Skip to main content

ATHEROSCLEROSIS

 1. ATHEROSCLEROSIS

Atherosclerotic diseases, particularly coronary artery disease and stroke, are 

leading causes of death (Mozaffarian et al., 2015). Atherosclerosis is a chronic 

process that progresses in a clinically silent manner for decades. The devas-

tating consequence of lesion evolution is the abrupt formation of thrombi, 

due to lesion rupture or erosion that occludes blood flow, with the attendant 

overt clinical symptoms (Bentzon et al., 2014) (Figure 1). The slow progres-

sion of human atherosclerosis, combined with tissue inaccessibility and the 

absence of effective imaging techniques, provides challenges for characteri-

sation of lesion evolution. Hence, the understanding of mechanisms underly-

ing the evolution of human atherosclerotic lesion progression is incomplete.


FIGURE 1 A section of a human atherosclerotic coronary artery. The lesion contains a large

lipid-rich core, containing cholesterol crystals, derived from apoptotic and/or necrotic foam cells.

A thin fibrous cap, consisting primarily of collagen underlies a compromised endothelial cell layer.

Smooth muscle cells have migrated from the media into the intima where they proliferate and can

become lipid-rich foam cells. Macrophage-derived and smooth muscle cell-derived foam cells

populate the edges of the growing lesion and the fibrous cap. The lesion reduces the artery lumen

diameter. A lesion with a thin fibrous cap covering a large lipid core is prone to rupture and rapid

thrombus formation resulting in an acute coronary event.


As a result, there has been a dependence on information gleaned from ani-

mal models of the disease (Getz and Reardon, 2012). One feature common

to all animal models is the need to induce hypercholesterolaemia for lesion

development, which is achieved by dietary and/or genetic manipulations. The

mechanisms described in this chapter are derived predominantly from stud-

ies in apolipoprotein E (ApoE−/−) or low-density lipoprotein (LDL) receptor

(Ldlr−/−)-deficient mice. While many factors influence the initiation, severity

and nature of lesions in hypercholesterolaemic animals, the basic tenet of this

chapter is that aberrant lipoprotein metabolism is a requirement for athero-

sclerosis development.

Atherosclerosis develops in specific regions of the arterial tree that are

characterised by turbulent blood flow. The initial morphological change is the

subendothelial (intimal) deposition of aggregated lipoproteins (Tabas et al.,

2007). These aggregates promote chemotaxis of leukocytes through several

mechanisms, including oxidation, and enzymatic and nonenzymatic prote-

olysis of apolipoproteins. In addition, monocyte/macrophage adhesion to the

arterial endothelium occurs in the initial stages of experimental atherosclero-

sis, followed by progressive subendothelial accumulation of macrophages in

both experimental and human lesions (Moore and Tabas, 2011). In addition to

recruiting more macrophages, these cells avidly take up native and modified 

ipoproteins resulting in lipid droplets filled predominantly with cholesteryl 

esters (CE), a process known as ‘foam cell’ formation. Macrophage uptake and 

clearance of subendothelial lipoproteins are likely beneficial during the initial 

stages of atherosclerosis.

The resulting dysregulation of lipid metabolism alters macrophage pheno-

type and compromises crucial functions of the cell (Moore et al., 2013). Foam 

cells that accumulate in lesions have a decreased ability to migrate from the 

lesion, which contributes to chronic inflammation and lesion progression. More 

complex plaques involve other immune cells and vascular smooth muscle cells 

(VSMCs). In advanced lesions, macrophages continue as major contributors to 

lesion growth and the inflammatory response through secretion of proinflam-

matory mediators and reactive oxygen and nitrogen species, as well as matrix-

degrading proteases. These processes lead to eventual cell death by either 

necrosis or apoptosis. Dead macrophages release their lipid contents and tissue 

factors, resulting in the formation of a prothrombotic necrotic core, which is a 

critical component of unstable plaques. Instability contributes to the rupture of a 

thin fibrous cap that covers the plaque and leads to subsequent thrombus forma-

tion that underlies an acute myocardial infarction or stroke (Figure 1).

Although many cell types, including monocytes, dendritic cells, lympho-

cytes, eosinophils, mast cells, endothelial cells and smooth muscle cells, contrib-

ute to lesion formation and growth, foam cells are central to the pathophysiology 

of atherosclerosis. Therefore, this chapter will focus on the relationship of 

altered lipid and lipoprotein metabolism to the mechanisms underlying mono-

cyte recruitment, macrophage foam cell formation, the inflammatory response, 

cell death, macrophage emigration and the resolution of inflammation (Moore 

et al., 2013). Initially, we will review critical components and pathways in lipid 

metabolism that contribute to atherosclerosis, followed by a review of emerging 

mechanisms underlying the influence of lipid metabolism in atherosclerosis at 

various stages of lesion development or regression. We also review the current 

development of therapeutic strategies for treatment of atherosclerotic diseases.

2. LIPOPROTEIN TRANSPORT IN ATHEROSCLEROSIS

2.1 Low-Density Lipoprotein

The most atherogenic dyslipidaemia is hypercholesterolaemia, especially 

familial hypercholesterolaemia (FH) (Sahebkar and Watts, 2013a,b). FH is 

characterised by greatly increased plasma concentrations of LDL cholesterol, 

primarily due to downregulation, deficiency or dysfunction of LDL receptors 

(Chapter 17). Pathogenic mutations causing FH frequently occur in the LDL 

receptor gene and less frequently in apolipoprotein B (apoB) and proprotein 

convertase subtilisin/kexin type 9 (PCSK9) genes. As a consequence, plasma 

LDL cholesterol concentrations increase, due to an inability of hepatic LDL 

receptors to clear LDL particles. Overproduction of LDL also contributes to 

elevated LDL. LDL penetrates the subendothelium and accumulates within the subendothelium and accumulates within 


IGURE 2 A schematic of exogenous and endogenous lipoproteins involved in atherogen-

esis. Elevated low-density lipoproteins (LDL) promote atherosclerosis as typified by patients with

familial hypercholesterolaemia. Very-low-density lipoproteins (VLDL), their remnants (IDL) and

chylomicron remnants also amplify atherogenesis. Lp(a) readily enters the arterial intima and pro-

motes atherosclerosis. Low levels of HDL or dysfunctional HDL are unable to promote reverse

cholesterol transport from lesion macrophages, contributing to the formation of foam cells.


the intima of susceptible arteries (Figure 2). LDL then aggregates and/or is

readily oxidised, resulting in an inflammatory response as well as foam cell

formation in both macrophages and smooth muscle cells. This process leads

to premature atherosclerosis in patients with hypercholesterolaemia, particu-

larly those with FH.

2.2 Very-Low-Density Lipoprotein

Another dyslipidaemia that predisposes to premature atherosclerosis is famil-

ial combined hyperlipidaemia (FCH) (Sahebkar and Watts, 2013a). FCH is the

most common form of inherited dyslipidaemia and is genetically heteroge-

neous, resulting from variations in multiple genes involved in the metabolism

and clearance of plasma lipoproteins (Chapter 15). Combined hyperlipidaemia

is often accompanied by additional metabolic abnormalities, including adipose

tissue dysfunction, increased fatty acid flux to the liver and insulin resistance

(Chapter 19). The metabolic phenotype of this dyslipidaemia is hepatic overpro-

duction of very-low-density lipoprotein (VLDL), elevated plasma VLDL tria-

cylglycerol (TAG), apoB and small dense LDL (Figure 2). These lipoproteins

also readily accumulate within the arterial intima, eliciting inflammatory and

other cellular responses linked to atherogenesis. 


Hypertriglyceridaemia is a dyslipidaemia characterised by increased plasma

VLDL TAG concentrations and in the more severe form, elevated chylomicrons

and chylomicron remnants (Hegele et al., 2014) (Chapter 16). Hypertriglyceri-

daemia is associated with increased risk of coronary heart disease. Heritability

accounts for more than 50% of the individual variations in plasma TAG concen-

trations. Genome-wide association studies identified gene variants, including

APOC3, LPL, TRIB1, GCKR and APOA5 that modulate plasma TAG and the

risk for atherosclerosis. Hypertriglyceridaemia results from hepatic overpro-

duction and/or decreased catabolism of TAG-rich lipoproteins, accumulation

of small dense LDL particles and low concentrations of high-density lipopro-

tein (HDL) (Figure 2). This dyslipidaemia is often linked to obesity, metabolic

syndrome and type 2 diabetes (Chapter 19). Although small dense LDL par-

ticles and dysfunctional HDL contribute, atherosclerosis associated with this

dyslipidaemia is primarily due to retention of TAG-rich lipoprotein remnants in

the artery wall. Remnants are retained by intimal extracellular matrix proteins,

and can be oxidised and readily induce foam cell formation. Fatty acids from

these particles readily elicit an inflammatory response and promote lipotoxicity

within cells involved in atherosclerosis.

2.3 Remnants of VLDL and Chylomicrons

The most compelling evidence that remnants of TAG-rich lipoproteins are ath-

erogenic comes from patients with dysbetalipoproteinaemia who have a very

high incidence of premature atherosclerosis (Brahm and Hegele, 2015). In this

rare disorder, remnants of VLDL and chylomicrons accumulate in plasma due

to a genetic variant of APOE (APOE2) bound to the surface of these particles

(Chapter 15). Normally, apoE is the ligand responsible for the clearance of chy-

lomicron remnants and VLDL remnants by hepatic LDL receptors; however,

apoE2 binds poorly to these receptors, resulting in delayed remnant clearance.

When coupled to an overproduction of intestinal or hepatic TAGs (Chapter 16),

remnants attain high plasma concentrations, thereby increasing their exposure

to the arterial intima (Figure 2).

2.4 Lipoprotein(a)

Elevated plasma concentrations of lipoprotein(a) (Lp(a)) are strongly associ-

ated with increased risk of atherosclerosis, although the definitive function of

this lipoprotein is still unclear (Koschinsky and Boffa, 2014) (Figure 2). Lp(a)

is composed of LDL with an additional single molecule of apo(a) that is bound

covalently to apoB-100 (Chapter 16). Plasma Lp(a) concentrations are not mod-

ulated by diet, age, sex or physical activity, but are largely determined by varia-

tion at the LPA gene locus, making elevated Lp(a) highly heritable.

The apo(a) gene contains a variable number of tandem repeats that encode

a kringle moiety identical to kringle IV of plasminogen. This leads to Lp(a) 

isoforms that differ in molecular mass. There is a strong and inverse association 

between plasma Lp(a) concentration and molecular mass of apo(a). Smaller apo(a) 

isoforms possess greater atherogenic potential. Similar to LDL, Lp(a) is retained 

within the arterial intima through extracellular matrix interactions, and induces 

chemokine secretion, inflammation and foam cell formation. Plasma Lp(a) carries 

oxidised phospholipids, and its atherogenicity may be associated with delivery 

of these proinflammatory lipids to lesions. Lp(a) can also induce apoptosis in 

endoplasmic reticulum (ER)-stressed macrophages, thereby contributing to lesion 

vulnerability and thrombosis. In addition, apo(a) homology to plasminogen may 

inhibit the fibrinolytic pathway, making Lp(a) a thrombogenic lipoprotein.

2.5 High-Density Lipoprotein

HDL cholesterol concentrations are inversely related to the risk of cardiovas-

cular disease and death (Luscher et al., 2014). However, there is increasing 

awareness that HDL function plays a more important role in protection from 

atherosclerotic disease, a feature not captured in measures of plasma HDL con-

centration. Many dyslipidaemias are associated with low plasma concentrations 

and/or dysfunctional HDL. Causes of low HDL cholesterol are not well under-

stood, but involve enhanced catabolism of the cholesteryl ester-depleted HDL 

particle (Chapter 15).

HDL possesses anti-inflammatory and antioxidant properties, but its prin-

cipal antiatherogenic function is due to its ability to mediate cholesterol export 

from cells involved in atherosclerosis and subsequent elimination of HDL 

cholesterol through reverse cholesterol transport (Chapter 15) (Figure 2). In 

experimental models of atherosclerosis, promotion of macrophage cholesterol 

export attenuates progression or induces lesion regression. Other functions of 

HDL impact atherogenesis. HDL-associated paraoxanase-1 (PON-1) normally 

prevents HDL from oxidative modification. Reduced levels of HDL-associated 

PON-1 activity lead to the generation of modified HDL that inhibits endothelial 

cell nitric oxide synthase (eNOS) activation, thereby losing its anti-inflammatory 

properties. These HDL are also defective in cholesterol export.

3. LIPOPROTEIN RECEPTORS AND LIPID TRANSPORTERS

Lipoproteins are complex macromolecules that are recognised by a variety of 

receptors (Chapter 17) and function to facilitate physiological and pathological 

processes. There is direct in vivo evidence for a role of lipoprotein receptors in 

the development of atherosclerosis as summarised in Table 1.

3.1 LDL Receptors

LDL receptors were defined by the classic studies of Goldstein and Brown 

in which they described a process for transporting large lipoprotein particles nsporting large lipoprotein particles


(∼24nm) across the cell membrane. The first step in this process is the interac-

tion of the apoB of LDL with the cysteine-rich receptor-binding domain of LDL

receptors. Engagement of apoB with LDL receptors results in endocytosis of the

entire LDL particle with subsequent delivery to the lysosome where the particle

is degraded to its components. Delivery of free (unesterified) cholesterol to the

ER induces homeostatic mechanisms that inhibit continuous exogenous delivery

or endogenous cholesterol synthesis (Chapters 17 and 11). Deficiency of LDL

receptors increases plasma cholesterol and accelerates atherosclerosis. Humans

with homozygous FH develop severe atherosclerotic disease within two decades

of life if untreated. Deficiency of LDL receptors in rabbits also leads to pro-

nounced hypercholesterolaemia and accelerated atherosclerosis. Genetic deple-

tion of LDL receptors in mice leads to a modest hypercholesterolaemia, which is

greatly augmented by feeding diets that are enriched in saturated fat and/or cho-

lesterol. LDL receptors can also be negatively regulated by PCSK9. Expression


of gain-of-function PCSK9 mutants leads to depletion of LDL receptors, hyper-

cholesterolaemia and atherosclerosis (Dadu and Ballantyne, 2014).

3.2 Scavenger Receptors

LDL receptors are downregulated by increased cellular cholesterol to pre-

vent sterol engorgement, which is a characteristic of cells in atherosclerotic

lesions. Therefore, it is unlikely that LDL promotes atherosclerosis through

this receptor. As an alternative, it was proposed that in hypercholesterolaemic

states, an increased residence time of LDL particles in the subendothelial

space leads to modifications, resulting in enhancement of macrophage uptake

that is not regulated by cellular sterol content. Acetylation of LDL was the

first modification identified that enhanced uptake in macrophages. Binding of

acetylated LDL to macrophages is saturable, consistent with a receptor-mediated

interaction. These recognitions led to the concept that hypercholesterolae-

mia promotes atherosclerosis through modified LDL uptake by ‘scavenger’

receptors (Chapter 17).

The subsequent effort to identify scavenger receptors led to the cloning of

a trimeric membrane glycoprotein that was expressed as two major splice vari-

ants. This receptor is now referred to as scavenger receptor class A or SR-A

(Canton et al., 2013). Although expression of the cloned protein demonstrated

the ability of acetylated LDL to bind to SR-A in cultured cells, studies in which

SR-A was genetically manipulated in atherosclerosis-susceptible mice demon-

strated a full spectrum of effects on lesion size from being increased, unaffected

or decreased (Canton et al., 2013) (Table 1). SR-A is now recognised as the

receptor for a wide array of ligands that influence atherosclerosis, and is also an

integrin-independent adhesion molecule (Chapter 17). Therefore, it is unclear

whether the multiple ligands of SR-A contribute to the variable effects observed

on atherosclerotic lesion formation.

There are now a large number of other receptors that are collectively classed

as scavenger receptors. Most of these receptors bind to modified LDL, in which

acetylation or oxidation is the most common modification. In addition to SR-A,

the major members of this class that have been studied for effects on experimen-

tal atherosclerosis are CD36, SR-B1 and LOX1 (Canton et al., 2013) (Chapter

17). CD36 was originally described as a fatty acid transporter and was subse-

quently identified as a receptor for copper-oxidised LDL. It is highly expressed

in cultured macrophages and atherosclerotic lesions. Like SR-A, the data on

the role of CD36 in atherosclerosis in either ApoE−/− or Ldlr−/− mice have been

inconsistent (Table 1).

SR-B1 was identified as a structural variant of CD36 with similar binding

characteristics for acetylated LDL. While many of the same ligands bind both

CD36 and SR-B1, LDL only binds to SR-B1. It was subsequently demonstrated

that SR-B1 is an HDL receptor that facilitates bidirectional movement of cho-

lesterol between cells and HDL particles (Canton et al., 2013) (Chapter 15).

Mice with deficiencies of both SR-B1 and apoE exhibit an array of cardiovas-

cular pathologies at an early age, including accelerated atherosclerosis. Lesions

in these mice are present in coronary arteries, which is uncommon in most small

animal models of atherosclerosis. Overexpression of SR-B1 in Ldlr−/− mice

decreases atherosclerosis with concomitant decreases in plasma HDL choles-

terol concentrations. Deleting SR-B1 increases atherosclerosis in Ldlr−/− mice

with increases in plasma cholesterol concentrations and very large HDL par-

ticles (Table 1). The effects of SR-B1 on plasma lipoprotein concentrations and

characteristics do not appear to affect atherosclerosis since deletion of SR-B1

in bone marrow-derived cells augments lesion size in mice, without discernible

effects on plasma cholesterol.

Although macrophages have been the major focus of expression for most

scavenger receptors, endothelial dysfunction is also crucial in atherosclerotic

lesions. A screen of an endothelial cell cDNA library identified a protein des-

ignated as lectin-like oxidised LDL (oxLDL) receptor or LOX-1 that binds

oxLDL (Chapter 17). LOX-1 is also expressed in macrophages and smooth

muscle cells of atherosclerotic lesions. LOX1 deficiency reduces atherosclero-

sis, whereas its overexpression in endothelial cells increases atherosclerosis in

hypercholesterolaemic mice, suggesting that this protein contributes to lesion

development (Table 1).

3.3 Low-Density Lipoprotein Receptor-Related Protein

LDL receptor-related protein (LRP1), a multifunctional protein in the LDL recep-

tor superfamily, is present on the surface of multiple cell types (Chapter 17). Since

its discovery by Herz and colleagues in 1988, more than 40 ligands have been

identified to interact with LRP1, implicating complex functions of this protein

(Chapter 17). This large protein contains a 515-kDa α chain (amino-terminal part)

and an 85-kDa β chain (membrane-bound) carboxyl terminal fragment. Deficiency

of LRP1 in macrophages reduces VLDL uptake, and enhances the secretion of

inflammatory mediators including monocyte chemoattractant protein-1 (MCP-1,

CCL2), interleukin (IL)-1, IL-6 and TNFα. Deficiency of LRP1 in macrophages

does not affect plasma cholesterol concentrations or lipoprotein–cholesterol

profiles, but augments atherosclerosis in hypercholesterolaemic mice (Table 1).

In addition to macrophages, LRP1 is abundant in VSMCs. Deficiency of LRP1

in VSMCs augments atherosclerosis in hypercholesterolaemic mice (Gonias and

Campana, 2014).

3.4 ATP-Binding Cassette Subfamily (ABCs)

Cells normally respond to excessive lipid accumulation by upregulation of path-

ways that promote export of cholesterol and other lipids (Chapter 15). Although

passive diffusion of cholesterol from the plasma membrane occurs, macrophage

foam cells possess several transporters that export lipids, including members 

of the ATP-binding cassette subfamily (ABCs) ABCA1 and ABCG1. ABCA1

facilitates cholesterol export to lipid-poor apoA-I within pre-β-HDL, whereas

ABCG1 facilitates export to mature HDL particles (Figure 2). Macrophage

expression of ABCA1 and ABCG1 is upregulated in response to increased cel-

lular cholesterol concentrations by liver X receptors (LXR). This ligand-acti-

vated nuclear receptor acts as a sterol sensor and is transcriptionally activated by

metabolites of lipoprotein-derived cholesterol, including the oxysterols 27-OH-

cholesterol, 24-OH-cholesterol, 7-keto-cholesterol and desmosterol, a choles-

terol precursor. All of these metabolites accumulate in macrophage foam cells.

Activation of LXR also inhibits inflammatory responses. In experimental mod-

els, strategies that increase activation or expression of LXR or ABC transport-

ers attenuate atherosclerosis, whereas inhibition of their expression promotes

atherosclerosis.

4. CONTRIBUTIONS OF LIPOPROTEIN-MEDIATED

INFLAMMATION TO ATHEROSCLEROSIS

Multiple cell types are involved in the development of atherosclerosis. One

well-recognised mechanistic theory is that endothelial dysfunction of the arte-

rial wall is the initial step in provoking monocyte adhesion, followed by mac-

rophage infiltration into the subendothelial space (intima) to form lipid-laden

foam cells. Resident cells of the arterial wall together with leukocytes recruited

from peripheral blood orchestrate a series of inflammatory responses in the arte-

rial wall, thereby promoting the progression of atherosclerosis. The ‘response-

to-retention’ hypothesis (Tabas et al., 2007) proposes that lipoproteins normally

diffuse across the endothelial barrier, become trapped in the subendothelium,

stimulate inflammatory responses and promote accumulation of lipid-laden

macrophages. It is generally accepted that accumulated lipids within macro-

phages are derived from plasma lipoproteins through uptake by endocytic,

phagocytic, receptor-involved or nonreceptor-mediated processes. This section

updates the current knowledge regarding lipid and lipoprotein-stimulated cellu-

lar changes and cell-specific manipulations of components that have been stud-

ied in atherosclerotic animal models.

4.1 Cell Types Involved in Atherosclerotic Lesions

Associations between inflammation and atherosclerosis are complex, they

involve multiple cell types and a network of signalling pathways. A subtle

change in this complex system may lead to substantial changes in a spectrum

of signalling pathways. Hypercholesterolaemia induces leukocytosis (Murphy

et al., 2014), thereby increasing their likelihood of recruitment into the suben-

dothelial space. Modified lipoproteins can activate endothelial cells to secrete

adhesion molecules such as vascular cell adhesion molecule 1 (VCAM-1),

intracellular adhesion molecule 1 (ICAM-1) and selectins. These molecules, together with chemoattractant mediators including complement factors and

MCP-1, promote monocyte adhesion to the impaired endothelial barrier and

emigration into the intima. Monocytes become activated to macrophages, which

take up native and modified lipoproteins to form lipid-laden foam cells. Among

all leukocyte types, macrophages are the predominant cell type in atheroscle-

rosis and are major contributors to the development of lesions. Therefore, the

contributions of this cell type to atherosclerosis will be reviewed in detail in

Section 4.2. Lipoproteins accumulating in the intimal space (intima) and their

uptake by macrophages contribute to the secretion of multiple proatherogenic

cytokines such as IL-1, IL-6 and TNFα (Figure 3). FIGURE 3 Pathways for monocyte recruitment and macrophage retention, foam cell forma-

tion, apoptosis, lipid export and emigration in plaques. GR1+LY6Chi monocytes are recruited

to mouse atherosclerotic lesions using chemokine–chemokine receptor pairs (CCR2:CCL2 and

CCR5:CCL5) to infiltrate the intima, which is facilitated by endothelial adhesion molecules,

including P-selectin and P-selectin glycoprotein ligand 1 (PSGL1), intercellular adhesion molecule

1 (ICAM1) and vascular adhesion molecule 1 (VCAM1). Recruited monocytes differentiate into

macrophages in the intima, where they interact with atherogenic lipoproteins (see text) resulting

in foam cell formation. Foam cells secrete proinflammatory cytokines, chemokines and retention

factors that induce macrophage proliferation, promote foam cell accumulation within lesions and

amplify the inflammatory response. The inflammatory milieu and excess lipid accumulation induce

macrophage ER stress and apoptosis. Cell death, together with defective efferocytosis, results in

the formation of a necrotic core, which is characteristic of advanced lesions. The promotion of

lipid unloading by ABCA1 and cholesterol export to lipid-poor apoA-I reverses cellular lipid accu-

mulation and induces plaque regression. Events that stimulate efferocytosis lead to reverse trans-

migration to the lumen and macrophage emigration to adventitial lymphatics. LFA1, lymphocyte

function-associated antigen 1; VLA4, very late antigen 4. Adapted from Moore et al. (2013). In addition to macrophages, T lymphocytes are present in atherosclerotic

lesions and, like macrophages, accumulate in early lesions (Libby, 2012). LDL

stimulates T lymphocytes to produce interferon-γ (IFN-γ), which contributes

to further recruitment of T lymphocytes. In contrast to T lymphocytes, athero-

sclerotic lesions contain minimal B lymphocytes. oxLDL stimulates antibody

production from B lymphocytes that accumulate in atherosclerotic lesions. It

is generally accepted that T lymphocytes augment atherosclerosis, whereas

antibodies produced by a subclass of B lymphocytes, termed B1 cells, protect

against formation and development of atherosclerosis.

Neutrophils are the most numerous leukocyte type in blood. This cell type

is present in blood for only 1–2days until activated, which promotes migration

into tissues. In the innate immune system, neutrophils are the first line in acute

inflammatory responses. The role of neutrophils in atherosclerosis has not been

appreciated until recently. Neutrophils become activated and are recruited to

sites of atherosclerotic lesions, but the mechanisms by which neutrophils con-

tribute to atherosclerosis are unclear (Weber and Noels, 2011).

VSMCs are the predominant cell type in the normal arterial medial layer.

Vascular inflammation leads to VSMC proliferation, and their migration into

atherosclerotic lesions. Recent studies revealed that progenitor cells present in

the adventitia of the arterial wall have the capability to become VSMCs and

contribute to the development of atherosclerosis. VSMCs secrete extracellular

matrix that stabilises lesions through the formation of a fibrous cap. VSMCs

also have the capacity to transform into foam cells by accumulating lipoprotein-

derived lipids; however, VSMC foam cells lose their capacity to secrete extra-

cellular matrix. Lipoproteins, such as oxLDL, induce the proliferation of

smooth muscle cells. In human coronary lesions, recent studies found that over

50% of foam cells were derived from smooth muscle cells, and these cells

expressed the macrophage marker CD68 (Allahverdian et al., 2014). These

discoveries have made research on vascular smooth muscle-related mecha-

nisms of atherosclerosis more challenging (Tellides and Pober, 2015; Zhang

and Xu, 2014).

4.2 Foam Cells

4.2.1 Recruitment of Circulating Monocytes

Activation of endothelium promotes recruitment of circulating monocytes

(Moore et al., 2013) (Figure 3). Of significance, hypercholesterolaemia is

associated with increased numbers of circulating monocytes. Their precursors,

hematopoietic stem and progenitor cells, become enriched in cholesterol and

deficient in cholesterol export, leading to cell proliferation. Circulating mono-

cytes in mice consist of at least two major subsets, Ly6Chi and Ly6Clow mono-

cytes. Ly6Chi cells constitute the majority of cells recruited to the intima, and

represent precursors of M1 classically activated macrophages and thus are key

participants in foam cell formation and inflammatory responses  Recruitment of circulating monocytes into atherosclerotic lesions requires

integration of at least three discrete processes. These are capture, rolling and

transmigration and each step is regulated by several molecular factors (Figure 3).

Molecules on the surface of activated endothelial cells mediate capture and

rolling of monocytes. This depends on their immobilisation by endothelial cell

chemokines, P-selectin, VCAM1 and ICAM1, which interact with chemokine

receptors and integrins on monocytes. Extravasation of monocytes across the

endothelium and into plaques is mediated by the interaction of monocyte che-

mokine receptors with the chemokines PECAM1 or VCAM1 that are secreted by

endothelial cells, macrophages and smooth muscle cells. In addition, neuronal

guidance cues expressed by endothelial cells, including ephrin B2, netrin 1 and

semaphorin 3A, can increase monocyte recruitment or inhibit the chemokine-

directed monocyte migration (Figure 3).

4.2.2 Foam Cell Formation

Macrophage foam cell formation represents one of the earliest stages of lesion

development, and continues throughout lesion evolution (Moore and Tabas,

2011) (Figure 4). Although macrophages can take up apoB-containing lipopro-

teins through the LDL receptor, this receptor is downregulated during foam

cell formation by increased cellular cholesterol. Modified lipoprotein uptake by

scavenger receptors, which is not regulated by intracellular cholesterol, contrib-

utes to foam cell formation. Multiple means of lipoprotein modification have

been described; however, the relevant pathways that promote foam cell forma-

tion in vivo remain to be fully elucidated.

Oxidative stress in the arterial intima promotes lipoprotein modification that

generates ‘damage’ signals that are recognised by scavenger receptors on macro-

phages and other cells of the innate immune system. oxLDL has been identified

in both human and mouse lesions and antibodies that recognise oxidation-

specific epitopes of LDL have also been discovered. Mediators of lipopro-

tein oxidation, including 12/15-lipoxygenase, myeloperoxidase, free radicals

including superoxide, hydrogen peroxide and nitric oxide, have been identified

in the artery wall. Lipoproteins modified through these mechanisms are readily

endocytosed by scavenger receptors in vitro (Figure 4). Scavenger receptors,

including SR-A1 (encoded by MSR1), macrophage receptor with collagenous

structure (MARCO; also known as SR-A2), CD36 (also known as platelet gly-

coprotein 4), SR-B1, LOX1, scavenger receptor expressed by endothelial cells

1 (SREC1) and scavenger receptor for phosphatidylserine and oxidised LDL

(SR-PSOX; also known as CXCL16) all can bind oxidised lipoproteins and pro-

mote unimpeded foam cell formation (Section 3.2). SR-A1 and CD36 mediate

the majority of uptake of oxidised lipoproteins by macrophages in vitro. These

receptors internalise lipoproteins into late endosomes and lysosomes, where

lysosomal acid lipase mediates the hydrolysis of lipoprotein CE to cholesterol

and fatty acids (Figure 4). Endolysosomal cholesterol is trafficked to the ER

or plasma membrane by NPC1 and NPC2. In the ER, cholesterol undergoes. FIGURE 4 Mechanisms controlling macrophage lipoprotein uptake, lipid export and intra-

cellular cholesterol trafficking. Macrophages internalise native LDL, VLDL and oxidised lipo-

proteins within the lesion by scavenger receptors outlined in Table 1. Internalised lipoproteins and

their lipids are digested in the lysosome. Cholesterol is transferred to the plasma membrane for

export from the cell or to the endoplasmic reticulum for esterified by acyl-coenzyme A:cholesterol

acyltransferase 1 (ACAT1) and ultimately stored as CE in cytosolic lipid droplets. Stored lipids

are released for export by either neutral cholesteryl ester hydrolase 1 (NCEH1)-mediated lipolysis

or via lipophagy (not shown). Cellular cholesterol accumulation increases oxysterol synthesis and

activation of LXR–retinoid X receptor (RXR) transcription factor complex that upregulates expres-

sion of ABCA1 and ABCG1. These transporters transfer cholesterol to lipid-poor apoA-I to form

nascent HDL or to lipidate mature HDL particles. Exported cholesterol is esterified by lecithin

cholesterol acyltransferase (LCAT). Excessive cellular cholesterol can interfere with the function of

the ER as well as induce cholesterol crystal formation in the lysosome, resulting in activation of the

inflammasome. If prolonged, this results in cell death by apoptosis. Increased cholesterol content of

membrane-associated lipid rafts increases proinflammatory Toll-like receptor 4 (TLR4) signalling

that activates nuclear factor-κB (NF-κB), resulting in the production of proinflammatory cytokines

and chemokines. Adapted from Moore et al. (2013). reesterification by acyl-CoA:cholesterol acyltransferase 1 (ACAT1) to CE that

are stored within neutral lipid droplets that appear ‘foamy’ under the micro-

scope (Figure 4).

Although ApoE−/− mice deficient in both SR-A1 and CD36 have reduced

lesion inflammation, macrophage apoptosis and plaque necrosis, loss of these

receptors did not reduce foam cell formation or lesion size (Moore et al., 2005).

Together with the results of human clinical trials of antioxidant vitamins E and

C that failed to show a reduction of cardiovascular events, these studies have

prompted the field to consider alternative mechanisms for foam cell formation. Proteases and lipases within the arterial intima also mediate lipoprotein modi-

fications, particularly LDL aggregation (Moore et al., 2013). Extracellular matrix

glycoproteins within the intima contribute to this process by retaining apoB-con-

taining lipoproteins and by modulating the activity of enzymes, including group

IIA secretory phospholipase A2 (PLA2G2A), PLA2G5 and PLA2G10, as well as

secretory sphingomyelinase. These lipolytic enzymes produce modified forms of

LDL that are taken up by macrophages independent of scavenger receptors. Evi-

dence from mouse studies supports a function for PLA2 in atherosclerosis progres-

sion, and circulating PLA2 levels in humans correlate with risk for atherosclerosis.

Although foam cell formation by native apoB-containing lipoproteins was not orig-

inally considered a major pathway, recent studies have documented macrophage

uptake of LDL by pinocytosis in vitro and in the arterial intima leading to foam cell

formation (Figure 4). This receptor-independent endocytic pathway also delivers

cholesterol to the endolysosomal compartment and stimulates ACAT1-mediated

cholesterol esterification. Thus, in vivo, it is likely that atherogenic LDL and other

apoB-containing lipoproteins induce foam cell formation by multiple pathways.

Excessive uptake of lipoproteins eventually results in defective lipid metab-

olism within macrophages, ultimately leading to increased lesion complexity.

When retained within lipid droplets, CE is metabolically inert, whereas cho-

lesterol within cell membranes can be toxic. Enrichment of macrophage ER

membranes with cholesterol, which occurs with excessive lipoprotein uptake,

eventually suppresses its esterification by ACAT1, resulting in further cholesterol

accumulation. Furthermore, plasma membranes become cholesterol enriched,

leading to amplified inflammatory signalling through Toll-like receptors (TLR;

see Section 4.6) (Figure 4). In addition, trafficking of cholesterol from lyso-

somes becomes compromised, which inhibits macrophage cholesterol export

and further enhances the inflammatory response. This dysregulated lipid metab-

olism contributes to macrophage ER stress, and if prolonged, can ultimately

lead to apoptosis and cell death (Figure 4). Efficient clearance of apoptotic cells

from lesions by surrounding macrophages (known as efferocytosis) requires the

engulfing cells to metabolise lipids derived from the apoptotic cells (Figure 3).

Therefore, excess lipoprotein uptake by macrophages compromises cellular lipid

metabolism, promotes apoptosis and suppresses efferocytosis, which leads to

secondary necrosis and a necrotic core (Figure 4). A necrotic core, together with

a thinned fibrous cap, is characteristic of plaques more vulnerable to rupture.

4.3 Macrophage Polarisation

On entry into the arterial intima, monocytes are polarised to M1 macrophages,

known as classically activated inflammatory cells, or M2 macrophages, known

as alternately activated cells that resolve inflammation (Moore et al., 2013). It

is thought that M1 macrophages are derived from Ly6Chi monocytes, whereas

M2 macrophages are from Lys6Clow monocyte precursors. In vivo, it is likely

that macrophage phenotype is more complex. Furthermore, the factors in lesion 

microenvironments that promote macrophage polarisation remain incompletely

defined. M1 macrophages represent foam cell precursors and in human plaques

they are enriched in lipids and localised to areas distinct from the less inflam-

matory M2 macrophages. In mouse models, M1 macrophages become predomi-

nant in complex lesions, whereas enrichment of M2 macrophages occurs in

lesions in which regression of atherosclerosis has been induced. Administration

of the M2-polarising cytokine IL-13 to Ldlr−/− mice drives lesion macrophages

to M2-like cells and inhibits lesion progression. Recent evidence indicates that

oxLDL induces a distinct macrophage phenotype that has been termed Mox,

characterised by the increased expression of NRF2, although the function of

these cells in atherosclerosis has not been defined.

4.4 Inflammatory Responses

Innate immune activation is considered a central feature of the pathogenesis of

atherosclerosis and is largely a consequence of dysregulated lipid metabolism

within developing lesions (Moore et al., 2013). Lipids, oxidised lipids and other

ligands that accumulate within lesions trigger macrophage receptors includ-

ing scavenger receptors, TLRs and nucleotide-binding oligomerisation domain

(NOD)-like receptors that in turn activate inflammatory responses (Figure 4).

Cholesterol crystals are detected in extracellular spaces and within macro-

phages in both early and more advanced lesions. Cholesterol crystals induce the

NLRP3 inflammasome, which leads to the processing and secretion of several

proinflammatory cytokines (Figure 4). The exposure of macrophages to fatty

acids derived from VLDL or VLDL remnants also activates the expression of

proinflammatory cytokines, independent of TLRs.

TLRs are a class of membrane-spanning, noncatalytic receptors that play

critical roles in innate immune responses. Multiple ligands, including oxLDL

and oxidised phospholipids, activate TLR2- and/or TLR4-related signalling

cascades to promote inflammation. In the presence of lipid ligands, CD36

and SRA interact with TLRs, resulting in activation of myeloid differentiation

factor (MyD) 88 and c-Jun N-terminal kinases (JNK), which promote apoptosis

(Figure 4). Their role in atherosclerosis is supported by studies in ApoE−/− mice

and Ldlr−/− mice, whereby genetic deficiency of TLR2, TLR 4 or the TLR

adapter protein MyD88 reduces lesion development. However, despite consistent

effects of global deficiency of TLRs and MyD88 in reducing atherosclerosis in

mice, their absence in bone marrow-derived cells has been conflicting. These

studies suggest that the primary impact of TLR signalling on atherosclerosis is

not through hematopoietic cells, including macrophages.

4.5 Atherosclerotic Lesion Macrophage Retention and Emigration

Atherogenesis is related to macrophage accumulation within lesions that

is determined by monocyte recruitment, macrophage proliferation, foam

 cell formation and macrophage emigration and death (Moore et al., 2013)

(Figure 3). Atherogenic lipoproteins in the intima are primary determinants

of macrophage recruitment. Macrophage emigration occurs in early athero-

sclerotic plaques, but the rate of egress decreases with lesion progression.

In mouse models, emigration is an important factor linked to attenuation

of progression or the induction of lesion regression. Lesional macrophages

are subject to both retention and emigration signals. Macrophage foam cells

exhibit increased expression of the neuro-immune guidance cues, netrin 1 and

semaphorin 3E, both of which induce macrophage chemostasis (Figure 3).

Deficiency of netrin 1 attenuates lesion progression and increases mac-

rophage emigration. The signals that guide macrophages to exit lesions,

by transmigration through the endothelium to the lumen or by migrating

through the media to the adventitial lymphatics, are not well defined, but

likely involve macrophage expression of the CC-chemokine receptor CCR7

and the CC-chemokine ligands CCL19 and CCL21 that regulate cell homing

the lymph. As lesions progress, the continued presence of foam cells in a

lipid-rich lesion environment leads to cholesterol- and saturated fatty acid-

induced cytotoxicity, and ER stress, thereby increasing apoptosis (Figure 4).

Efferocytosis, the ability of macrophages to clear apoptotic cells through

receptors including tyrosine protein kinase MER (MERTK) and LRP1,

becomes compromised by cholesterol accumulation in the engulfing mac-

rophage. Defective efferocytosis contributes to secondary necrosis as well

as to formation and expansion of lipid-rich necrotic cores, which, in turn,

contribute to lesion rupture.

4.6 Atherosclerotic Lesion Regression

With the exception of early lesions, which are dominated by foam cells,

atherosclerosis was originally considered irreversible (Moore et al., 2013).

Recent studies demonstrating macrophage emigration from plaques in mice,

and that inflammation-resolving and tissue-remodelling M2 macrophages

are present in human and animal lesions, suggest that regression in humans

may be achievable. Some of the determinants of lesion regression have

recently emerged (Figure 3). Common to all mouse models of regression is

the requirement for aggressive lowering of plasma lipids and the observation

that within the regressing lesion there is a marked decrease in macrophage

number. Of those remaining, a majority display an M2 phenotype. In mac-

rophages from regressing lesions, the expression of genes including netrin

1, semaphorin 3E and members of the cadherin family is downregulated,

whereas CCR7 and cell motility factors are upregulated, thereby enhancing

the migratory ability of macrophages. Although other key factors that pro-

mote regression remain to be identified, improved lipid metabolism, includ-

ing marked attenuation of foam cell formation, is critical for regression to

be achieved. 5. NEW EMERGING MECHANISMS OF LIPID METABOLISM

INFLUENCING ATHEROSCLEROSIS

5.1 MicroRNAs

MicroRNAs (miRs) are a class of small noncoding RNAs that participate in a

diversity of biological and pathophysiological processes through posttranscrip-

tional regulation of gene expression. Many miRs regulate lipoprotein metabo-

lism in the circulation and/or local tissues (Novak et al., 2014). A major focus

of recent research is regulation of HDL metabolism by miRs. HDL particles are

carriers of miRs in plasma. Conversely, miRs also regulate expression of many

genes associated with HDL biosynthesis, uptake and metabolism.

miR-33 is the most intensely studied miR that regulates HDL metabolism.

miR-33 is embedded in intron 16 of the sterol regulatory element-binding fac-

tor (SREBP) gene, a transcription factor involved in cholesterol homeostasis

(Chapter 11). Although many studies reported that inhibition of miR-33 by anti-

sense oligonucleotides (ASO) in mice and monkeys or genetic deficiency of miR-

33 in mice increased plasma HDL cholesterol concentrations (Figure 5), other

studies have demonstrated that inhibition of miR-33 did not increase, or only

increased transiently, plasma HDL cholesterol concentrations. As with conflict-

ing findings in regulation of HDL cholesterol, effects of miR-33 on atherosclero-

sis are also inconsistent. In hypercholesterolaemic mice, an ASO against miR-33

or its genetic deficiency prevented, or had no effect on, the development of ath-

erosclerosis, or regressed preexisting atherosclerosis. These conflicting findings

admonish further studies before regulation of miR-33 can be used in human

studies. In addition to conflicting findings in mouse models, multiple differences

between mice and humans warrant careful interpretation. For example, miR-33

has differential effects in mice and humans on the expression of Niemann–Pick

C1 (NPC1) and ABCG1. These differences may be partially attributed to the

finding that mice only have miR-33a, whereas humans and primates have both

miR-33a and miR-33b (Fernandez-Hernando and Moore, 2011; Naar, 2013).

Several other miRs have also been shown to regulate lipoprotein metabo-

lism. We provide a brief review of those that have been studied in atherosclerotic

animal models. HDL carries more miRs than LDL particles. miR-155 is one of

the few exceptions and exhibits higher abundance in LDL than in HDL. miR-

155 regulates inflammatory responses. In addition, enhancement of miR-155

increases oxLDL uptake into cultured macrophages and impairs macrophage

cholesterol export to apoA-I, and suppression of miR-155 leads to increases

of cholesterol export to apoA-I in the absence of changes in the expression of

ABCA1 and ABCG1. Inhibition of miR-155 by an antagonist of a microRNA

(antagomiR) system reduced atherosclerotic lesion size in ApoE−/− mice fed a

high-fat, high-cholesterol diet without affecting plasma total cholesterol con-

centrations. miR-155 deficiency in bone marrow-derived cells reduced athero-

sclerotic lesions in ApoE−/− mice, but augmented atherosclerotic lesion size in

Ldlr−/− mice (Nazari-Jahantigh et al., 2015). It is unclear whether miR-155 has FIGURE 5 Existing and potential therapeutic targets for the treatment of lipid metabolism

and prevention or treatment of atherosclerosis. The therapeutic objective is to decrease the con-

centration of atherogenic lipoproteins or increase antiatherogenic lipoproteins in the circulation,

thereby improving lesion pathology or plaque regression. Statins inhibit the hepatic enzyme HMG-

CoA reductase, leading to increased expression of the LDLR and stimulation of the hepatic clearance

of LDL and VLDL remnants. Cholesterol-absorption inhibitors (ezetimibe) reduce the absorption

of cholesterol from the intestine by inhibiting the transporter NPC1L1 and are very effective when

combined with statins. Inhibition of PCSK9 with injected monoclonal antibodies blocks LDLR deg-

radation and increases hepatic cell surface LDLR, which results in an increased hepatic clearance of

LDL, VLDL remnants and Lp(a). Inhibition of microsomal TAG transfer protein (MTP) decreases

the assembly of VLDL in the liver and chylomicrons in the intestine by blocking the transfer of TAG

and CE to apoB-100 or apoB-48. CETP inhibitors increase HDL cholesterol and decrease LDL

cholesterol levels as well as Lp(a). CETP normally transfers CE from HDL to VLDL, IDL and LDL,

where theoretically this can promote the atherogenicity of these lipoproteins. ApoB antisense oligo-

nucleotides (ASO) inhibit the translation and synthesis of apoB and therefore decrease the secretion

of apoB-containing lipoproteins (VLDL) into plasma. An apoC-III ASO inhibits the synthesis and

secretion of apoC-III, a protein that normally delays the catabolism of TAG-rich lipoproteins. An

apo(a) ASO inhibits the hepatic synthesis of apo(a), therefore inhibiting the formation of Lp(a).

Inhibition of ACAT-2 with ASOs in mice inhibits the secretion of both chylomicrons and VLDL into

plasma and attenuates atherosclerosis in mice. Specific DGAT-2 inhibitors decrease hepatic VLDL

secretion and LDL cholesterol in experimental animals and are currently under development for

clinical use. MicroRNA-33 (miR-33) inhibits the expression of ABCA1 and ABCG1 and decreases

HDL formation. miR-33 ASOs or anti-miR-33 in mice increases cellular ABCA1 expression and

plasma HDL cholesterol concentrations and attenuate atherosclerosis in mice. differential effects between Ldlr−/− mice and ApoE−/− mice, leading to different

contributions of miR-155 to atherosclerosis in these two mouse strains.

miR-30c targets the 3′ untranslated region of microsomal triacylglyceride

transfer protein (MTP) mRNA. In addition to reducing MTP activity and apoB

secretion, miR-30c diminishes lipid synthesis in an MTP-independent manner. Lentiviral transduction of miR-30c reduced, and anti-miR-30c increased, plasma

cholesterol concentrations in C57BL/6 or ApoE−/− mice fed a Western diet, an

effect attributed to changes in secretion of plasma apoB-containing lipoproteins.

Consistent with modulation of plasma apoB-containing lipoproteins, transduc-

tion of miR-30c in female ApoE−/− mice fed a Western diet attenuated athero-

sclerosis, whereas inhibition of this miR augmented lesion development.

Given their multitargeting features, some circulating miRs might serve as

biomarkers to predict the development of atherosclerosis, and inhibiting or

enhancing certain miRs might be a good therapeutic strategy. However, the

most comprehensively studied miRs, such as miR-33, still require further char-

acterisation and thorough preclinical testing prior to applying therapeutic strate-

gies to humans (Naar, 2013).

5.2 Inflammasomes

Cholesterol crystal accumulation in macrophages is a distinguishing feature

of atherosclerosis from initiation through advanced stages of lesion develop-

ment, and is both a consequence of lipid overloading in cells and a critical

contributor to the progression of atherosclerosis (Figures 1 and 4). Choles-

terol crystals activate immune cells and induce inflammation through multiple

mechanisms. One proposed mechanism is that cholesterol crystals activate

nucleotide-binding oligomerisation domain receptors (NLRP)3 inflamma-

somes (Figure 4) (Lu and Kakkar, 2014).

The inflammasome is a cytosolic caspase-activating molecular complex that

contributes to inflammatory responses. Uptake of oxLDL leads to activation of

NLRP3 inflammasomes in cultured macrophages, and increases IL-1β release.

As a consequence, NLRP3 deficiency in bone marrow-derived cells reduces

hypercholesterolaemia-induced atherosclerosis in mice. This is a leukocyte-

specific effect of NLRP3 inflammasomes because whole body deficiency of

NLRP3 inflammasome components such as NLRP3, apoptosis-associated

speck-like protein or caspase-1 have no effect on atherosclerosis development

in hypercholesterolaemic mice.

oxLDL uptake increases IL-1β secretion in macrophages, an important

cytokine representing NLRP3 inflammasome activation. However, global defi-

ciency of IL-1β, but not its deficiency in bone marrow-derived cells, affects ath-

erosclerosis in hypercholesterolaemic mice, implicating macrophage NLRP3

inflammasomes in promotion of atherosclerosis through an IL-1β-independent

mechanism. Activation of NLRP3 inflammasomes also results in release of

IL-1α. Comparable to effects of IL-1β, global deficiency of IL-1α reduces

hypercholesterolaemia-induced atherosclerosis in mice. Fatty acids, especially

oleic acid, increase secretion of IL-1α in macrophages independent of NLRP3

inflammasome activation. Deficiency of IL-1α in bone marrow-derived cells

reduces atherosclerosis. Therefore, although activation of NLRP3 inflamma-

somes increases both IL-1α and IL-1β, these two cytokines contribute to athero-

sclerosis through different mechanisms. Currently, a clinical trial, CANTOS (NCT01327846), is ongoing to deter-

mine whether inhibition of IL-1β has beneficial effects on coronary artery

disease. Despite the conflicting findings in animal models, completion of this

clinical trial will provide insights into the effects and mechanisms of this IL-1

subtype in the development of atherosclerosis in humans (Lu and Kakkar, 2014).

5.3 Trimethylamine and Trimethylamine-N-oxide

For decades, physicians and research investigators have noted that consumption

of food abundant in saturated fat and cholesterol increases risk for atheroscle-

rosis. These foods are also rich in trimethylamine (TMA). Recently, potential

mechanisms by which a TMA-enriched diet promotes atherosclerosis have been

examined. In addition to food, TMA is produced by intestinal microbiota diges-

tion of choline and phosphatidylcholine. On absorption, TMA is oxidised by

hepatic flavin monooxygenases to generate trimethylamine-N-oxide (TMAO).

TMAO has been shown to augment atherosclerosis in mice through the reg-

ulation of multiple components of lipid metabolism. For example, TMAO

decreases mRNA abundance of ABCA1 and ABCG1 in cultured mouse perito-

neal macrophages, resulting in attenuated cholesterol export to apoA-I (Chapter

15). TMAO increases CD36 and SR-A in macrophages, modulates bile acid

metabolism and sterol transporters in both the liver and the intestine, and pro-

motes cholesterol delivery but diminishes reverse cholesterol transport in vivo

(Tang and Hazen, 2014). However, TMAO does not influence mRNA levels of

LDL receptor or cholesterol synthesis genes (Chapters 11 and 17). Effects of

TMAO on atherosclerosis development are also supported by findings in human

cohort studies. Plasma concentrations of TMAO are associated with common

events of atherosclerosis such as myocardial infarction, stroke or death (Tang

and Hazen, 2014; Micha et al., 2010).

6. TRADITIONAL AND EVOLVING LIPID-LOWERING

THERAPIES FOR THE TREATMENT OF ATHEROSCLEROSIS

In addition to lifestyle changes, lipid-regulating agents are widely used to

improve dyslipidaemia in high-risk patients (Chan et al., 2014). Current guide-

lines recommend statins as first-line lipid regulators. Inhibitors of cholesterol

absorption or bile acid sequestrants represent additional or alternative agents

for patients who are intolerant to statins or require optimal reduction in plasma

LDL cholesterol. However, many statin-treated patients, including those with

metabolic syndrome or type 2 diabetes, have significant residual atherosclerosis

risk, due in part to persistent abnormalities in TAG-rich lipoproteins and HDL

(Chapter 19). Treatment approaches involve the additional use of other lipid-

regulating agents to treat atherogenic dyslipidaemia by harnessing their comple-

mentary mechanisms of action. Increased understanding of the biological and

molecular mechanisms underlying the atherosclerotic process at its interface  with lipoprotein metabolism, together with progress in our comprehension of

the molecular genetics of lipid disorders, has highlighted several new targets for

therapeutic intervention, as well as novel treatment options (Figure 5).

6.1 Statins

Inhibition of HMG-CoA reductase, a rate-limiting enzyme in hepatic cholesterol

synthesis by statins, results in the reduction of intracellular cholesterol content

that in turn induces an increase in SREBP-2-mediated hepatic LDL receptor

synthesis (Sahebkar and Watts, 2013b) (Chapter 11). This increases the clear-

ance of atherogenic lipoproteins, particularly LDL, as well as chylomicron rem-

nants and VLDL remnants (Figure 5). Statins are the most efficacious agents for

lowering the plasma concentration of LDL cholesterol and apoB-100. Statins

also decrease chylomicron remnants and VLDL TAGs, reduce small dense

LDL particles and modestly increase HDL cholesterol. Statins decrease hepatic

apoB-100 production, although results are inconsistent. Divergent results have

also been reported on the effects of statins on HDL metabolism. However, there

is no evidence that statin-induced HDL effects contribute to cardiovascular ben-

efit. Irrespective of its effects, clinical trials have consistently demonstrated that

statin therapy reduces cardiovascular events.

6.2 Fibrates

Several trials demonstrate that fibrates decrease atherosclerotic events in

patients with the metabolic syndrome and type 2 diabetes, which is character-

ised by increased plasma VLDL TAG and reduced plasma HDL cholesterol (Do

et al., 2014). Fibrates are agonists of the nuclear hormone receptor, peroxisome

proliferator-activated receptor-α (PPAR-α). Fibrates decrease plasma TAG up

to 50%, LDL cholesterol up to 20%, increase HDL cholesterol up to 20% and

decrease small dense LDL particles. The mechanistic action of fibrates is not

fully understood, but they decrease TAG substrate availability to liver by stimu-

lating fatty acid oxidation, thereby decreasing hepatic VLDL secretion. Fibrates

also promote VLDL lipolysis by activating lipoprotein lipase (LPL) and reduc-

ing apoC-III gene expression. Fibrates increase the expression of apoA-I and

ABCA1 by activating LXR, thereby promoting reverse cholesterol transport.

6.3 Niacin

Niacin has the theoretical potential to be an optimal drug for the treatment of ath-

erosclerotic diseases (Do et al., 2014). Niacin lowers plasma TAG concentrations

by up to 30%, LDL cholesterol up to 15%, and increases HDL cholesterol by up

to 25% and causes a shift of small dense LDL to large buoyant LDL particles.

Niacin is one of the few agents that can decrease plasma Lp(a). The mechanism

by which niacin modulates lipid metabolism has not been elucidated. Niacin decreases hepatic secretion of VLDL TAG by inhibiting fatty acid mobilisation

from peripheral adipocytes, thus decreasing the hepatic synthesis and secretion

of VLDL and the concentrations of its products, intermediate-density lipopro-

tein (IDL) and LDL. Niacin also increases plasma HDL cholesterol and apoA-I

concentrations by decreasing clearance of HDL apoA-I. Despite this desirable

profile of action on plasma lipoproteins, and positive results from early clini-

cal trials, two recent clinical trials failed to demonstrate significant benefits of

niacin on cardiovascular events in statin-treated patients (Rached et al., 2014).

6.4 Cholesterol Absorption Inhibitors

Ezetimibe selectively lowers plasma LDL cholesterol concentrations (Do

et al., 2014). Ezetimibe inhibits the function of Niemann–Pick 1-Like 1 pro-

tein (NPC1L1) resulting in reduced intestinal cholesterol absorption, attenuated

cholesterol transport in chylomicron remnants and reduced hepatic cholesterol

content (Figure 5). SREBP-2-mediated upregulation of hepatic LDL recep-

tors stimulates hepatic uptake of plasma apoB-100-containing lipoproteins and

accounts for up to 20% decreases in plasma LDL cholesterol. Ezetimibe also

significantly decreases LDL particle number, but has an inconsistent effect on

LDL particle size. Ezetimibe decreases intrahepatic TAGs, independent of body

weight, visceral fat and insulin sensitivity, although the mechanism remains

unclear. Inhibition of cholesterol absorption and hepatic cholesterol synthe-

sis with ezetimibe in combination with a statin, has complementary effects

on the fractional catabolism of apoB-containing lipoproteins. Accordingly, a

combination of ezetimibe and a low-dose statin can more effectively decrease

LDL cholesterol than higher doses of statin alone. Although some short-term

imaging studies have reported little or no benefit of ezetimibe treatment, the

recent Improved Reduction of Outcomes: Vytorin Efficacy International Trial

(IMPROVE-IT) study demonstrated significant benefits of ezetimibe for the

reduction of cardiovascular events.

6.5 ω-3 Polyunsaturated Fatty Acids

Fish oils are a rich source of ω-3 polyunsaturated fatty acids (PUFAs), mainly

eicosapentenoic acid (EPA) and docosahexenoic acid (DHA). Fish oils are rec-

ommended as an adjunct to diet for the reduction of VLDL–TAG and prevention

of acute pancreatitis (Rached et al., 2014). Human studies demonstrate that ω-3

PUFA supplementation decreases hepatic VLDL production and accordingly

reduces VLDL–TAG. There are no consistent effects on plasma concentra-

tions of LDL cholesterol, HDL cholesterol or HDL–apoA-I. The mechanistic

action of ω-3 PUFAs on plasma TAGs includes inhibition of diacylglycerol

acyltransferase (DGAT), fatty acid synthase and acyl-CoA carboxylase enzyme

activities, resulting in decreased TAG and fatty acid synthesis. ω-3 PUFAs also

enhance fatty acid β-oxidation by stimulating PPAR-α. However, they are  much 

weaker PPAR-α agonists than fibrates. Although some early trials have shown

cardiovascular benefits, a recent meta-analysis has cast doubts on the therapeu-

tic efficacy of ω-3 PUFA and concluded that its supplementation is not associ-

ated with decreased risk of atherosclerosis-related events.

6.6 Proprotein Convertase Subtilisin/Kexin Type 9 Inhibitors

PCSK9 targets the LDL receptor for degradation and was discovered as the third

gene involved in autosomal dominant hypercholesterolaemia. Some mutations

of PCSK9 enhance its function and cause hypercholesterolaemia, whereas loss-

of-function mutations are associated with hypocholesterolaemia and reduced

cardiovascular risk. By enhancing hepatic LDL receptor degradation, PCSK9

increases plasma LDL cholesterol. PCSK9 is primarily regulated transcription-

ally by SREBP2 and the secreted protein binds the epidermal growth factor-like

repeat A domain of the LDLR, which results in attenuated LDLR recycling

and its targeting to lysosomes for degradation. While statins effectively reduce

plasma LDL cholesterol in many patients, statins also upregulate PCSK9 via

SREBP2, thereby limiting statin’s efficacy in reducing plasma LDL cholesterol.

Antibodies against PCSK9 have been developed that increase the numbers of

LDL receptors available at the cell surface and reduce plasma LDL cholesterol

(Figure 5) (Rached et al., 2014). These include human mononclonal antibod-

ies or humanised antibodies that are injected intravenously or subcutaneously.

In phase II and III trials, anti-PCSK9 therapy of patients lowers LDL choles-

terol by 40–65% in both heterozygous FH and non-FH patients, with or without

statin and ezetimibe treatment. Lp(a) concentrations were also decreased sig-

nificantly. Other strategies to inhibit PCSK9, including ASO therapies, small

interfering RNAs and recombinant adnectins are under development. PCSK9

inhibition is a promising new approach for treatment of hypercholesterolaemia

and potentially atherosclerosis.

6.7 ASO Therapies

ASOs are short, single-stranded, synthetic analogues of natural nucleic acids

designed to bind to a target mRNA in a sequence-specific manner. Injection

of a short complementary ASO sequence binds to mRNA and induces either

selective degradation of the complex by endogenous nucleases or inhibition of

mRNA processing and/or function. The most advanced ASO is mipomersen

that is designed to inhibit hepatic apoB-100 synthesis (Rached et al., 2014; Do

et al., 2014) (Figure 5). Phase II and III trials revealed that mipomersen reduced

plasma concentrations of apoB, LDL cholesterol, non-HDL cholesterol and

TAGs by 20–65% in patients with different forms of hyperlipidaemia, includ-

ing heterozygous and homozygous FH, and in patients treated with statins and

ezetimibe. Mipomersen reduced Lp(a) levels by 25% by inhibiting its synthesis.

The main side effects involve injection-site reactions and hepatic steatosis. Beyond apoB and PCSK9, several other potential ASO targets for treat-

ment of hyperlipidaemia have been identified, including apoC-III and Lp(a)

(Figure 5). ApoC-III normally inhibits LPL-mediated lipolysis of chylomicrons

and VLDL, and attenuates hepatic clearance of their remnants. Human genetic

studies have associated decreased plasma concentrations of apoC-III with lower

TAG concentrations and diminished cardiovascular events. In animal models

and Phase I human trials, an apoC-III ASO inhibitor produced potent, selec-

tive reductions in plasma apoC-III and TAG concentrations as well as marked

increases in HDL-C. ASOs with the potential to lower PCSK9 and Lp(a) plasma

concentrations are in early stages of development.

6.8 Microsomal Triacylglyceride Transfer Protein Inhibitors

MTP is critical for the formation and secretion of apoB-containing lipopro-

teins from the liver and intestine (Do et al., 2014) (Chapter 16). MTP trans-

fers TAG, CE and phospholipid to apoB within the cell during the lipoprotein

assembly process. Mutations in the MTP gene lead to a rare condition known

as abetalipoproteinaemia, in which plasma apoB-containing lipoproteins are

undetectable. In animal models, MTP inhibition results in profound reductions

in plasma TAG and cholesterol concentrations (Figure 5). Although early MTP

inhibitors reduced LDL cholesterol, further development of most inhibitors

has been discontinued due primarily to hepatic fat accumulation. Lomitapide

is the only systemic MTP inhibitor currently in development. Lomitapide sub-

stantially reduced levels of LDL cholesterol in homozygous FH and in clini-

cal trials proved very effective in reducing LDL cholesterol in patients with

homozygous FH as well as in patients with moderate hypercholesterolaemia,

either as monotherapy or when combined with ezetimibe. However, variable

gastrointestinal side effects, minor elevations in liver transaminase levels and

increases in hepatic fat were reported. Lomitapide was recently approved for

treatment of homozygous FH, as this drug was considered to provide the ben-

efits of cardiovascular protection that outweighed risks of increased hepatic fat.

An intestine-targeted MTP inhibitor has been shown to decrease both VLDL

and chylomicron production and resulted in weight loss without elevations of

liver enzymes or increases in hepatic fat, suggesting that this approach may

be more applicable to treatment of a broader range of lipid disorders (Rached

et al., 2014).

6.9 ACAT and DGAT Inhibitors

One of the important steps in atherogenesis and cholesterol accumulation in the

arterial wall is esterification of cholesterol in macrophages, which promotes

foam cell formation. This provided a rationale for the development of ACAT

inhibitors; however, early versions inhibited both ACAT isoforms (ACAT-1 and -2)

(Rached et al., 2014). Imaging based clinical trials failed to demonstrate reduced atherosclerosis progression in patients with FH. By contrast, an ACAT-

1-selective inhibitor K-604 is under development by Kowa Pharmaceuticals.

ACAT-2 is the isoform responsible for cholesterol esterification in the liver and

intestine and the provision of CE for lipoprotein synthesis (Figure 5). Genetic

deletion of ACAT-2 or pharmacological inhibition of ACAT-2 with ASOs in

mice inhibits the secretion of both chylomicrons and VLDL into plasma and

attenuates atherosclerosis in Ldlr−/− mice. As a result, specific inhibitors of

ACAT-2 are currently under development.

DGATs esterify the third fatty acid to DAG to form TAG in adipose tissue, the

intestine and the liver. Studies in mice with DGAT-1 deficiency or mice treated

with DGAT-1 inhibitors demonstrated reduced plasma TAGs, hepatic steatosis and

obesity, which were paralleled by improved insulin resistance. In patients with

severe hypertriglyceridaemia, with LPL deficiency, a DGAT-1 inhibitor decreased

fasting plasma TAG levels. Clinical trials are ongoing in patients with coronary

artery disease to further substantiate these findings. DGAT-2 is the isoform respon-

sible for TAG synthesis in liver and intestine and the provision of CE for lipopro-

tein synthesis (Figure 5). Specific DGAT-2 inhibitors decrease plasma and liver

TAG in experimental animals and are currently under development for clinical use.

6.10 High-Density Lipoprotein Modulating Drugs

Epidemiological evidence demonstrates that low HDL cholesterol concentra-

tions predict cardiovascular events across multiple populations. However, thera-

peutic strategies to target HDL have not yet achieved success (Rached et al.,

2014). Three HDL cholesterol-raising drugs, two early cholesteryl ester trans-

fer protein (CETP) inhibitors and niacin, have failed in prospective interven-

tion trials due to off-target side effects or lack of efficacy. Current therapeutic

approaches are focussed on improved HDL metabolism, increased HDL particle

number and enhanced HDL function. Inhibition of CETP lipid transfer activ-

ity elevates plasma HDL cholesterol concentrations by 30–140% and reduces

concentrations of LDL cholesterol and Lp(a) by up to 40% (Figure 5). Large

cardiovascular end-point trials of two recently developed CETP inhibitors are

ongoing. Other approaches that transiently increase HDL particle numbers and

enhance HDL functionality include infusions of reconstituted HDL, administra-

tion of apoA-I mimetic peptides, antagonists of miR33 (antimiR33) (Figure 5)

and injection of delipidated HDL. These strategies involve structural remodel-

ling of endogenous HDL particles and are targeted towards export of cholesterol

from the atherosclerotic plaque (Figure 5). The impact of these agents on cardio-

vascular events remains to be evaluated.

7. FUTURE DIRECTIONS

The pathogenesis and mechanisms of atherosclerosis are complex. In the

past several decades, lipid and lipoprotein-related mechanisms have been studied extensively, and medical treatments of atherosclerosis targeting

lipid metabolism have been improved. However, many questions remain

unresolved: (1) Although inflammation is recognised as a critical mechanism in

hypercholesterolaemia-induced atherosclerosis, the therapeutic effects of directly

targeting inflammation have not been translated into human use. (2) There is

substantial experimental evidence that HDL reduces atherosclerosis; however,

efforts to increase HDL cholesterol were hampered by either off-target effects or

lack of beneficial effects on atherosclerotic diseases. (3) Currently, there is only

weak evidence that drugs regress preexisting atherosclerosis in humans. Several

animal studies have demonstrated that therapeutic intervention can induce regres-

sion of atherosclerosis, but only in the setting of significant lowering of plasma

cholesterol concentrations. (4) Therapeutic approaches that directly influence

mechanisms of atherogenesis within the arterial wall, including those that spe-

cifically target cellular lipid metabolism, have yet to be developed or evaluated.

In future studies, in addition to continuously exploring the complex mechanisms

of lipid/lipoprotein-related atherosclerosis in animal models, it will be important

to apply convincing findings from animal models into human clinical trials.