering effect of statins from their
inhibition of smooth muscle proliferation and anti-infl ammatory mechanisms because of
monocyte recruitment, and reduction in the prominent role of oxidized LDL in initi-
macrophage production of matrix-degrading ating infl ammatory cascades. Nonetheless,
enzymes. In vitro studies suggest that statins accumulating clinical and experimental data
may also reduce infl ammation by inhibiting suggest that at least part of the benefi t of

129
Chapter 5

statins derives from mechanisms other than endothelium and may increase the permeabil-ity
LDL lowering. of the vessel wall to lipoproteins. In ad-dition to
causing direct endothelial damage, increased
hemodynamic stress can augment the number
Tobacco Smoking of scavenger receptors on mac-rophages, thus
Numerous studies have shown that tobacco enhancing the development of foam cells.
smoking increases the risk of atherosclerosis Cyclic circumferential strain, in-creased in
and ischemic heart disease. Even minimal hypertensive arteries, can enhance smooth
smoking increases the risk, and the heaviest muscle cell production of proteogly-cans that
smokers have the greatest risk of bind and retain LDL particles, pro-moting their
cardiovascu-lar events. accumulation in the intima and facilitating their
Tobacco smoking could lead to athero- oxidative modification. Angio-tensin II, a
sclerotic disease in several ways, including mediator of hypertension, acts not only as a
enhanced oxidative modification of LDL, de- vasoconstrictor, but also as a stimu-lator of
creased circulating HDL levels, endothelial oxidative stress (through activation of NADPH
dysfunction owing to tissue hypoxia and in- oxidases, a source of superoxide anion, O 2) −

creased oxidant stress, increased platelet ad- and as a proinflammatory cytokine. Thus,
hesiveness, increased expression of soluble hypertension may also promote athero-genesis
LAM, inappropriate stimulation of the sym- by contributing to a pro-oxidant and
pathetic nervous system by nicotine, and inflammatory state.
displacement of oxygen by carbon monoxide
from hemoglobin. Extrapolation from animal
Antihypertensive Therapy
experiments suggests that smoking not only
accelerates atherogenesis, but also Like dyslipidemias, treatment of hypertension
increases the propensity for thrombosis— should start with lifestyle modifications, but
both compo-nents of the “vulnerable patient.” often requires pharmacologic intervention. The
Fortunately, smoking cessation can reverse Dietary Approaches to Stop Hyperten-sion
some of the adverse outcomes. People who (DASH) studies demonstrate that a diet high in
stop smoking greatly reduce their likelihood of fruits and vegetables, with dairy prod-ucts low in
coronary heart disease, compared with those fat and an overall reduced sodium content,
who continue to smoke. In one study, after three significantly improves systolic and diastolic
years of cessation, the risk of cor-onary artery blood pressures. Regular exercise can also
disease for former smokers be-came similar to reduce resting blood pressure levels. Many
subjects who never smoked. medications effectively lower blood pressure, as
described in Chapters 13 and 17.
Hypertension
Diabetes Mellitus and the
Elevated blood pressure (either systolic or
dia-stolic) augments the risk of developing Metabolic Syndrome
athero-sclerosis, coronary heart disease, and The global incidence of diabetes mellitus is
stroke (see Chapter 13). The association of estimated at 170 million people and projected to
elevated blood pressure with cardiovascular grow 40% worldwide by 2030. In the United
disease does not appear to have a specific States alone, 18.2 million people are diabetic,
threshold. Rather, risk increases continuously and projections suggest that one in every three
with pro-gressively higher pressure values. children born in 2000 will eventually develop the
Systolic pressure predicts adverse outcomes condition. With a three- to fivefold in-creased
more reli-ably than does diastolic pressure, risk of acute coronary events, 80% of diabetic
particularly in older persons. patients succumb to atheroscle-rosis-related
Hypertension may accelerate atherosclero- conditions, including coronary heart disease,
sis in several ways. Animal studies have shown stroke, and peripheral artery disease.
that elevated blood pressure injures vascular Accordingly, some consider diabetes

130
Atherosclerosis

an atherosclerotic risk equivalent, elevating it little as 30 minutes per day can protect
to the same risk category as that of people against cardiovascular mortality.
with a history of myocardial infarction.
The predisposition of diabetic patients to
Estrogen Status
atherosclerosis may relate in part to the non-
enzymatic glycation of lipoproteins (which Cardiovascular disease dominates over other
enhances uptake of cholesterol by scavenger causes of mortality in women, including breast
macrophages, as described earlier), or to a pro- and other cancers. Before menopause, women
thrombotic tendency and antifibrinolytic state have a lower incidence of coronary events than
that is often present. Diabetics frequently have men. After menopause, however, men and
impaired endothelial function, gauged by the women have similar rates. This ob-servation
reduced bioavailability of NO and increased suggests that estrogen (the levels of which
leukocyte adhesion. Tight control of serum decline after menopause) may have
glucose levels in diabetic patients reduces the atheroprotective properties. Physiologic estro-
risk of microvascular complications, such as gen levels in premenopausal women raise HDL
retinopathy and nephropathy. At least one study and lower LDL. Experimentally, estrogen also
has also demonstrated a reduction in exhibits potentially beneficial antioxidant and
macrovascular outcomes, such as myocardial antiplatelet actions and improves endothelium-
infarction and stroke, in patients with type 1 dependent vasodilation.
diabetes who followed an intense antidiabetic Early observational studies suggested that
regimen. In addition, control of hyperten-sion hormone therapy reduced the risk of coronary
and dyslipidemia in diabetic patients artery disease in postmenopausal women,
convincingly reduces the risk of cardiac and prompting many physicians to prescribe such
cerebrovascular complications. medications for cardiovascular prevention
The metabolic syndrome (previously purposes. However, the Heart and Estrogen/
known as the “insulin resistance syndrome” or Progestin Replacement Study demonstrated an
“syndrome X”) is a descriptor for a clustering of association between such hormone use and an
risk factors, including hypertension, hyper- early increased risk of vascular events in
triglyceridemia, reduced HDL, cellular insulin women with preexisting coronary disease.
resistance (often leading to glucose intoler- Subsequent randomized primary preven-tion
ance), and visceral obesity (excessive adipose studies from the Women’s Health Initia-tive
tissue in the abdomen). This constellation as- were terminated prematurely because estrogen-
sociates with a high risk for atherosclerosis in plus-progestin treatment increased
both diabetic and nondiabetic patients, and the cardiovascular risk by 24% overall, with a
National Health and Nutrition Examina-tion striking 81% higher risk during the fi rst year of
Survey estimates that an astounding 44% of therapy. Of note, these troubling outcomes did
Americans have the metabolic syndrome based not appear in the cohort of patients ran-domized
on current guidelines. The presence of insulin to the estrogen-only arm of the study. Further
resistance in this syndrome appears to promote analyses will help determine if safe hormone
atherogenesis long before affected persons therapies are possible. Mean-while, because
develop overt diabetes. currently available clinical trial data do not show
that gonadal hormone therapy is
Lack of Physical Activity cardioprotective and that it may ac-tually be
harmful, such therapy should not be
Exercise may mitigate atherogenesis in several commenced for the sole goal of reducing
ways. In addition to its beneficial effects on the cardiovascular risk.
lipid profile and blood pressure, exercise en-
hances insulin sensitivity and endothelial pro-
duction of NO. Long-term prospective studies of
Biomarkers of Cardiovascular Risk
both men and women indicate that even modest Despite identification of the well-established
activities, such as brisk walking, for as risk factors just described, one out of five

131
Chapter 5

cardiovascular events occurs in patients lacking levels do appear to be at increased risk.


these attributes. In conjunction with growing Recent GWAS and Mendelian randomization
knowledge about the pathogenesis of athero- analyses also support a causal link between
sclerosis, several novel markers of risk have Lp(a) and cardiovascular events.
emerged. These biomarkers serve three pri- Diet and exercise have little impact on
mary roles: (1) as a means to help stratify the Lp(a) levels. Of current lipid-lowering agents,
risk of atherosclerotic disease and thus guide niacin has the greatest effect on Lp(a),
the choice of therapies, (2) as clinical mea- lowering its concentration by as much as
sures to assess treatment success, and (3) as 20%. However, thus far, there is no evidence
potential targets of new therapeutic regimens. that reduction of Lp(a) by drug therapy
improves cardiovascu-lar outcomes.
Homocysteine
C-Reactive Protein and Other
Some studies have shown a significant rela-
Markers of Inflammation
tionship between elevated circulating levels of
the amino acid homocysteine and the in- Because the pathogenesis of atherosclerosis in-
cidence of coronary, cerebral, and peripheral volves inflammation at every stage, markers of
artery disease. The mechanism by which ho- inflammation have undergone evaluation as
mocysteine might increase atherosclerotic risk predictors of cardiac risk. Recall that the pro-
remains undetermined, but current evidence cess of lipoprotein entry and modification in the
suggests that abnormally high levels may pro- vessel wall triggers the release of cytok-ines,
mote oxidative stress, vascular inflammation, followed by leukocyte infiltration, more cytokine
and platelet adhesiveness. Although folic acid release, and smooth muscle migra-tion into—
and other vitamin B supplements reduce high and proliferation within—the in-tima. Involved
serum homocysteine levels, clinical trials have cytokines (e.g., IL-6) mobilize to the liver and
not shown that such therapy reduces athero- incite increased production of acute-phase
sclerotic disease or its complications. reactants, including CRP, fibrino-gen, and
serum amyloid A.
Of these molecules, CRP has shown the
Lipoprotein (a) greatest promise as a marker of low-grade
Lipoprotein (a), referred to as Lp(a) and pro- systemic inflammation associated with athero-
nounced “L-P-little-a,” has been identifi ed as sclerotic disease. Large studies of apparently
an independent risk factor for coronary artery healthy men and women indicate that those with
disease in some studies. Lp(a) is a variant of higher basal CRP levels have a greatly
LDL whose major apolipoprotein (apo B-100) increased risk of adverse cardiovascular out-
links by a disulfi de bridge to another protein, comes, independent of serum cholesterol
apo(a). Apo(a) structurally resembles plas- levels. Recent prospective studies affirm high-
minogen, a plasma protein important in the sensitivity CRP as an independent predictor of
endogenous lysis of fi brin clots (see Chapter myocardial infarction, stroke, peripheral artery
7). Thus, the detrimental effect attributed to disease, and sudden cardiac death. Although it
Lp(a) may relate to competition with normal serves as a marker of risk not captured by
plasminogen activity. Lp(a) is able to enter traditional algorithms, there is no convincing
the arterial intima, and in vitro studies have evidence that CRP itself is actually a mediator
shown that it encourages infl ammation and of atherogenesis.
thrombosis. Recent data support the use of CRP levels
Lp(a) levels in the population are skewed to guide therapy. The JUPITER trial, a study
and not normally distributed, showing a trail- of 17,800 healthy individuals with above-
ing prevalence of the higher levels. As with median levels of CRP but without elevated
homocysteine, not all population studies sup- LDL, dem-onstrated a reduced incidence of
port a link between Lp(a) and cardiovascular major cardio-vascular events among patients
events, though people with the highest Lp(a) who were treated with a statin.

132
Atherosclerosis

Genetics it will continue to be a menace as the popu-


lation ages and as developing countries em-
Genetic predisposition, as refl ected by fam-ily
brace the adverse dietary and activity habits
history, represents a major nonmodifi able risk
of a Western lifestyle. Ongoing research of
factor for atherosclerosis. While directly
the biology of atherosclerosis, as well as
causative genes remain elusive, recent GWAS
advances in therapeutic procedures and
have identifi ed a number of loci associated with
medications, will undoubtedly continue to
atherosclerotic disease. The strongest
further our abili-ties to combat this condition.
connection with CAD and myocardial in-farction
Yet we have not fully capitalized on what we
localizes to chromosome 9p21.3. This region
already know— that much cardiovascular risk
contains genes that code for two cyclin-
is modifi able. Effective control of the risk
dependent kinase inhibitors that are involved in
factors described earlier remains a critical
regulation of the cell cycle and may participate
component to tame this global scourge. It is
in TGF-# inhibitory path-ways. Other
here that the patient– physician relationship
associations with CAD include loci on
and the role of medical professionals as
chromosome 6q25.1, which maps to a gene
community leaders advo-cating healthy
that encodes a mitochondrial C1-tetra-
lifestyles remain of cardinal importance.
hydrofolate synthase involved in methionine
synthesis, and chromosome 2q36.3, a region
devoid of known functional genes. It is too early SUMMARY
to know whether such fi ndings will ultimately
result in enhanced identifi cation, prevention, 1. Early in atherogenesis, injurious and in-
and treatment of atherosclerotic disease. flammatory stimuli activate endothelial and
smooth muscle cells. The resulting
cascade of events recruits immune cells to
the ves-sel wall, fueling a persistent
Infectious Agents in Atherogenesis inflammatory state believed to underlie
Several studies have identifi ed infectious progression of the disease (see Fig. 5.2).
agents (e.g., herpes viruses, Chlamydia 2. Mechanisms that contribute to
pneu-moniae) within some atherosclerotic atherosclero-sis shape the forming plaque
lesions, raising the question of their potential over decades (see Figs. 5.3 through 5.7).
role in atherogenesis. These studies have Plaques can dis-play features associated
generated substantial controversy, and proof with clinical sta-bility or propensity to
of a causal role is lacking. Although it is provoke thrombotic events (so-called
unclear if infec-tions truly play a role in “vulnerable” plaques; see Fig. 5.8).
atherogenesis, viral and microbial products 3. Clinical expression of atherosclerosis com-
could plausibly drive aspects of monly results from narrowing of the vessel
atherogenesis. To date, a number of well- lumen, from calcification or weakening of the
powered trials have not shown that antibiotic arterial wall, or from plaque disruption with
treatment directed against infec-tious agents superimposed thrombus formation. Common
reduces the risk of future car-diac events in manifestations include angina pectoris,
survivors of acute coronary syndromes. myocardial infarction, stroke, and peripheral
arterial disease (see Fig. 5.10).
4. Modifi able risk factors for atherosclerosis
Outlook
include dyslipidemia, smoking, hyperten-
Despite accumulating knowledge of the patho- sion, and diabetes. Nonmodifi able risk
genesis of atherosclerosis and its clinical se- factors include advanced age, male gen-
quelae, this disease remains a major cause of der, and a family history of premature cor-
death in the modern world. Although improve- onary disease. Novel biomarkers, such as
ments in cardiovascular care have reduced CRP, may prove useful in further defi ning
age-adjusted mortality from this condition, risk.

133

You might also like