Sie sind auf Seite 1von 11

REVIEW

CURRENT
OPINION The central role of arterial retention of cholesterol-
rich apolipoprotein-B-containing lipoproteins in the
pathogenesis of atherosclerosis: a triumph
of simplicity
Jan Borén a,b and Kevin Jon Williams a,b,c

Purpose of review
Today, it is no longer a hypothesis, but an established fact, that increased plasma concentrations of
cholesterol-rich apolipoprotein-B (apoB)-containing lipoproteins are causatively linked to atherosclerotic
cardiovascular disease (ASCVD) and that lowering plasma LDL concentrations reduces cardiovascular
events in humans. Here, we review evidence behind this assertion, with an emphasis on recent studies
supporting the ‘response-to-retention’ model – namely, that the key initiating event in atherogenesis is the
retention, or trapping, of cholesterol-rich apoB-containing lipoproteins within the arterial wall.
Recent findings
New clinical trials have shown that ezetimibe and anti-PCSK9 antibodies – both nonstatins – lower
ASCVD events, and they do so to the same extent as would be expected from comparable plasma LDL
lowering by a statin. These studies demonstrate beyond any doubt the causal role of apoB-containing
lipoproteins in atherogenesis. In addition, recent laboratory experimentation and human Mendelian
randomization studies have revealed novel information about the critical role of apoB-containing
lipoproteins in atherogenesis. New information has also emerged on mechanisms for the accumulation in
plasma of harmful cholesterol-rich and triglyceride-rich apoB-containing remnant lipoproteins in states of
overnutrition. Like LDL, these harmful cholesterol-rich and triglyceride-rich apoB-containing remnant
lipoprotein remnants become retained and modified within the arterial wall, causing atherosclerosis.
Summary
LDL and other cholesterol-rich, apoB-containing lipoproteins, once they become retained and modified
within the arterial wall, cause atherosclerosis. This simple, robust pathophysiologic understanding may
finally allow us to eradicate ASCVD, the leading killer in the world.
Keywords
apolipoprotein-B, atherosclerosis, Mendelian randomization studies, prospective randomized controlled
trials, proteoglycan

INTRODUCTION
Atherosclerotic cardiovascular disease (ASCVD) is a
Department of Molecular and Clinical Medicine, University of Gothen-
the leading cause of death globally. Widespread burg, bSahlgrenska University Hospital, Gothenburg, Sweden and
c
in developed areas of the world and now becoming Section of Endocrinology, Diabetes, & Metabolism, Department of
more common in poorer countries, deaths from Medicine, Lewis Katz School of Medicine at Temple University,
Philadelphia, Pennsylvania, USA
ASCVD are projected to increase to more than
Correspondence to Jan Borén, MD, PhD, Wallenberg Laboratory,
2.3  107 per year by 2030. It is tempting to think
Sahlgrenska University Hospital, Gothenburg, Sweden.
of atherosclerosis as a disease of the modern Tel: +46 733 764264; e-mail: jan.boren@wlab.gu.se
lifestyle. Yet atherosclerosis has plagued humans Curr Opin Lipidol 2016, 27:473–483
for thousands of years and has been detected in
DOI:10.1097/MOL.0000000000000330
4000-year-old Egyptian mummies.
This is an open-access article distributed under the terms of the Creative
Today, it is no longer a hypothesis, but an Commons Attribution-Non Commercial-No Derivatives License 4.0, where
established fact, that increased plasma concen- it is permissible to download and share the work provided it is properly
trations of cholesterol-rich apolipoprotein-B cited. The work cannot be changed in any way or used commercially.

0957-9672 Copyright ß 2016 Wolters Kluwer Health, Inc. All rights reserved. www.co-lipidology.com

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


Atherosclerosis: cell biology and lipoproteins

Thus, cholesterol per se is not the cause of this disease.


KEY POINTS In contrast, cholesterol-rich apoB-lipoproteins
 Recent studies provide new support for the ‘response- possess special properties that render them uniquely
to-retention’ hypothesis – namely, that the key initiating pathogenic in ASCVD.
event in atherosclerotic cardiovascular disease
(ASCVD) is the retention, or trapping, of cholesterol-rich
apoB-containing lipoproteins within the arterial wall. The response-to-retention hypothesis of
atherogenesis
 Accordingly, the most effective therapies against
ASCVD – LDL-lowering drugs – are based upon the The response-to-retention hypothesis drew on work
principle that decreasing the concentration of apoB- from the 1940s to the 1980s showing that lipopro-
lipoproteins in the circulation decreases the probability teins can interact with proteoglycans of the arterial
that they will enter and become retained in wall [14–17]. Lipids and apoB accumulate at lesion-
the subendothelium. prone sites before gross morphological changes
 With the rise of overnutrition, obesity, and the occur [18–21], and retention of apoB-lipoproteins
atherometabolic syndrome, cholesterol-rich and is seen throughout the progression of atherosclero-
triglyceride-rich apoB-containing remnant lipoproteins sis. The consequences of the retention of apoB-lip-
(C-TRLs) have emerged as major drivers of human oproteins include, not only an accumulation of
ASCVD worldwide, also due to their retention within lipid, but also prolonged exposure of these particles
the arterial wall. to local enzymes and other factors within the vessel
 Using our extensive knowledge of the pathogenesis of wall. The retained and modified apoB-lipoproteins
atherosclerosis, we can now reclassify nearly all trigger cellular responses within the artery wall that
epidemiologic risk factors for ASCVD events into accelerate further lipoprotein retention and lesion
causative agents, exacerbating factors, and mere development [9,11,22].
bystander phenomena, thereby focusing therapeutic
efforts on causative and exacerbating factors.
Influx of apolipoprotein-B-lipoproteins
 At this point, we know enough about the into the artery wall
pathophysiology of atherosclerosis that eradication of Lipoproteins normally flux into and out of the
this major killer has become a genuine possibility. arterial wall [20,23]. The molecular mediators of this
transendothelial movement of lipoproteins remain
incompletely characterized. Recent evidence has
(apoB)-containing lipoproteins are causatively suggested roles for caveolin-1 [24,25] and the scav-
linked to ASCVD and that lowering LDL concen- enger receptor class B type I (SR-BI) [26], but
trations with statins and nonstatins reduces athero- additional mediators are likely [27].
&& &&
sclerotic cardiovascular events in humans [1,2 –4 , ApoB-lipoproteins up to only approximately
& &
5 ,6 ]. Despite the clarity of the evidence today, the 70 nm in diameter [i.e., chylomicron remnants,
role of LDL was questioned and even ridiculed for smaller VLDL, IDL, LDL, and lipoprotein(a)] can
many years [7]. During that period, numerous com- efficiently cross an intact endothelium, and among
peting hypotheses [8–11] were articulated to explain these, the smaller ones pass more readily than the
the initiating events in atherogenesis. After decades larger ones [23]. This size limitation explains why
of research, however, there is now a large body of patients with lipoprotein lipase deficiency do not
evidence to support the ‘response-to-retention hy- develop atherosclerosis despite very high plasma
pothesis’ – namely, that the key initiating event in levels of large apoB-containing chylomicrons.
atherogenesis is the retention, or trapping, of choles- Studies in-vivo of the influx and egress of LDL
terol-rich apoB-containing lipoproteins within the particles into and out of the arterial intima
arterial wall. The retained lipoproteins and their have shown that, although the endothelial layer
byproducts provoke a series of strikingly maladaptive remains intact over most atherosclerotic lesions,
local responses that cause plaque initiation, growth, its permeability to LDL becomes abnormally high
and evolution (Fig. 1). [20,28,29]. The precise stimuli and molecular mech-
Modern research on atherosclerosis began anism(s) responsible for this increased permeability
with the insightful and pioneering ‘cholesterol are still not elucidated, and it was not known until
hypothesis’ of Anichkov and Khalatov [12,13]. As a recently if the increased endothelial permeability
point of clarification, however, we now know for LDL could be reversed. To examine this issue,
that many things that carry cholesterol near Bartels et al. [30 ] fed LDL receptor null (Ldlr / )
&&

arteries – such as normal HDL and erythrocytes mice on a high-cholesterol diet for up to 5 months
(which transport a similar amount of cholesterol in to increase their plasma concentrations of apoB-
blood as LDL does) – do not cause atherosclerosis. lipoproteins, thereby provoking the formation of

474 www.co-lipidology.com Volume 27  Number 5  October 2016

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


The pathogenic simplicity of atherosclerosis Borén and Williams

LUMEN
LDL IDL Remnant

Monocyte
ap
sc
rou
Fib

Ps
MM
INTIMA

LPL Necrotic
debris
SMase T-cell
TF
UC-rich material
PGs IL-4,
IFN-γ Lipid-rich core

se s
Ea a s e
Ch rot e
SM
Ci

P
n-m Foam cell with LP-derived
igr
ati lipids, unable to emigrate
on
MEDIA

Retention Responses

FIGURE 1. The response-to-retention model of initiation and progression of atherosclerosis. Arrows are color-coded to indicate
crucial mechanisms in the retention of cholesterol-rich atherogenic apolipoprotein-B-lipoproteins within the arterial wall, which
is the key initiating step in atherogenesis (yellow) and then local responses to the retained and modified lipoproteins that lead
to plaque growth and evolution (red). The main text describes molecular mechanisms for early retention and aggregation of
apoB-lipoproteins within the arterial wall and then the acceleration of lipoprotein retention after plaque initiation and other
subsequent maladaptive responses. Two key enzymes implicated in the retention and aggregation of apoB-lipoproteins and
hence atherogenesis are the secretory sphingomyelinase (produced by endothelium, particularly when activated, and by
plaque macrophages) and lipoprotein lipase (produced by plaque macrophages and acting as a ‘bridge’ to facilitate further
adherence of apoB-lipoproteins to arterial matrix). ChEase, cholesteryl esterase; LP, lipoprotein; LPL, lipoprotein lipase; MMPs,
matrix metalloproteinases; PGs, proteoglycans; Remnant, cholesterol-rich and triglyceride-rich apoB-containing remnant
lipoprotein (C-TRL remnant); SMase, sphingomyelinase; SMC, smooth muscle cell, including immature SMCs; TF, tissue factor;
UC, unesterified cholesterol. Source: Adapted from [11].

atherosclerotic plaques. To investigate the effect of caveolin-1 and SR-BI were not examined. Bartels
&&
an abrupt lowering of plasma apoB-lipoprotein con- et al. [30 ] ruled out a nonspecific process by
centrations on aortic endothelial permeability to showing that the anti-apoB ASO treatment had no
LDL and the fractional degradation rates of apoB- effect on endothelial permeability to Evans blue dye.
lipoproteins within the atherosclerotic lesions, In addition, correction of the hypercholesterolemia
these authors used an anti-apoB antisense oligonu- caused the fractional degradation rates of apoB
cleotide (ASO) to reduce hepatic apoB synthesis. within the plaques to fall sharply. Most importantly,
Concentrations of apoB-containing lipoproteins in the rapid decreases in permeability to LDL and in the
plasma fell sharply to nearly undetectable levels fractional degradation of LDL that enters the plaque
within 3 days after administration of the ASO. may turn out to be crucial for subsequent plaque
&
The authors then analyzed the aortic plaques after stabilization and atherosclerosis regression [6 ].
1 or 4 weeks of ASO treatments. Lowering of plasma
apoB-lipoprotein concentrations reduced LDL per- Retention of atherogenic apolipoprotein-
meability into the arterial wall within 1 week of ASO B-lipoproteins within the artery wall as the
treatment, and this effect preceded any detectable key pathogenic event in atherosclerosis
changes in the cellular composition or size of the Because the capacity of transendothelial transport
&&
atherosclerotic lesions [30 ]. The molecular mech- is high even in normal arterial segments, it is
anisms involved still remain unclear, and effects on unlikely that influx of LDL and other smaller

0957-9672 Copyright ß 2016 Wolters Kluwer Health, Inc. All rights reserved. www.co-lipidology.com 475

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


Atherosclerosis: cell biology and lipoproteins

apoB-lipoproteins from the bloodstream is rate performed that do not change plasma concen-
limiting in determining their concentrations within trations of apoB-lipoproteins in hypercholesterole-
the artery wall. Instead, mechanisms for the selec- mic animals but specifically decrease or increase
tive retention of a tiny percentage of the apoB-lip- the retention of these particles within the arterial
oproteins that enter the artery wall drive their local wall. These interventions correspondingly decrease
accumulation and hence atherogenesis. Support for or increase atherogenesis. Such interventions
this model came from early work by Schwenke and include mutagenesis of the proteoglycan/GAG-
Carew showing that LDL entry did not differ binding domain of apoB100, thereby slowing LDL
between normal arterial sites that are susceptible retention and atherogenesis [36], manipulations
versus resistant to subsequent plaque development. of arterial matrix to alter its affinity for apoB
&& &
Although the same amount of LDL entered, the [31,37–39,40 ,41 ], and knockouts of arterial-wall
susceptible sites, though healthy, already showed enzymes specifically implicated in apoB-lipoprotein
greater LDL accumulation [20]. More recent support retention and aggregation [42–45].
comes from molecular studies by Tran-Lundmark The first of these studies, which used mutagen-
et al. who crossed glycosaminoglycan (GAG)- esis of apoB100 to block its direct adherence
deficient perlecan (Hspg2D3/D3) mice with hypercho- to arterial matrix, emphasizes the importance of
lesterolemic apolipoprotein-E (apoE)-null (Apoe / ) LDL quality, as opposed to simply focusing on its
mice. Because perlecan participates both as a per- concentrations in plasma. Other variations in LDL
meability barrier and as a proretentive molecule quality may also affect its atherogenicity, such as
within the murine arterial wall, the influx of LDL surface lipid composition [46] and core lipid com-
and other apoB-lipoproteins into the aortic wall in position (which influences the conformation of
vivo was increased in Hspg2D3/D3/Apoe / mice com- apoB and hence its interactions with proteoglycans)
pared with Apoe / controls, whereas subendothe-
&&
[47,48 ,49]. In addition, the protein composition of
lial retention of LDL was paradoxically lower. In apoB-lipoproteins influences their interactions
other words, this ingenious genetic manipulation with proteoglycans. As discussed above, apoE medi-
of perlecan separated the two processes. Impor- ates binding of lipoproteins to arterial GAGs,
tantly, these authors demonstrated a significant although apoE also facilitates hepatic disposal of
reduction of atherosclerotic lesions in Hspg2D3/D3/ atherogenic lipoproteins. In contrast, the small
Apoe / mice compared with Apoe / controls, indi- exchangeable apolipoprotein, apolipoprotein-C
cating the key role of decreased apoB-lipoprotein (apoC)-III, increases the affinity of LDL for arte-
retention regardless of the increase in endothelial rial-wall proteoglycans, whereas decreasing hepatic
permeability to LDL entry. Thus, most apoB- uptake of apoB-lipoproteins [50], thereby increasing
lipoprotein particles that entered the arterial wall the accumulation of atherogenic lipoproteins
left without contributing to the growth of the within the vessel wall [51,52]. The mechanism has
atherosclerotic lesion [31]. been unclear because apoC-III by itself lacks posi-
In earliest atherogenesis, negatively charged tively charged domains to bind arterial-wall proteo-
proteoglycans in the extracellular matrix of the glycans [51]. Hiukka et al. [53] demonstrated that an
arterial intima bind and trap apoB-lipoproteins via increased apoCIII : apoB molar ratio in LDL isolated
electrostatic interactions with specific positively from patients with type 2 diabetes correlated with
charged aminoacyl residues in the full-length hep- lower contents of unesterified cholesterol, sphingo-
atic form of apoB, apoB100 (residues 3359–3369) myelin, ceramide, and GM1. Importantly, this com-
[32], and in the truncated intestinal form, apoB48 bination of compositional changes was associated
(residues 84–94) [33]. Moreover, apoB48-lipopro- with increased proteoglycan binding [53]. The
teins typically contain numerous molecules of apoE, mechanism remains unclear but one possibility is
an apoprotein that has a proteoglycan-binding that a more fluid monolayer on the LDL particle
domain almost identical to the proteoglycan- surface may facilitate the insertion of more mol-
binding sequence in apoB100. Proteoglycans are ecules of apoC-III and permit conformational
negatively charged due to the sulfate and carboxylic changes in apoB100 that increase its proteoglycan-
acid groups in their GAG side-chains. In experimen- binding affinity. In contrast, however, the apoC-
tal studies, several arterial-wall proteoglycans have III : apoB ratio in LDL from nondiabetic control
been shown to be important for apoB-lipoprotein patients showed no correlations with the content
retention and atherosclerosis, most notably the of these lipids, indicating less global changes in LDL
matrix-associated proteoglycans biglycan, perlecan, composition [53].
and versican [9,34,35]. Regarding the subendothelial arterial matrix,
Consistent with the response-to-retention recent work has implicated proteases [54] and other
&
model, numerous other interventions have been factors [55,56 ] in the regulation of the arterial-wall

476 www.co-lipidology.com Volume 27  Number 5  October 2016

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


The pathogenic simplicity of atherosclerosis Borén and Williams

content of LDL-binding proteoglycans and hence atherogenesis. Unraveling the molecular mechan-
&& &
atherosclerosis. She et al. [40 ,41 ] recently focused isms from disturbed flow to matrix proliferation and
on a less well known proteoglycan in the vascula- increased lipoprotein retention will be a fruitful area
ture, the neural/glia antigen 2 (NG2) proteoglycan. for future research.
Although NG2 is undetectable in the intima New work has focused on apoB-lipoprotein
of normal murine arteries, early atheromata in retention as a potential therapeutic target. One
Apoe / mice contain immature synthetic smooth strategy is the creation of mAbs that block apoB-
&&
muscle cells (s-SMCs) that express NG2 [40 ]. Thus, binding sites on GAG chains within the arterial
&
the atherogenic effects of NG2 seem to emerge with matrix [63,64 ]. Interestingly, the antigen-binding
accumulation of s-SMCs [21,57]. She et al. also dem- site (the CDR3 of the heavy chain) of one of
onstrated a remarkable role for NG2-positive s-SMCs these antibodies contains an arginine motif
in trapping LDL and then presenting it to macro- that resembles the major GAG-binding sequence
&
phages. It is a novel mechanism for the presentation in apoB100 [33,36,64 ]. The antibodies decrease
of undesirable material to macrophages that, we intra-arterial lipoprotein retention and hence the
speculate, may have evolved to facilitate the progression of atherosclerosis in hypercholesterole-
mic Apoe / mice [63,64 ].
&
disposal of certain infectious agents or cellular
debris [58,59]. In the context of retained apoB-
lipoproteins, however, the process becomes malad- Cholesterol-rich and triglyceride-rich
aptive. Thus, NG2-positive s-SMCs dramatically apolipoprotein-B-containing remnant
increase the uptake of LDL by macrophages within lipoproteins, which are metabolically
the arterial wall, promoting foam-cell formation, distinct from LDL, have emerged as a major
whereas NG2-deficient s-SMCs reduce LDL uptake. driver of human atherosclerosis
Despite worse obesity and worse hyperlipidemia, Lipid and lipoprotein concentrations are often
double knockout mice (Ng2 / /Apoe / ) developed assessed in fasting plasma, but the idea that particles
less atherosclerosis than did Apoe / controls. originating in the postprandial state may contribute
Surprisingly, the NG2 GAG chains were not required to atherosclerosis appeared in the literature at least
&&
for the binding of LDL to NG2 [40 ]. Furthermore, as far back as the 1940s [65,66]. In that era, however,
NG2 also bound acetylated LDL, indicating that the overwhelming driver of human atherosclerosis
NG2 has the capacity to bind LDL through hydro- was LDL, which is not primarily a postprandial
phobic interactions. These features make NG2 particle.
&&
unique amongst LDL-binding proteoglycans [40 ] The situation has changed dramatically since
and may be related to its normal biologic functions. then. With the rise of overnutrition, obesity, and
Why does atherosclerosis preferentially develop the atherometabolic syndrome, harmful choles-
at specific sites in the arterial tree? These sites are terol-rich and triglyceride-rich apoB-containing
often associated with branches and bifurcations, remnant lipoproteins (C-TRLs) have emerged as a
&
which expose the endothelium to disturbed laminar major driver of human ASCVD worldwide [67 ]. The
blood flow and shear stress [60]. Most conventional characteristic dyslipoproteinemia of the atherome-
explanations for increased atherosclerosis suscepti- tabolic syndrome increases plasma concentrations
bility at these sites have focused on biomechanical of C-TRL remnants, not LDL. The major defect is
alterations of endothelial cells, such as increased impaired hepatic removal of C-TRLs from plasma
&
permeability and elaboration of cytokines [61], [68 ]. Like LDL [9], C-TRL remnants become retained
&
but none of these proposed mechanisms has been within the arterial wall [6 ,11,33,69]. Mendelian
definitely demonstrated. In a recent advance, Stef- randomization studies have clearly demonstrated
fensen et al. showed that induction of disturbed their causal role in human ASCVD events [70]. Of
laminar flow in the straight segment of the other- note, each C-TRL particle contains up to 40 times
wise atherosclerosis-resistant common carotid more cholesterol compared with an LDL particle.
artery of mice provokes matrix proliferation in the Unfortunately, patients treated with optimal statin
&& &&
nearby arterial wall, transforming it into a lipopro- therapy [71] and even PCSK9 inhibitors [3 ,4 ]
tein-retaining site – but without altering LDL influx. exhibit considerable residual risk for ASCVD events,
This provides experimental evidence for a causal which may occur, in large part, because these agents
chain linking disturbed laminar flow patterns lower fasting or postprandial C-TRL levels by only
to increased arterial matrix deposition, apoB- 4–25% [72–74]. There is therefore a need to find
&&
lipoprotein retention, and atherosclerosis [62 ]. new therapeutic approaches to restore normal, rapid
Like the prior work cited above [20,31], this study hepatic removal of C-TRLs from plasma, to lower
contradicts the notion of a key role for disturbed residual ASCVD risk in obesity, the atherometabolic
endothelial permeability to LDL in early phases of syndrome, and type 2 diabetes.

0957-9672 Copyright ß 2016 Wolters Kluwer Health, Inc. All rights reserved. www.co-lipidology.com 477

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


Atherosclerosis: cell biology and lipoproteins

Aggregation and modifications of retained Ruuth et al. [46] recently demonstrated that
apolipoprotein-B-lipoproteins within the susceptibility of plasma LDL to aggregation upon
artery wall exposure to SMase ex vivo varies considerably
Following retention of cholesterol-rich apoB- amongst human patients, predicts future deaths
lipoproteins within the artery wall, the lipoproteins from ASCVD, and can be altered by diet. This work
have been shown to undergo several modifications adds to a growing body of data implicating SMase in
with important biological consequences. For human atherosclerosis [9,75]. Crucially, the work
example, proteoglycan-bound LDL in vitro forms identifies LDL aggregability in the presence of SMase
aggregates that resemble material seen in vivo, and as a new, potentially modifiable cardiovascular risk
retained lipoproteins are more susceptible to further factor. Aggregation-prone LDL – dubbed ‘unstable’
modifications. Key enzymes implicated in apoB-lip- LDL – exhibits several compositional character-
oprotein retention, aggregation, and atherogenesis istics, including an increased sphingomyelin : phos-
include the secretory sphingomyelinase (S-SMase), phatidylcholine (SM : PC) ratio [46]. Interestingly,
lipoprotein lipase, and the nonpancreatic secretory an increase in dietary polyunsaturated fatty acids
group V phospholipase-A2 (PLA2-V) [9,42–45,75] and fiber decreased the SM : PC ratio and stabilized
(Fig. 1). These enzymes, particularly lipoprotein LDL, whereas an increase in dietary sucrose caused
lipase, bridge between LDL and proteoglycans inde- the opposite. Consistent with these findings, a diet
pendently of the physical state of apoB and thereby rich in saturated fats has been shown to increase
cause a shift in the retentive mechanism from a serum SMase activity and, consequently, increase
low-affinity process (apoB-GAG binding) in the LDL aggregation in mice [81]. These results further
pristine arterial wall to a high-affinity process (lip- emphasize the importance of LDL quality in human
oprotein-GAG binding) in an established atheroma. ASCVD. Moreover, measurement of LDL instability
This shift in retentive mechanism has direct clinical in the presence of SMase may serve as a predictive
consequences: a lifetime plasma LDL cholesterol biomarker for the identification of patients at unrec-
concentration of 80 mg/dl almost always protects ognized risk of cardiovascular death who might
from atherosclerosis [76] but pre-existing plaques benefit from specific, targeted interventions.
grow if the LDL-cholesterol is 80 mg/dl [77].
Acidity in deeper parts of normal intima and Retained lipoproteins induce maladaptive
within atherosclerotic plaques enhances lipolytic, cellular responses in the arterial wall that
proteolytic, and other modifications of apoB-con- accelerate further apolipoprotein-B-
taining lipoproteins that strongly increase their lipoprotein retention
affinity for proteoglycans and may therefore accel- Retained and aggregated lipoproteins provoke a series
erate their retention and the ensuing maladaptive of local biological responses that eventually include a
&
cellular responses in the arterial intima [78 ]. In maladaptive reaction dominated by abnormally per-
a fascinating advance, Sneck et al. elucidated the sistent macrophages and T cells that accelerate lip-
mechanism for SMase-induced aggregation of LDL. oprotein retention and other features of plaque
The enzyme induces a massive, global confor- development (Fig. 1). Aggregated apoB-lipoproteins
mational change in apoB that exposes hydrophobic are avidly taken up by macrophages [9,82] and by
motifs that apparently adhere from one digested vascular SMCs [83] leading to foam-cell formation.
LDL particle to the next [79]. These processes stimulate the release of proathero-
Several lines of evidence implicate aggregation genic factors that induce the synthesis of proteogly-
of apoB-lipoproteins within the arterial wall as a cans with enhanced affinity for atherogenic
major accelerator of lipoprotein retention and athe- lipoproteins [9,82]. In addition, monocyte/macro-
rogenesis. Once LDL and other apoB-lipoproteins in phages recruited into atheromata secrete proretentive
the arterial wall have aggregated, diffusion back into enzymes, notably lipoprotein lipase, sphingomyeli-
plasma becomes essentially impossible, due to the nase, and PLA2, that accelerate further retention of
large size of the aggregates and their greatly enhanced atherogenic lipoproteins. In addition, persistent
affinity as multivalent ligands for arterial matrix, macrophages in atherosclerotic plaques secrete pro-
as well as conformational changes that expose teases that weaken the overlying fibrous cap, and
additional positively charged domains on apoB. Lipid these cells release tissue factor, a potent procoagulant.
hydrolysis and other modifications of the retained, These maladaptive responses favor plaque rupture,
aggregated lipoproteins release biologically active robust clot formation that often occludes the arterial
byproducts that recruit macrophages and other cells lumen, and downstream ischemia (Fig. 1).
into the developing lesion, while also blocking their A critical question is why retained apoB-lipopro-
emigration, thereby accelerating disease progression teins induce this abnormal persistence of macro-
(Fig. 1; recently reviewed in [80]). phages and T cells? It has been hypothesized that

478 www.co-lipidology.com Volume 27  Number 5  October 2016

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


The pathogenic simplicity of atherosclerosis Borén and Williams

specific cellular and molecular programs that cause and their affinity for arterial-wall proteoglycans.
immune cells to remain in place may have evolved as The affinity is modulated by the lipid and
a defense against Mycobacterium tuberculosis, an acid- protein composition of the lipoprotein particles.
fast bacillus that is not killed after phagocytosis and In particular, apoC-III on LDL increases its
would therefore be spread throughout the body by proteoglycan binding.
emigrating macrophages. Interestingly, retained and (3) The instability of retained lipoproteins, that is,
modified apoB-containing lipoproteins within the how readily they aggregate in the presence of
arterial wall elicit many of the same antiemigration arterial-wall enzymes, particularly S-SMase.
&
signals as M. tuberculosis does [6 ,11,84–86]. Unfortu- This property has been linked to LDL lipid com-
nately, the result is a crippling of the reticuloendo- position and is affected by diet.
thelial system, which otherwise has a huge capacity (4) The susceptibility of aggregated and retained
that could easily handle a few grams of intramural lipoprotein to further modifications within
cholesterol and other debris. the artery wall.
(5) The ability of the modified lipoproteins to
New data regarding possible roles for HDL induce a maladaptive cellular response.
in the response-to-retention model of
atherogenesis Thus, it can be hypothesized that patients with
The pathogenic importance in atherosclerosis of atherogenic apoB-lipoproteins characterized by high
plasma HDL-cholesterol (HDL-C) levels has come binding affinity to arterial proteoglycans, increased
into doubt, due to recent failures of clinical trials susceptibility to aggregate, and features that promote
of agents that raise plasma HDL-C concentrations a hyperresponsive maladaptive cellular infiltrate may
[87,88] and the lack of ASCVD protection from develop atherosclerosis despite only moderately elev-
human gene polymorphisms that raise plasma con- ated plasma levels of cholesterol. It remains to be
centrations of HDL-C [89]. In animal models, how- demonstrated whether assessments of these specific
ever, injections of HDL, overexpression of apoA-I, qualities of LDL and C-TRLs will add predictive power
and strategies to enhance HDL function delay beyond conventional risk factors or help identify
atherosclerosis progression and even promote plaque groups of patients for targeted therapies.
stabilization and regression [11,85,88]. In the
context of the response-to-retention hypothesis,
roles for HDL in every step have been hypothesized Reclassification of risk factors for
[87] – namely, interfering with the irreversible bind- atherosclerosis into causative agents,
ing of plasma LDL to arterial wall proteoglycans [90– exacerbators, and mere bystander
92], blocking SMase-induced aggregation of LDL phenomena
[79,93], removing toxic lipids, and resolving the Our understanding of the pathogenesis of ASCVD is
maladaptive inflammatory infiltrate [94–98]. Along based on laboratory investigation, human epidemi-
these lines, the apoA-I mimetic peptide 4F was ology, genetic studies, and clinical trials. Collec-
recently shown to block SMase-induced LDL aggre- tively, these studies have demonstrated beyond
gation and the increase in binding of the modified any doubt that cholesterol-rich apoB-lipoproteins
&
LDL particles to human aortic proteoglycans [99 ]. In are causally linked to atherogenesis and that low-
contrast, abnormal HDL or apoA-I within the arterial ering the plasma concentration of LDL is the most
&
wall may have adverse effects [96,100,101 ,102]. effective therapy to date against ASCVD. The critical
step is that cholesterol-rich apoB-lipoproteins from
plasma become pathogenic only after their reten-
Atherogenicity of apolipoprotein-B- tion within the arterial wall.
lipoproteins Using our extensive knowledge of the pathogen-
Based on our present understanding for how esis of atherosclerosis, we can now reclassify nearly all
cholesterol-rich apoB-lipoproteins induce athero- epidemiologic risk factors into causative agents,
sclerosis, the atherogenicity of apoB-containing exacerbating factors, and bystander phenomena
lipoproteins may depend on (Table 1) [80]. Causative and exacerbating factors
are targets for therapy, bystander phenomena are not.
(1) Their plasma concentrations, because high
levels of circulating apoB-lipoproteins increase
the probability that these particles will enter CONCLUSION
and be retained in the subendothelium. It is hard to unravel the root cause of a condition
(2) Their ability to become retained in the artery from an end-stage lesion. Atherosclerosis was no
wall. This is influenced by their size (<70 nm) exception. For more than a century and a half, many

0957-9672 Copyright ß 2016 Wolters Kluwer Health, Inc. All rights reserved. www.co-lipidology.com 479

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


Atherosclerosis: cell biology and lipoproteins

Table 1. Reclassification of human epidemiologic risk factors for ASCVD events into causative agents, exacerbating factors,
and mere bystander phenomena
Pathophysiologic
Epidemiologic risk factors for ASCVD reclassification Evidence from human RCTs or human MRS

Elevated plasma concentrations of total Causative LDL: proven in RCTs of statins and nonstatins and in MRS; Other
cholesterol, LDL-cholesterol, non-HDLc, apoB-lipoproteins: Proven in MRS
C-TRL remnants, and apoB
Tobacco smoking Exacerbating Lifetime low plasma LDL levels from genetics or unusual lifestyles
protect smokers from ASCVD events
Diabetes mellitus Exacerbating Likewise for diabetes: lifetime low LDL levels protect patients with
diabetes from ASCVD events
Hypertension Exacerbating Likewise for hypertension (but extreme hypertension still causes other
health problems)
Male sex (age) Exacerbating Likewise for elderly men, many of whom no longer develop clinically
significant ASCVD
Elevated plasma glucose levels per se Exacerbating Modest genetic elevations in plasma glucose in MRS detectably
increase ASCVD, but in the context of abundant plasma apoB-
lipoproteins
Low plasma HDLc concentrations Exacerbating or No benefit yet in RCTs of HDLc-raising treatments; no benefit of
bystander higher HDLc in MRS
Elevated plasma homocysteine Bystander Disproven in RCTs to lower plasma homocysteine levels
concentrations
Chlamydia pneumoniae infection; Bystander Disproven in RCTs of antibiotics
periodontal disease (gingivitis)
Elevated plasma C-reactive protein Bystander Disproven in MRS
Oxidized LDL Bystander Disproven in many large RCTs of antioxidant supplements.
Moreover, several components of the heart-healthy lifestyle are
pro-oxidant, such as exercise, dietary PUFAs, and moderate
alcohol consumption

ASCVD, atherosclerotic cardiovascular disease; C-TRL, cholesterol-rich and triglyceride-rich apoB-containing remnant lipoprotein; HDLc, HDL-cholesterol; MRS,
Mendelian randomization studies; RCTs, randomized controlled trials.

theories had been articulated to explain how a nor- (J.B. and K.J.W.), ALF-medel Västra Götalandsregionen
mal artery becomes atherosclerotic. As a testament (J.B. and K.J.W.), and the Ruth and Yonatan Ben-Avra-
to the importance of this area, most of these theories ham Fund (K.J.W.).
were tested in clinical trials. As a consequence,
it is now established that increased plasma Conflicts of interest
concentrations of cholesterol-rich apoB-containing Dr Williams reports purchasing stock in Gemphire
lipoproteins are causatively linked to ASCVD. Therapeutics, Inc., a company focused on the treatment
Accordingly, the most effective therapies against of dyslipidemias.
atherothrombotic cardiovascular disease – LDL-
lowering drugs – are based upon the principle that
decreasing the concentration of apoB-lipoproteins REFERENCES AND RECOMMENDED
in the circulaton decreases the probability that they READING
will enter and be retained in the subendothelium. Papers of particular interest, published within the annual period of review, have
been highlighted as
At this point, we know enough about the patho- & of special interest
physiology of atherosclerosis that eradication of this && of outstanding interest

major killer has become a genuine possibility. 1. Mihaylova B, Emberson J, Blackwell L, et al., Cholesterol Treatment Trialists
(CTT) Collaborators. The effects of lowering LDL cholesterol with statin
Acknowledgements therapy in people at low risk of vascular disease: meta-analysis of individual
data from 27 randomised trials. Lancet 2012; 380:581–590.
None. 2. Cannon CP, Blazing MA, Giugliano RP, et al. Ezetimibe added to statin
&& therapy after acute coronary syndromes. N Engl J Med 2015; 372:2387–
2397.
Financial support and sponsorship A seminal study demonstrating that a nonstatin, ezetimibe, that lowers plasma LDL
levels decreases atherosclerotic cardiovascular disease (ASCVD) events in hu-
The authors acknowledge support from the Swedish mans to exactly the same extent as would be expected from the same degree of
Research Council (J.B.), Swedish Heart-Lung Foundation LDL lowering from a statin.

480 www.co-lipidology.com Volume 27  Number 5  October 2016

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


The pathogenic simplicity of atherosclerosis Borén and Williams

3. Robinson JG, Farnier M, Krempf M, et al. Efficacy and safety of alirocumab in 30. Bartels ED, Christoffersen C, Lindholm MW, Nielsen LB. Altered metabolism
&& reducing lipids and cardiovascular events. N Engl J Med 2015; 372:1489– && of LDL in the arterial wall precedes atherosclerosis regression. Circ Res
1499. 2015; 117:933–942.
&& &&
Refs. [3 ,4 ] present initial data that another nonstatin, anti-PCSK9 antibodies, The authors show that rapid lowering of plasma concentrations of cholesterol-rich
that substantially lowers plasma LDL levels substantially decreases ASCVD events apoB-lipoproteins decreases the permeability of the endothelium overlying ather-
in humans. omata to LDL and decreases the fractional degradation of LDL that enters the
4. Sabatine MS, Giugliano RP, Wiviott SD, et al. Efficacy and safety of plaque. This finding may be important for our understanding of plaque stabilization
&& evolocumab in reducing lipids and cardiovascular events. N Engl J Med and atherosclerosis regression.
2015; 372:1500–1509. 31. Tran-Lundmark K, Tran PK, Paulsson-Berne G, et al. Heparan sulfate in
&& &&
Refs. [3 ,4 ] present initial data that another nonstatin, anti-PCSK9 antibodies, perlecan promotes mouse atherosclerosis: roles in lipid permeability, lipid
that substantially lowers plasma LDL levels substantially decreases ASCVD events retention, and smooth muscle cell proliferation. Circ Res 2008; 103:
in humans. 43–52.
5. Hegele RA, Gidding SS, Ginsberg HN, et al. Nonstatin low-density lipopro- 32. Borén J, Olin K, Lee I, et al. Identification of the principal proteoglycan-binding
& tein—lowering therapy and cardiovascular risk reduction—statement from site in LDL. A single-point mutation in apo-B100 severely affects proteogly-
ATVB Council. Arterioscler Thromb Vasc Biol 2015; 35:2269–2280. can interaction without affecting LDL receptor binding. J Clin Invest 1998;
An important overview of historical and recent data that LDL lowering by nonstatins 101:2658–2664.
decreases ASCVD events in humans. 33. Flood C, Gustafsson M, Richardson PE, et al. Identification of the proteo-
6. Williams KJ, Tabas I, Fisher EA. How an artery heals. Circ Res 2015; glycan binding site in apolipoprotein B48. J Biol Chem 2002; 277:32228–
& 117:909–913. 32233.
&&
A commentary on Bartels ED, Christoffersen C, et al. 2015 (reference [30 ]). 34. Camejo G, Fager G, Rosengren B, et al. Binding of low density lipoproteins
7. Steinberg D. Thematic review series: the pathogenesis of atherosclerosis. An by proteoglycans synthesized by proliferating and quiescent human arterial
interpretive history of the cholesterol controversy: Part I. J Lipid Res 2004; smooth muscle cells. J Biol Chem 1993; 268:14131–14137.
45:1583–1593. 35. Tannock LR. Proteoglycan-LDL interactions: a novel therapeutic target?
8. Capron L. Pathogenie de l’atherosclerose: mises à jour des trois theories Atherosclerosis 2014; 233:232–233.
dominantes. Ann Cardiol Angeiol (Paris) 1989; 38:631–634. 36. Skålén K, Gustafsson M, Rydberg EK, et al. Subendothelial retention of
9. Williams KJ, Tabas I. The response-to-retention hypothesis of early ather- atherogenic lipoproteins in early atherosclerosis. Nature 2002; 417:750–
ogenesis. Arterioscler Thromb Vasc Biol 1995; 15:551–561. 754.
10. Ventura HO. Profiles in cardiology. Rudolph Virchow and cellular pathology. 37. Vikramadithyan RK, Kako Y, Chen G, et al. Atherosclerosis in perlecan
Clin Cardiol 2000; 23:550–552. heterozygous mice. J Lipid Res 2004; 45:1806–1812.
11. Williams KJ, Tabas I. Lipoprotein retention – and clues for atheroma 38. Huang F, Thompson JC, Wilson PG, et al. Angiotensin II increases vascular
regression. Arterioscler Thromb Vasc Biol 2005; 25:1536–1540. proteoglycan content preceding and contributing to atherosclerosis devel-
12. Anitschkow N, Chalatow S. Über experimentelle Cholesterinsteatose und opment. J Lipid Res 2008; 49:521–530.
ihre Bedeutung für die Entstehung einiger pathologischer Prozesse. Zentrbl 39. Tang T, Wilson PG, Thompson JC, et al. Prevention of TGFß induction
Allg Pathol Pathol Anat 1913; 24:1–9. attenuates angII-stimulated vascular biglycan and atherosclerosis in Ldlr /
13. Konstantinov IE, Mejevoi N, Anichkov NM, Nikolai N. Anichkov and his theory mice. J Lipid Res 2013; 54:2255–2264.
of atherosclerosis. Tex Heart Inst J 2006; 33:417–423. 40. She ZG, Chang Y, Pang HB, et al. NG2 ablation reduces low-density
14. Faber M. The human aorta; sulfate-containing polyuronides and the deposi- && lipoprotein retention of synthetic smooth muscle cells and atherogenesis.

tion of cholesterol. Arch Pathol (Chic) 1949; 48:342–350. Arterioscler Thromb Vasc Biol 2016; 36:49–59.
15. Camejo G, Lopez A, Vegas H, Paoli H. The participation of aortic proteins in The authors describe a novel role for NG2-positive s–smooth muscle cells in
the formation of complexes between low density lipoproteins and intima- trapping LDL and then presenting this LDL to macrophages. Despite worse
media extracts. Atherosclerosis 1975; 21:77–91. obesity and worse hyperlipidemia, double knockout mice (Ng2 / /Apoe / )
16. Iverius P-H. The interaction between human plasma lipoproteins and con- developed less atherosclerosis than did Apoe / controls. The authors show that
nective tissue glycosaminoglycans. J Biol Chem 1972; 247:2607–2613. NG2 has the capacity to bind LDL through hydrophobic interactions. These
17. Vijayagopal P, Srinivasan SR, Radhakrishnamurthy B, Berenson GS. Inter- features make NG2 unique amongst LDL-binding proteoglycans and may be
action of serum lipoproteins and a proteoglycan from bovine aorta. J Biol related to its normal function in the arterial intima.
Chem 1981; 256:8234–8241. 41. Fogelstrand P, Borén J. Catch and release: NG2-coated vascular smooth
18. Smith EB, Slater RS. Lipids and low density lipoproteins in intima in relation to & muscle cells capture lipoproteins for macrophages. Arterioscler Thromb
its morphological characteristics. Ciba Found Symp 1973; 12:39–62. Vasc Biol 2016; 36:7–8.
19. Tamminen M, Mottino G, Qiao JH, et al. Ultrastructure of early lipid accu- A commentary on She et al. 2016 [40&&].
mulation in ApoE-deficient mice. Arterioscler Thromb Vasc Biol 1999; 42. Bostrom MA, Boyanovsky BB, Jordan CT, et al. Group V secretory phos-
19:847–853. pholipase A2 promotes atherosclerosis: evidence from genetically altered
20. Schwenke DC, Carew TE. Initiation of atherosclerotic lesions in cholesterol- mice. Arterioscler Thromb Vasc Biol 2007; 27:600–606.
fed rabbits. II. Selective retention of LDL vs. selective increases in LDL 43. Öörni K, Kovanen PT. PLA2-V: a real player in atherogenesis. Arterioscler
permeability in susceptible sites of arteries. Arteriosclerosis 1989; 9:908– Thromb Vasc Biol 2007; 27:445–447.
918. 44. Gustafsson M, Levin M, Skålén K, et al. Retention of low-density lipoprotein in
21. Nakashima Y, Fujii H, Sumiyoshi S, et al. Early human atherosclerosis: atherosclerotic lesions of the mouse: evidence for a role of lipoprotein lipase.
accumulation of lipid and proteoglycans in intimal thickenings followed by Circ Res 2007; 101:777–783.
macrophage infiltration. Arterioscler Thromb Vasc Biol 2007; 27:1159– 45. Devlin CM, Leventhal AR, Kuriakose G, et al. Acid sphingomyelinase pro-
1165. motes lipoprotein retention within early atheromata and accelerates lesion
22. Borén J, Gustafsson M, Skålén K, et al. Role of extracellular retention of low progression. Arterioscler Thromb Vasc Biol 2008; 28:1723–1730.
density lipoproteins in atherosclerosis. Curr Opin Lipidol 2000; 11:451– 46. Ruuth M, Nguyen SD, Vihervaara T, et al. Unstable LDL – novel mechanism of
456. atherogenesis and link to cardiovascular deaths. 15 April 2016, Krogerup
23. Sloop CH, Dory L, Roheim PS. Interstitial fluid lipoproteins. J Lipid Res 1987; Højskole, Humlebæk, Denmark: Oral presentation at the 22nd annual Scan-
28:225–237. dinavian atherosclerosis conference, Scandinavian Society for Atherosclero-
24. Frank PG, Pavlides S, Cheung MW, et al. Role of caveolin-1 in the regulation sis Research (SSAR); 2016.
of lipoprotein metabolism. Am J Physiol Cell Physiol 2008; 295:C242–248. 47. Coronado-Gray A, van Antwerpen R. The physical state of the LDL core
25. Fernández-Hernando C, Yu J, Suárez Y, et al. Genetic evidence supporting a influences the conformation of apolipoprotein B-100 on the lipoprotein
critical role of endothelial caveolin-1 during the progression of atherosclero- surface. FEBS Lett 2003; 533:21–24.
sis. Cell Metab 2009; 10:48–54. 48. Melchior JT, Sawyer JK, Kelley KL, et al. LDL particle core enrichment in
26. Armstrong SM, Sugiyama MG, Fung KY, et al. A novel assay uncovers an && cholesteryl oleate increases proteoglycan binding and promotes athero-

unexpected role for SR-BI in LDL transcytosis. Cardiovasc Res 2015; sclerosis. J Lipid Res 2013; 54:2495–2503.
108:268–277. The authors tested the hypothesis that the increased atherosclerosis associated
27. Kraehling JR, Rajagopal C, Sessa WC. Identification of novel mechanisms of with LDL core enrichment in cholesteryl oleate results from an increased affinity of
LDL uptake into endothelium. Naunyn-Schmiedeberg’s Arch Pharmacol the LDL particle for arterial proteoglycans. Results strongly support their hypoth-
2014; 387 (Suppl 1):S12; Presented at the 80th Annual meeting, Deutsche esis. ‘This is an important observation when considering that monounsaturated fats
Gesellschaft für Experimentelle und Klinische Pharmakologie und Toxikolo- such as olive oil are still being promoted as healthy and protective against heart
gie e.V. [German Society for Experimental and Clinical Pharmacology and disease’, state the authors.
Toxicology (registered association)], 1–3 April 2014, Hannover, Germany. 49. Flood C, Gustafsson M, Pitas RE, et al. Molecular mechanism for changes in
28. Nielsen LB, Stender S, Jauhiainen M, Nordestgaard BG. Preferential influx proteoglycan binding on compositional changes of the core and the surface
and decreased fractional loss of lipoprotein(a) in atherosclerotic compared of low-density lipoprotein-containing human apolipoprotein B100. Arterios-
with nonlesioned rabbit aorta. J Clin Invest 1996; 98:563–571. cler Thromb Vasc Biol 2004; 24:564–570.
29. Nielsen LB, Gronholdt ML, Schroeder TV, et al. In vivo transfer of lipopro- 50. Borén J, Watts GF, Adiels M, et al. Kinetic and related determinants of plasma
tein(a) into human atherosclerotic carotid arterial intima. Arterioscler Thromb triglyceride concentration in abdominal obesity: multicenter tracer kinetic
Vasc Biol 1997; 17:905–911. study. Arterioscler Thromb Vasc Biol 2015; 35:2218–2224.

0957-9672 Copyright ß 2016 Wolters Kluwer Health, Inc. All rights reserved. www.co-lipidology.com 481

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


Atherosclerosis: cell biology and lipoproteins

51. Olin-Lewis K, Krauss RM, La Belle M, et al. ApoC-III content of apoB- 72. Hassing HC, Mooij H, Guo S, et al. Inhibition of hepatic sulfatase-2 in vivo: a
containing lipoproteins is associated with binding to the vascular proteogly- novel strategy to correct diabetic dyslipidemia. Hepatology 2012; 55:1746–
can biglycan. J Lipid Res 2002; 43:1969–1977. 1753.
52. Davidsson P, Hulthe J, Fagerberg B, et al. A proteomic study of the 73. Blom DJ, Hala T, Bolognese M, et al. A 52-week placebo-controlled trial of
apolipoproteins in LDL subclasses in patients with the metabolic syndrome evolocumab in hyperlipidemia. N Engl J Med 2014; 370:1809–1819.
and type 2 diabetes. J Lipid Res 2005; 46:1999–2006. 74. Holman RR, Koren MJ, Roth E, et al. Evaluation of the glycemic effects and
53. Hiukka A, Ståhlman M, Pettersson C, et al. ApoCIII-enriched LDL in type 2 efficacy and safety of evolocumab (AMG 145) in subjects with or without
diabetes displays altered lipid composition, increased susceptibility for dysglycemia or metabolic syndrome. Diabetes 2015; 64 (suppl. 1):A68; Oral
sphingomyelinase, and increased binding to biglycan. Diabetes 2009; presentation at the 75th Scientific Sessions of the American Diabetes
58:2018–2026. Association, Chicago, IL, USA, 7 June 2015.
54. Didangelos A, Mayr U, Monaco C, Mayr M. Novel role of ADAMTS-5 in 75. Tabas I, Li Y, Brocia RW, et al. Lipoprotein lipase and sphingomyelinase
proteoglycan turnover and lipoprotein retention in atherosclerosis. J Biol synergistically enhance the association of atherogenic lipoproteins with
Chem 2012; 287:19341–19345. smooth muscle cells and extracellular matrix. A possible mechanism for
55. Seidelmann SB, Kuo C, Pleskac N, et al. Athsq1 is an atherosclerosis low density lipoprotein and lipoprotein(a) retention and macrophage foam
modifier locus with dramatic effects on lesion area and prominent accumula- cell formation. J Biol Chem 1993; 268:20419–20432.
tion of versican. Arterioscler Thromb Vasc Biol 2008; 28:2180–2186. 76. Cohen JC, Boerwinkle E, Mosley TH Jr, Hobbs HH. Sequence variations in
56. Tang T, Thompson JC, Wilson PG, et al. Biglycan deficiency: increased aortic PCSK9, low LDL, and protection against coronary heart disease. N Engl J
& aneurysm formation and lack of atheroprotection. J Mol Cell Cardiol 2014; Med 2006; 354:1264–1272.
75:174–180. 77. Nissen SE, Tardif JC, Nicholls SJ, et al. Effect of torcetrapib on the
Regulation of vascular proteoglycans seems to be coordinated because mice progression of coronary atherosclerosis. N Engl J Med 2007; 356:1304–
made genetically deficient in biglycan exhibit increased vascular perlecan content. 1316.
57. Nakashima Y, Wight TN, Sueishi K. Early atherosclerosis in humans: role of 78. Öörni K, Rajamäki K, Nguyen SD, et al. Acidification of the intimal fluid: the
diffuse intimal thickening and extracellular matrix proteoglycans. Cardiovasc & perfect storm for atherogenesis. J Lipid Res 2015; 56:203–214.
Res 2008; 79:14–23. In this review, the authors summarize the effects of acidic extracellular pH on
58. Maus F, Sakry D, Biname F, et al. The NG2 proteoglycan protects oligoden- atherogenesis, focusing on subendothelial retention of apoB-containing lipopro-
drocyte precursor cells against oxidative stress via interaction with OMI/ teins, altered macrophage biology, and abnormalities in HDL particles. Acidity
HtrA2. PLoS One 2015; 10:e0137311. enhances lipolytic, proteolytic, and other modifications of LDL and other apoB-
59. Matsumoto H, Kumon Y, Watanabe H, et al. Accumulation of macrophage- containing lipoproteins, and strongly increases their affinity for proteoglycans, and
like cells expressing NG2 proteoglycan and Iba1 in ischemic core of rat brain may thus accelerate their retention and the ensuing maladaptive cellular responses
after transient middle cerebral artery occlusion. J Cereb Blood Flow Metab within the arterial intima.
2008; 28:149–163. 79. Sneck M, Nguyen SD, Pihlajamaa T, et al. Conformational changes of apoB-
60. Peiffer V, Sherwin SJ, Weinberg PD. Does low and oscillatory wall shear 100 in SMase-modified LDL mediate formation of large aggregates at acidic
stress correlate spatially with early atherosclerosis? A systematic review. pH. J Lipid Res 2012; 53:1832–1839.
Cardiovasc Res 2013; 99:242–250. 80. Williams KJ, Fisher EA. Apolipoprotein-B: the crucial protein of atherogenic
61. Kwak BR, Bäck M, Bochaton-Piallat ML, et al. Biomechanical factors in lipoproteins. In: Wang H, Patterson C, editors. Atherosclerosis: risks,
atherosclerosis: mechanisms and clinical implications. Eur Heart J 2014; mechanisms, & therapies. Hoboken, NJ, USA: John Wiley & Sons, Inc.;
35:3013–3020. [ESC Position Paper] 2015. pp. 291–312.
62. Steffensen LB, Mortensen MB, Kjolby M, et al. Disturbed laminar blood flow 81. Deevska GM, Sunkara M, Morris AJ, Nikolova-Karakashian MN. Character-
&& vastly augments lipoprotein retention in the artery wall: a key mechanism ization of secretory sphingomyelinase activity, lipoprotein sphingolipid con-
distinguishing susceptible from resistant sites. Arterioscler Thromb Vasc Biol tent and LDL aggregation in ldlr / mice fed on a high-fat diet. Biosci Rep
2015; 35:1928–1935. 2012; 32:479–490.
The authors provide experimental evidence for a causal chain linking disturbed 82. Tabas I, Williams KJ, Borén J. Subendothelial lipoprotein retention as the
laminar flow patterns to increased arterial matrix deposition and then increased initiating process in atherosclerosis: update and therapeutic implications.
apoB-lipoprotein retention and atherosclerosis. This study contradicts the notion Circulation 2007; 116:1832–1844.
of a key role for disturbed endothelial permeability to LDL in early phases of 83. Lao KH, Zeng L, Xu Q. Endothelial and smooth muscle cell transformation in
atherogenesis. atherosclerosis. Curr Opin Lipidol 2015; 26:449–456.
63. Brito V, Mellal K, Portelance SG, et al. Induction of antianti-idiotype anti- 84. Moore KJ, Sheedy FJ, Fisher EA. Macrophages in atherosclerosis: a dynamic
bodies against sulfated glycosaminoglycans reduces atherosclerosis in balance. Nat Rev Immunol 2013; 13:709–721.
apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2012; 85. Williams KJ, Feig JE, Fisher EA. Rapid regression of atherosclerosis: insights
32:2847–2854. from the clinical and experimental literature. Nat Clin Pract Cardiovasc Med
64. Delgado-Roche L, Brito V, Acosta E, et al. Arresting progressive athero- 2008; 5:91–102.
& sclerosis by immunization with an antiglycosaminoglycan monoclonal anti- 86. Bracho-Sanchez E, Ramkhelawon B, Desvignes L, et al. Induction of neural
body in apolipoprotein E-deficient mice. Free Radic Biol Med 2015; guidance molecule expression in macrophages and dendritic cells by My-
89:557–566. cobacterium tuberculosis. J Immunol 2013; 190 (Suppl 1):55.9.
The authors have earlier reported the preventive antiatherogenic properties of an 87. Williams KJ. What does HDL do? A new mechanism to slow atherogenesis –
antichondroitin sulfate antibody response. Results in this study further demon- but a new problem in type 2 diabetes mellitus. Atherosclerosis 2012;
strate the use of this antiglycosaminoglycan antibody-based immunotherapy as a 225:36–38.
novel approach to target apoB-lipoprotein retention, and hence atherosclerosis, at 88. Hewing B, Moore KJ, Fisher EA. HDL and cardiovascular risk: time to call the
different phases of progression. plumber? Circ Res 2012; 111:1117–1120.
65. Moreton JR. Atherosclerosis and alimentary hyperlipemia. Science 1947; 89. Voight BF, Peloso GM, Orho-Melander M, et al. Plasma HDL cholesterol and
106:190–191. risk of myocardial infarction: a mendelian randomisation study. Lancet 2012;
66. Zilversmit DB. Atherogenesis: a postprandial phenomenon. Circulation 380:572–580.
1979; 60:473–485. 90. Bihari-Varga M. Influence of serum high density lipoprotein on the low density
67. Williams KJ, Wu X. Imbalanced insulin action in chronic over nutrition: clinical lipoprotein-aortic glycosaminoglycan interactions. Artery 1978; 4:504–509.
& harm, molecular mechanisms, and a way forward. Atherosclerosis 2016; 91. Camejo G, Cortez MM, Lopez F, et al. Factors modulating the interaction of
247:225–282. LDL with an arterial lipoprotein complexing proteoglycan: the effect of HDL.
A comprehensive overview of imbalanced insulin action, also known as pathway- Acta Med Scand Suppl 1980; 642:159–164.
selective insulin resistance and responsiveness, in the pathogenesis of the 92. Umaerus M, Rosengren B, Fagerberg B, et al. HDL2 interferes with LDL
atherometabolic syndrome and type 2 diabetes mellitus, including the character- association with arterial proteoglycans: a possible athero-protective effect.
istic dyslipoproteinemia and accelerated ASCVD risk in these disorders. Atherosclerosis 2012; 225:115–120.
68. Taskinen MR, Borén J. New insights into the pathophysiology of dyslipidemia 93. Schissel SL, Tweedie-Hardman J, Rapp JH, et al. Rabbit aorta and human
& in type 2 diabetes. Atherosclerosis 2015; 239:483–495. atherosclerotic lesions hydrolyze the sphingomyelin of retained low-density
A review of kinetic studies of dyslipoproteinemia in the atherometabolic syndrome, lipoprotein. Proposed role for arterial-wall sphingomyelinase in subendothe-
implicating overproduction of cholesterol-rich and triglyceride-rich apoB-contain- lial retention and aggregation of atherogenic lipoproteins. J Clin Invest 1996;
ing remnant lipoproteins but more importantly an impairment in their clearance from 98:1455–1464.
the circulation by the liver. 94. De Nardo D, Labzin LI, Kono H, et al. High-density lipoprotein mediates anti-
69. Williams KJ, Chen K. Recent insights into factors affecting remnant lipopro- inflammatory reprogramming of macrophages via the transcriptional regu-
tein uptake. Curr Opin Lipidol 2010; 21:218–228. lator ATF3. Nat Immunol 2014; 15:152–160.
70. Varbo A, Benn M, Tybjærg-Hansen A, et al. Remnant cholesterol as a causal 95. Moore KJ, Fisher EA. High-density lipoproteins put out the fire. Cell Metab
risk factor for ischemic heart disease. J Am Coll Cardiol 2013; 61:427–436. 2014; 19:175–176.
71. Ridker PM, Danielson E, Fonseca FA, et al. Rosuvastatin to prevent vascular 96. Hewing B, Parathath S, Barrett T, et al. Effects of native and myeloperox-
events in men and women with elevated C-reactive protein. N Engl J Med idase-modified apolipoprotein A-I on reverse cholesterol transport and
2008; 359:2195–2207. atherosclerosis in mice. Arterioscler Thromb Vasc Biol 2014; 34:779–789.

482 www.co-lipidology.com Volume 27  Number 5  October 2016

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.


The pathogenic simplicity of atherosclerosis Borén and Williams

97. Niyonzima N, Samstad EO, Aune MH, et al. Reconstituted high-density 100. Chiba T, Chang MY, Wang S, et al. Serum amyloid A facilitates the binding
lipoprotein attenuates cholesterol crystal-induced inflammatory responses of high-density lipoprotein from mice injected with lipopolysaccharide
by reducing complement activation. J Immunol 2015; 195:257–264. to vascular proteoglycans. Arterioscler Thromb Vasc Biol 2011;
98. Bursill CA, Castro ML, Beattie DT, et al. High-density lipoproteins suppress 31:1326–1332.
chemokines and chemokine receptors in vitro and in vivo. Arterioscler 101. Huang Y, DiDonato JA, Levison BS, et al. An abundant dysfunctional
Thromb Vasc Biol 2010; 30:1773–1778. & apolipoprotein A1 in human atheroma. Nat Med 2014; 20:193–
99. Nguyen SD, Javanainen M, Rissanen S, et al. Apolipoprotein A-I mimetic 203.
& peptide 4F blocks sphingomyelinase-induced LDL aggregation. J Lipid Res Evidence that apoA-I modified by a specific enzyme, myeloperoxidase, in human
2015; 56:1206–1221. atheromata may be harmful.
The authors evaluated effects of the apoA-I mimetic peptide 4F on sphingomye-
linase (SMase)-modified LDL particles. The results demonstrated that 4F stabilizes 102. Nicholls S, Ray K, Ballantyne C, et al. Comparative effects of cholesteryl ester
LDL particles by preventing SMase-induced conformational changes in apoB-100, transfer protein inhibition, statin and ezetimide therapy on atherogenic and
thereby blocking SMase-induced LDL aggregation and the resulting increase in protective lipid factors: the Accentuate Trial. Innsbruck: 84th European
LDL retention. Atherosclerosis Society Congress; 2016 .

0957-9672 Copyright ß 2016 Wolters Kluwer Health, Inc. All rights reserved. www.co-lipidology.com 483

Copyright © 2016 Wolters Kluwer Health, Inc. All rights reserved.

Das könnte Ihnen auch gefallen