ATEROESCLEROSIS
ATEROESCLEROSIS
ATEROESCLEROSIS
doi:10.1152/physrev.00004.2012
INTRODUCTION
OVERVIEW OF ATHEROGENESIS
FUNDAMENTAL SIGNALING...
INITIATION OF ATHEROSCLEROSIS:...
INITIATION OF ATHEROSCLEROSIS:...
INITIATION OF ATHEROSCLEROSIS:...
CYTOKINES AND INNATE VERSUS...
EFFECTS OF SELECTED RISK FACTORS...
PROMOTION OF ATHEROSCLEROSIS
PROGRESSION OF ATHEROSCLEROSIS
FURTHER PLAQUE DESTABILIZATION...
PERSPECTIVES
1317
1319
1320
1355
1383
1392
1403
1430
1438
1454
1483
1484
I. INTRODUCTION
During the past 10 15 years, understanding of the molecular biology of atherosclerosis and related signaling pathways has progressed so rapidly that the field has become
essentially unrecognizable to most clinicians, many students of the life sciences, and even veteran investigators in
related disciplines, a lamentable knowledge gap that undoubtedly hampers progress in the field. For those interested persons who find themselves in this situation, this
review is intended to fill a portion of that gap, which may
have formed from lack of formal training, inattention to the
field, or merely the passage of time.
Much of the text may be considered as an introductory
primer or overview for a number of the signaling pathways
now central to investigations into the molecular biology of
1317
I.
II.
III.
IV.
V.
VI.
VII.
VIII.
IX.
X.
XI.
XII.
PAUL N. HOPKINS
With the use of yeast as a eukaryotic test organism, a remarkable 73% of 6,000 genes making up the yeast genome
were found unnecessary for survival (when tested one gene
at a time) and only 50% affected growth even in extreme
test conditions (613). When tested two genes at a time,
extensive interactive networks can be identified. One study
reported 117,000 significant interactions affecting yeast
growth out of 5.4 million potential interactions among the
1,700 genes tested. Informatics techniques allowed organization of many of these apparent interactions into overlapping networks with known functional utilities (RNA processing, mitochondrial activity, etc.) (352). In this study,
there were 39 interactions per gene on average and for only
10 genes were no interactions identified (C. L. Myers, personal communications).
Networks can act to buffer defects in single genes, distributing their effects to closely interacting network neighbors.
1318
350
200
150
100
50
0
13
While considering a number of these pathways, this reviewer was struck by one remarkable feature: redundancy.
In a redundant system, a duplicate or similar element may
take over the function of another element or provide a
somewhat different but overlapping utility. Often built into
cleverly designed mechanical or informatics systems, redundancy can ensure the operation of critically important activities even when one or more key components fail. In
living systems, the phenomenon of gene duplication, allowing the original or duplicated gene to diverge somewhat or
greatly in function, provides a mechanism for redundancy
to arise naturally. Alternate transcriptional start sites and
particularly alternate splice sites are found in 75% of
human genes and provide numerous isoforms with analogous flexibility of expression (861). The multiplicity of cytokines and multiple isoforms of various enzymes (such as
the thioredoxin and glutaredoxin systems) certainly illustrate redundancy, often showing overlapping if not entirely
duplicated functions. However, rather than simple linear
pathways with a few key back-up elements, the notion of
redundancy is taken to an entirely new level when considering the series of enormously interacting, branching networks that better describe living systems.
1319
PAUL N. HOPKINS
and take on the VSMC phenotype once in the plaque
(1755). Surprisingly, the well-studied cellular make-up of
atherosclerotic lesions in animal models, including VSMClike cells and various immune cells, has only recently been
confirmed in human plaques (1933), demonstrating general
comparability.
As the lipid-rich plaque progresses, accumulating macrophages (and other migrating cells) as well as activated endothelial cells secrete a host of proinflammatory cytokines,
matrix metalloproteinases (MMPs), and cathepsins, causing plaque fragility. IFN- strongly inhibits collagen formation by VSMC, further weakening the plaque and newly
forming fibrous cap (1042). The result can be catastrophic
thrombosis and downstream tissue infarction; but more often, there is limited mural thrombosis with subsequent organization of the thrombus, leading to saltatory growth of
lesions with relatively high fibrous tissue content. Other
precipitating changes in the plaque include erosions and,
more especially, eruption through the endothelium of underlying cholesterol crystals (4). Prior to such episodes of
thrombosis, there may be little, if any, encroachment of the
plaque into the arterial lumen due to outward remodeling of
the arterial wall to accommodate the growing plaque.
In reference to the tables that accompany the following
sections, the percentage increase or decrease in atherosclerosis reported in the table represents differences in the extent of atherosclerosis in the genetically manipulated or
targeted animals compared with control animals at the
same point in time. The length of time of the experiments
varies widely, and these times have only occasionally been
noted in the tables. This limitation is balanced somewhat by
utilizing atherosclerosis extent measured at the same point
in time in the manipulated animals and their controls. As
nearly all the studies cited were conducted in animals,
mostly mice, relevance to the human condition may at times
be questioned. Recent reviews regarding the relevance of
animal models to humans note some limitations but empha-
1320
1321
apoptosis. It is tempting to speculate that one of the antiatherogenic aspects of INSR and IGF-IR signaling relate to
inhibition of apoptosis. Other aspects of insulin signaling
likely contribute. For example, insulin was also found to
enhance endothelial cell-cell barrier function at adherens
junctions, and this could be seen as anti-inflammatory
(672).
PAUL N. HOPKINS
Insulin
O2
NOX1/2
Other?
Insulin receptor
GTP
Gab1
P
P
P
P
P
P
P
P
GDP Ras
(Shc) Grb2
IRS1/2
Sos
SHP2
GTP Ras
Src
PLC
Other adapters,
and targets
PIP2
PIP2
GTP Rac1
PIP3
SH2B1
RACK1
JAK2
STAT3
PIP3
O2
GDP Rac1
CIC3
O2
SOD1
Other effects
MAPK pathway
DAG
Other
effects
PKC
PDK
?Gef
IP3
PTP
SHP2
PIP3
P
P
P
P
Src
PTEN
MTM1
P
P
P
P
Ca2+
PKC
H2O2
ER
IP3R
PTP
Other effects
PTEN
MKP1
NOX activation
p47phox
GTP Rac1
CCND1
PIP3
PKB
GSK3
eIF2B
eIF2
4E-BP
PHLPP
Rheb
eNOS
activation,
anti-apoptotic
effects
eIF4E
mTORC1
mTOR
Protein sythesis
Cell growth
S6K1
S6
TSC1
TSC2
FOXO1,3
AMPK
Autophagy
14-3-3
SIRT1
Nucleus
IRS1/2, iNOS, p21, p27Kip1, FASL, BIM, Noxa, MnSOD, GADD45, Egr1
FOXO1,3
FOXO1,3:
cell cycle progression
inflammation
apoptosis
antioxidants
FIGURE 2. Receptor tyrosine kinase signaling as illustrated by INSR (insulin receptor). Overall, cellular effects
are dependent on simultaneous activation of multiple pathways. The multiplicity of activation pathways is
achieved, in part, by binding of several different adaptor proteins (shown in amber) to either INSR or IRS1/2.
Not shown are specific pathways leading to increased expression of GLUT4 by insulin (as occurs primarily in
muscle and adipose). CCND1, cyclin D1; MTM1, myotubular myopathy 1, also known as myotubularin. See text
for other abbreviations.
1322
Other
targets
Table 1. Genes related to fundamental signaling pathways present in multiple cell types (78 genes tested)
Gene
Model
% A
Function, Comment
INS2/ Akita in
LDLR/
1224% males,
130%
females
INS2/ Akita in
apoE/
1211%
Congenic 6T allele in
apoE/
188%
PAPP-A
PAPP-A/ apoE/
270%
INSR
SMC-specific Tg PAPP-A
o in apoE/
ESMIRO apoE/
1250%
152%
145%
MphIRKO in apoE/
Macrophage-specific
IGF-IR/ in apoE/
250%
157%
FGFR2
Endothelial cell-specific
Tg of constitutively
active FGFR2 in
apoE/
169%
IRS1
INSR/ IRS1/
apoE/
199%
IGF-IR
Continued
1323
IGF-I
PAUL N. HOPKINS
Table 1.Continued
Gene
IRS2
Model
IRS2/ apoE/
FLC IRS2/ into
apoE/
Function, Comment
160
238%
IRS2/ apoE/
128%
142%
PI3K (PIK3CG)
EC-specific Gab1/
apoE/
p110/ apoE/
Akt1
Akt1/ apoE/
152%
FOXO1
FOXO1KR/KR LDLR/
139%
FOXO3
EC-specific FOXO1/,
FOXO3/, and
FOXO4/ in LDLR/
277%
FOXO4
FOXO4/ apoE/
1128%
HDAC3
shHDAC3 apoE/
1280%
SIRT1
SIRT1/ apoE/
1112%
Gab1
252%
Continued
1324
% A
Table 1.Continued
Gene
Model
% A
PKC/ apoE/
275%
PKC3
p47phox
PKC3/ apoE/
p47phox/ apoE/
250%
270%
NOX2 (gp91phox)
NOX2/y apoE/
248%
NOX1
NOX1/y apoE/
228%
PTEN
PTEN/ apoE/
G6PDH
G6PDH/y apoE/
243%
AMPK2
AMPK2/ LDLR/
148%
NS
MAPK-related pathways
/
Grb2
apoE
Grb2
Grb2/ BMT
and
ASK1
ASK1/ apoE/
AIP1
AIP1/ apoE/
p38
macrophage-specific
p38 deletion (Cre-lox)
in apoE/ mice
JNK1 (MAPK8)
JNK1/ apoE/
243%
1131%
186%
114% (NS)
NS
Continued
1325
PKC
Function, Comment
PAUL N. HOPKINS
Table 1.Continued
Gene
Model
% A
JNK2/ apoE/
253%
Egr1
Egr1/ apoE/
286%
MK2
MK2/ LDLR/
256%
MKP-1
MKP-1/ apoE/
250%
NR4A1
NR4A1/ BMT in
apoE/
1114%
ADAM17FVB/FVB
LDLR/
250%
TNF-
TNF-/ apoE/
260%
LT
LT/ C57BL/6
262%
Continued
1326
JNK2 (MAPK9)
Function, Comment
Table 1.Continued
Gene
Model
% A
1130%
TNFR1/ carotid
arteries transplanted
into apoE/ mice
270%
TNFR2/ atherogenic
diet fed C57BL/6J
mice
NS
TNFR2/ apoE/
243%
TRAF1
TRAF1/ LDLR/
232%
TRAF5
TRAF5/ LDLR/
144%
NEMO
23047%
Macrophage-specific
IKK2 deletion (Cre-lox)
in LDLR/ mice
162%
Endothelial-specific,
degradation-resistant
IB in apoE/
260%
190%
TNFR2
IKK2
IB
cIAP2
1131%
247%
Continued
1327
TNFR1/ atherogenic
diet fed C57BL/6J
mice
TNFR1
Function, Comment
PAUL N. HOPKINS
Table 1.Continued
Gene
Model
% A
NF-B1
241%
NR4A3
NR4A3/ apoE/
252%
A20 (TNFAIP3)
A20/ apoE/
160%
GR
NS
Function, Comment
SCD1
SCD1
LDLR
LYPLA3 (PLA2G15)
LYPLA3/ apoE/
GLA
GLA/0 apoE/
178%
178%
1104%
NF-B signaling including activation by IL-1 and other TNF superfamily receptors
IL-1Ra
IL-1Ra Tg LDLR/
240%
IL-1
IL-1/ C57BL/6
256%
IL-1
IL-1/ apoE/
233%
IL-1R1
IL-1R1/ apoE/
293%
Continued
1328
Table 1.Continued
Gene
Model
% A
MyD88
MyD88/ apoE/
273%
IRAK4
289%
OPG/ apoE/
127%
TRAF6
OPG (TNFRSF11B)
NS
Function, Comment
1300%
266%
ACE
243%
ACE2
258%
ACE2/y apoE/
1325%
ACE2/y LDLR/
138%
AT1R
AT1aR/ apoE/
254%
AT2R
223%
ROCK1
260%
ROCK2
245%
PYK2
PYK2/ apoE/
266%
Continued
1329
Renin
human AGT/renin Tg
C57BL/6J mouse
renin/ BMT in
LDLR/
PAUL N. HOPKINS
Table 1.Continued
Gene
Model
% A
p21Cip1/ apoE/
253%
STAT3
endothelial-specific
STAT3/ C57BL/6
mice fed an
atherogenic diet
264%
GRK2
GRK2/ BMT in
LDLR/
231%
ARRB2
ARRB2/ LDLR/
240%
Bcl10
266%
APLN
APLN/ apoE/
150%
APLNR
APLNR/ apoE/
234%
HSD11B2
HSD11B2/ apoE/
11719%
Continued
1330
p21Cip1 (CDKN1A)
Function, Comment
Table 1.Continued
Gene
Model
% A
Function, Comment
1109%
1331
This and most of the following tables detail gene knockout (KO) or transgenic (Tg) overexpression studies showing effects
on atherogenesis. Often a high-saturated-fat, high-cholesterol diet (Western diet) is fed, but this is not always indicated.
The Paigen diet adds cholate which promotes greater lipid elevations and atherogenicity. If available, the effect on
atherosclerosis in the aortic root is given (since it is the most consistently reported sites and quantification is reasonably
standardized). If multiple time points were reported, the longest follow-up time is generally presented here. If aortic root
quantification was not presented, the area of most advanced atherosclerosis was generally used. Percentage increases
or decreases are intended as only rough, semi-quantitative estimates for comparison. Reference numbers are given in
parentheses. EC, endothelial cells; VSMC, smooth muscle cells; DC, dendritic cells; BMT, bone marrow transplant (after
irradiation of recipient animals); Th1, type 1 T helper cell; Th2, type 2 T helper cell; Treg, regulatory T-cell; NS,
nonsignificant; % A, percent difference in atherosclerosis compared with control animal (e.g., a 100% increase is
equivalent to a twofold increase over control; a 75% decrease is equal to of the control).
PAUL N. HOPKINS
portant downstream target of PKB while control of
mTORC2 is less well understood.
1332
1333
PAUL N. HOPKINS
Rheb is required for activation of mTORC1 (mammalian
target of rapamycin, complex 1) (FIGURE 2). Active PKB
phosphorylates and inhibits TSC1/2 leading to GTP binding of Rheb and mTORC1 activation. Active mTORC1
then acts to increase protein synthesis by promoting phosphorylation of the ribosomal protein S6 (by S6K1), activating eIF4E, and inhibiting autophagy. mTOR is opposed in
all these actions by AMPK (AMP-activated protein kinase),
which senses low cellular energy levels as reflected by increasing AMP. AMPK activates a host of pathways to generally mobilize and increase cellular fuels and energy production while decreasing energy expenditure. AMPK signaling promotes production of new mitochondria and has
also been associated with longevity. Interestingly, knockout
of at least one isoform of AMPK increased atherosclerosis
(see TABLE 1), despite its opposition to at least some aspects
of insulin signaling.
1334
1335
NOX2 is upregulated in human coronary arteries from patients with CAD, even in areas without lesions (682).
NOX2 deficiency causes X-linked granulomatous disease.
Normal human subjects all experienced impairment of
flow-mediated dilation after 20 min of arm ischemia while
patients with NOX2 deficiency (or p47phox deficiency) did
not (1079), again demonstrating relevance of NOX2 in
short-term endothelial function. Endothelial NOX5 expression was sevenfold higher in early human coronary atherosclerotic lesions and contributes to ROS production (680).
The NOX5 gene was apparently lost in rodents and hence
cannot be studied by KO in mice. Hyperlipidemia appears
to contribute to age-related increases in arterial wall ROS
production, since endothelial NOX2 expression and superoxide levels markedly increased with age in ApoE-deficient
mice but not in wild-type mice.
dependent on active Ras. The tyrosine kinase Src (or another Src family kinase) is activated by INSR (and EGFR)
through the intermediary action of SHP2. Src amplifies
EGFR activation by phosphorylating EGFR tyrosines 845
and 1101, Gab1, STAT3, and STAT5a/b. Src also phosphorylates ADAM17, further activating its sheddase activity, which thereby releases more HB-EGF (89, 1004).
Similar amplification likely occurs with INSR. Perhaps not
surprisingly, insulin signaling through INSR acts synergistically with EGFR to increase the mitogenic effects of EGFR
signaling (180). Not shown in FIGURE 2 are a number of
INSR-interacting proteins (such as Cbl: Casitas B-cell lymphoma) or SOCS (suppressor of cytokine signaling) that initiate feedback inhibition and downregulation (89). Nor are the
complex pathways shown that regulate transport of GLUT4containing vesicles to and their fusion with the cell membrane,
since GLUT4 is not expressed on endothelial cells.
PAUL N. HOPKINS
phosphorylations in two separate hits on a Thr-X-Tyr
(TXY) motif in the activation loop. Thus MAP2K are rather
exceptional dual-function kinases with both serine/threonine and tyrosine kinase activities. Similarly, MAP2K acti-
Insulin
HB-EGF
Insulin receptor
EGFR
GDP Ras
P
P
P
Sos
GTP Ras
PAK, S621
Src, Y341
PKA, S43
14-3-3
Many inputs
MAP3K
Raf1
ASK1
TAK1
MEKK2/3
MAP2K
MEK1/2
MKK4/7
MKK3/6
MEK5
MAPK
ERK1/2
JNK
p38
ERK5
Inputs
MAPK
filter
Many outputs
Ca2+
Cytoprotection
p47phox
mRNA stabilization
NOX5
p47phox
Fos
Elk1
Jun
ICAM-1, TF, PDGF
Fos
Egr1
P
ATF2
1336
RSK1
RelA
MSK1
MK2
14-3-3
Activation loop
PKC, S338
RBD CRD
P
P
P
P
Raf1, S494
GTP Ras
Raf1 N
MAPK activity
Grb2 (Shc) P
Sos
Raf1, T491
GDP Ras
(Shc) Grb2
PKA&B, S259
P
P
P
P
PKA, S233
P
P
P
P
ADAM17
In FIGURE 3 only one MAP3K is shown for the JNK and p38
modules, whereas, in reality, a number of MAP3K can activate either or both JNK or p38. These MAP3K include
MEKK1 4, MLK2/3 (mixed lineage kinases 2 and 3, also
known as MAP3K10 and MAP3K11), ASK1 (apoptosis
signal-regulating kinase 1), NIK (NF-B-inducing kinase or
MAP3K14), and TAK1 (TGF--activated kinase-1 or
MAP3K7). A number of these MAP3K as well as their activating MAP4K are involved with NF-B activation as
well, illustrating important network behavior or cross-talk
between MAPK and NF-B activation pathways. Surprisingly, KO of ASK1, either whole body or in bone-marrow
derived cells only, resulted in an increase in atherosclerosis
with greater macrophage accumulation and less macrophage necrosis (see TABLE 1).
Several MAP4K are particularly relevant to endothelial cell
activation. These include PKC (which has multiple targets), (GCK; or MAP4K2), and PAK1/2 (p21 activated kinase 1 and 2). Flow-induced PAK activation in endothelial
cells apparently activates JNK through the MAP2K MKK4
(285, 692, 1353). JNK induces bone morphogenic protein 2
(BMP2), BMP4, and Toll-like receptor 4 (TLR4) and has
numerous other proinflammatory and pro-apoptotic effects
in endothelial cells (285) yet only JNK2 appeared to be
atherogenic (see TABLE 1).
4. MAPK, MAPK targets, and atherosclerosis
Once active, one of the targets of ERK1/2 (and p38) is
MSK1 (mitogen- and stress-activated protein kinase 1).
MSK1 phosphorylates serine 276 of the NF-B p65 subunit
after translocation to the nucleus (1467). This is a key modification of p65 which facilitates CBP/p300 binding and
FIGURE 3. Mitogen-activated protein kinase (MAPK) signaling. Each MAPK module consists of a series of three protein kinases, often held in
juxtaposition by a scaffold protein (shown in amber for MAPK scaffolds as well as other scaffolds or adaptor proteins). Other regulatory proteins such
as protein phosphatases are often attached to MAPK scaffolds (not shown here). Many more signals than shown here can impinge on a MAP3K. In
addition, terminal MAPKs have many more serine/threonine phosphorylation targets than illustrated. Graph on left illustrates kinetic features of
MAPK modules which act as noise filters at low upstream input, followed by rapid, easy, and full activation above the activation threshold, and limitation
of downstream activity with higher inputs. The figure on the right suggests that MAPK modules act as funnels to collect many signals, bottle necks to
filter and process these signals, and then have many diverse targets.
1337
The unique architecture of MAPK modules, with their scaffolds, associated phosphatases, and requirements for two
closely spaced phosphorylations to activate the terminal
kinases all work together to impart uniquely useful kinetic
properties, indicated in FIGURE 3. Thus MAPK modules
remain inactive, acting as noise filters at low level inputs,
become rapidly activated once inputs rise above a specific
level, and then display a stable, limited output activity even
when inputs rise to very high levels (1140). Furthermore,
the scaffold proteins (and other regulatory proteins) can
dampen or amplify the output depending on cell status or
context (1067, 1742). All these properties are highly desirable features of a biochemical switch controlling crucial
downstream processes and help explain the preservation of
MAPK pathways in essentially all eukaryotic organisms.
Furthermore, cellular effects vary markedly depending on
cell type, duration, and location of MAPK signaling. For
example, transient cytosolic activation of ERK can lead to
cell proliferation, while sustained nuclear activity can cause
terminal differentiation (933).
for the separation fraction where it was discovered). Various 14-3-3 isoforms often hold proinflammatory client proteins like Raf1 inactive in the cytoplasm as shown in FIGURE
3 (1537, 2103, 2125). There is a suggestion that upregulation of 14-3-3 proteins may be protective in atherosclerosis,
although assignment of specific roles for the different isoforms is difficult (218, 1052). Remarkably, however, phosphorylation of S621 on Raf1 creates a binding site for 143-3 which is mandatory for Raf1 activity (by providing a
binding site for ATP and a shield against S621 dephosphorylation) (419). Once active, Raf1 phosphorylates MEK1 or
MEK2 which can then phosphorylate and activate the terminal MAPK, ERK1, or ERK2.
PAUL N. HOPKINS
acetylation of p65 Lys310, allowing full transcriptional
competency for NF-B (see below); yet another example of
cross-talk between MAPK and NF-B signaling. Also indicated in FIGURE 3 are the generally proinflammatory activities of JNK and p38. Thus JNK activates transcription
factors including early growth factor 1 (Egr1), activating
transcription factor 2 (ATF2), and activator protein-1 (AP1), which contains either c-Fos, c-Jun, or both. Egr1 and
AP-1 generally induce proinflammatory gene transcription.
ApoE-deficient mice with Egr1 knockout had a marked reduction in atherosclerosis (see TABLE 1).
1338
In MKP-1 deficiency, an unknown mechanism led to impaired ERK1/2 expression which was linked to defective
migration, an unexpected anti-inflammatory effect on macrophages (820, 1612). In ASK1 KO, there was less early
macrophage apoptosis and hence larger lesions (2005).
There was also less macrophage necrosis which might actually be an advantage in more advanced lesions (although
this possibility was not tested). If ASK1-deficient endothelial cells were less prone to inflammation, this tendency was
apparently less important for lesion size than the macrophage effects. This may be seen as a disadvantage to wholebody KO studies in general, since differences in endothelial
and macrophage effects, which may act in the opposite
direction, may not be separable.
Endothelial insulin signaling has been proposed to act as a
two-edged sword, with the potentially proinflammatory
Grb2-Sos-Ras-Raf1-MAPK cascade and production of the
vasoconstrictor endothelin-1 (ET-1) on the one hand and
the relatively protective, antiapoptotic, anti-inflammatory,
and vasodilatory PKB-eNOS pathway on the other (1251,
1432). The finding of increased atherosclerosis with IRS2
knockout in ApoE-deficient mice which was directly proportional to plasma insulin concentrations is supportive of
this view (637). Based on the increased atherosclerosis seen
in mice with endothelial-specific INSR deletion, the overall
endothelial effect of normal insulin signaling can be taken
as protective. However, as with the unexpected findings for
MAPK signaling noted in the prior paragraph, macrophage-specific deletion of INSR decreased atherosclerosis
(TABLE 1). In both cases, the direction of the effect could
potentially be explained by survival effects for the particular cell type. Moreover, the insights gained would probably
have been obscured if only whole body KO experiments had
been conducted.
C. NF-B Activation
1. General features of NF-B signaling
Some departure from the outside-in order of presentation
is taken here to first introduce NF-B itself and to illustrate
the central significance of this master proinflammatory, antiapoptotic transcription factor. Signaling pathways related
to NF-B are exceedingly complex. Indeed, in the year 2012
alone, there were 5,683 publications listed in PubMed that
included NF-B in an indexed field. Several excellent,
recent reviews are available (1639, 1841, 1944).
Like most transcription factors, NF-B is a dimer. Most
commonly, RelA (also known as p65) is paired with p50.
1339
PAUL N. HOPKINS
TNF (soluble)
ASM
Ceramide
U
U
U
SODD
TNFR1
SPHK1
O2
O2
CIC3
O2
Rac1
O2
O2
O2
O2
RFK
O2
14-3-3
PKC
U U
26S
TRADD TRAF2
RIP1 U cIAP1/2
U LUBAC
U
U U
U U
U U
TAB1 TAB2
U U
TAK1
U U
U
U
U
U
U
U NEMO
cIAP1/2
NIK
PKC
NOX1/2
BMX
TRAF3
TRAF3
FAN
U
U A20
U
U U
p62 PKC
NIK
U
U
MEKK3
U
U
U
U
26S
NEMO
Ras
IRS1
SOD1
IKK2
IKK1
NIK
ERK1/2
JNK
p38
PAK1/2
Rho
ROCK
H2O2
RSK1
IB
RelA:p50
p100
U
U
U
RelB:p52
26S
ERK5
p52
AKIP1
p53
FAK
SIRT1
NIK
CBP
PKAc
PKC
NF-B
MSK1
S536: P
S311: P
K310: Ac
S276: P
K221: Ac
Y152: NO2
Y66: NO2
1340
RHD
TAD
p65 (RelA)
U
U
GEF-H1
TRAF2
cIAP1/2
Activated NOX2 produces superoxide within the endosome which can escape through anion channels such as
chloride channel 3 (ClC3) (1154) into the cytoplasm where
it is rapidly converted to hydrogen peroxide by superoxide
dismutase 1 (SOD1). Interestingly, oxidation of extracellular cysteines on TNFR1 and TNFR2 (which are intraendosomal after endocytosis) seems to enhance binding and subsequent signaling by TNF (1363). ROS and especially peroxynitrite increase PKC activity as well (551).
FIGURE 4. Introduction to NF-B signaling. Tumor necrosis factor- (TNF-)-mediated activation of NF-B signaling is representative of the TNF
receptor superfamily. Although not shown, MEKK1 recruitment and activation may be dependent on K63 ubiquitinated TRAF2 and RIP1. Also
not shown is the E3 ubiquitin ligase, SCF-TrCP (S-phase kinase-associated protein 1, Cullin1, F-box protein complex--transducin repeatcontaining protein) which mediates K-48 ubiquitin chain attachment onto IB after IB is phosphorylated by IKK2. Sphingosine-1-phosphate
(S1P) is a required cofactor for TRAF2 to function as a K63 E3 ubiquitin ligase. See text for additional details and abbreviations.
1341
PAUL N. HOPKINS
juxtaposition with active TAK1. Additional ubiquitination
of TAK1 and NEMO seems to increase TAK1 activity and
amplify the signal (498). Phosphorylation of IKK2 by
TAK1 activates IKK2 which phosphorylates IB, marking
it for K48 ubiquitination and proteosomal degradation.
The decrease in IB liberates RelA:p50 dimers to translocate to the nucleus. At the same time, removal of IB
exposes Rel A Ser276 which becomes a target of activated
PKA subunits (designated PKAc). Translocation of RelA:
p50 to the nucleus ensues, apparently mediated by the
adapter protein A-kinase-interacting protein 1 (AKIP1)
with enhancement by hydrogen peroxide (588).
1342
High NIK activity not only activates IKK1 but also IKK2,
apparently by IKK1 phosphorylation of IKK2 (686). When
activated, IKK1 as well as IKK2 have effects well beyond
NF-B translocation with nuclear phosphorylation targets
including the RelA subunit as well as other transcription
factors and cytoplasmic signaling proteins (355, 623,
1405).
The contribution of NIK to NF-B activation is often underappreciated. Both TNFR1 and TNFR2 seem to mediate
adhesion molecule expression, yet TNFR2 appears to have
a greater role for E-selectin and ICAM-1 (275). Deficiency
of either TNFR1 or TNFR2 strongly inhibited atherosclerosis in hyperlipidemic mice (276) (see TABLE 1). Prolonged
cytokine or other signaling leading to ROS generation (as
Transactivation involves posttranslational protein modifications of the NF-B dimer, allowing it to function as a
competent transcription factor whose activity is further
modulated by other coactivators and corepressors. Phosphorylation of RelA at Ser276 is critical in facilitating RelA
binding and acetylation by the coactivator CBP (CREB
binding protein or closely related p300) and binding with
other coactivators (such as cyclin T1/CDK9) which can direct expression of a more inflammatory subset of NF-B
target genes (623, 1320). Ser276 also vies for attention from
the E3 ubiquitin ligase SOCS1 (suppressor of cytokine signaling 1) that targets RelA for degradation and the oncogenic
kinase Pim1 (proviral integration site murine leukemia virus)
that can maintain RelA active (1307). Several other posttranslational modifications are shown in FIGURE 4 (623). Various
kinases not generally considered part of the NF-B signalosome can contribute to activation of nuclear NF-B,
such as endothelial focal adhesion kinase (FAK) (1414).
Nitration by peroxynitrite (on RelA tyrosines 66 and 152),
or nitrosylation by NO of cysteine 62 of p50, inhibits
NF-B signaling by causing dissociation from DNA and
removal from the nucleus by IB (623).
NF-B complexed with CBP/p300 displaces inhibitory histone deacetylases (e.g., HDAC13) and promotes activity
of histone acetyltransferases (HATs). Histone acetylation
leads to transcriptional activation of target genes by relaxing the winding of DNA around histones, making the promoter more accessible to the RNA polymerase II complex.
Interestingly, cigarette smoke extract powerfully inactivates
HDAC enzymes to promote inflammation while GR (glucocorticoid receptor)-bound cortisol recruits HDAC2 to inflammatory gene promoters to repress inflammatory gene
expression (623). Additionally, cortisol-GR exerts powerful anti-inflammatory effects by acting at glucocorticoidresponsive elements (GREs) as a positive transcription factor to various anti-inflammatory proteins (such as annexin
A1) (1407) and by binding to other adaptor proteins, in
particular GR interacting protein 1 (GRIP1) which binds
and acts as a repressor to most NF-B and AP-1 responsive
proinflammatory genes (308, 1482). Estrogen also induces
annexin A1 with diminished adhesion to endothelium by
estrogen-treated neutrophils (1267). In cholesterol-fed rabbits and ApoE-deficient mice, glucocorticoid treatment decreased plaque size (81, 549), while no effect was seen in
ovariectomized monkeys without estrogen replacement
(11). However, medroxyprogesterone blocked the protective effective of estrogen treatment in these animals. Macrophage-specific deletion of GR did not affect lesion size in
LDLR-deficient mice but resulted in lesions with less calcification (1437).
Silent mating type information regulation 2 (Sir2) homolog
1 (SIRT1) is an anti-inflammatory deacetylase that can inactivate NF-B by removing acetyl groups. Endothelial-spe-
1343
5. NF-B transactivation
PAUL N. HOPKINS
Table 2. NF-B activation of nuclear translocation by representative members of the TNFR superfamily of receptors
Downstream Signaling
Proteins
(Transmembrane
Receptor Adaptors)
Primary intracellular
adapters
recruited to the
cytoplasmic tail of
the ligated receptor.
Additional adapters,
kinases, and E3
ubiquitin ligases may
bind to the activated
receptor.
SODD (inhibitory,
displaced by TRADD)
TRADD (recruits TRAF2,
RIP1)
TRUSS (recruits TRAF2,
IKK complex)
MADD [recruits Grb2
with Sos. Required for
ERK1/2 activation by
TNF- (963)]
FAN (recruits other
adaptors required for
neutral
sphingomyelinase
activation. FAN also
functions as a GPCR
adapter.)
Secondary adapters
may serve multiple
roles as kinases and
ubiquitin ligases
TRAF2/5 (recruit
cIAP1/2, MLK3)
RIP1 (can recruit TRAF5.
With K63 chain also
binds p62. K63 chains
required to recruit
TAB1/TAB2/TAK1.)
p62 (scaffold that also
binds PKC and
MEKK3a)
NEMO (essential final
adapter in canonical
pathwaycomplex with
IKK1 and IKK2. Binds
K63 and linear chains.
Also involved in
recruitment of
MEKK1.)
ABIN1 (binds ubiquitin
chains on RIP1 and
inhibits caspase 8
binding to FADD, thus
preventing formation of
complex II.
Proximal kinases
(and targets)
recruited and
activated by
adapters or by each
other. May also
serve as E3 ligase
targets
RIP1 (AIP1)
PKC, PKC [TRAF2 Thr
117, directing K63
ubiquitination of TRAF2
and recruitment of
IKK1 and IKK2 (1033)]
CD40 (trimer)
none known
Continued
1344
TNFR1 (trimer)
Table 2.Continued
Downstream Signaling
Proteins
(Transmembrane
Receptor Adaptors)
IL-1R (IL-1RAcP)
TRAF2/5 (TRAF2/5,
RIP1?, TAK1, ASK1,
MEKK1, K63)
cIAP1/2 (RIP1, K63)
LUBAC (NEMO, linear)
IKK activating
kinases (target)
activated by ubiquitin
scaffold in canonical
pathway
TAK1c (IKK2)
ASK1 (MAP2K)
MEKK1 (IKK2)
MEKK3a (IKK1, MKK4/7,
MKK3/6)
MLK3 (MKK4/7)
PKC (IKK2, others)
SCF-TrCP (IBs, p100,
major E3 producing K48
chains)
A20 (RIP1, TRAF, NEMO,
TAK1, K63 chains.
Induced by NF-B.)
USP21 [RIP1 (1998)]
CYLD (similar to A20.
Constitutively active.
Particularly important for
NEMO, TRAF2, TRAF6,
and Bcl-3 K63 chain
removal.)
Cezanne (removes K48
chains from IB, K63
from RIP1)
RNF11
ITCH, TAXBP1 (TAXBP1
mediates A20 binding to
RIP1 and TRAF6. ITCH
may be the E3 that K48
ubiquitinates RIP1 in an
A20 complex. ITCH also
promotes ubiquitination
of cFLIP which releases
caspase 8.)
TAK1c (IKK2)
ASK1 (MAP2K)
MEKK3a (IKK1,
MKK4/7, MKK3/6)
PKC (IKK2, others)
TAK1c (IKK2)
SCF-TrCP (IBs,
p100, major E3
producing K48
chains)
A20 (RIP1, TRAF,
NEMO, TAK1, K63
chains. Induced by
NF-B.)
USP21 [RIP1 (1998)]
CYLD (similar to A20.
Constitutively active.)
Deactivating K48 E3
ligases and
deubiquitinases or
ubiquitin editing
enzymes (targets)
TRAF2 (receptor
binding causes K63
decoration by TRAF2
of cIAP1/2 which
alters target from
NIK to TRAF3, with
degradation of TRAF3
and release of NIK)
cIAP1/2 (TRAF3, K48)
ZFP91 (stabilizes NIK,
K63)
NIK (IKK1)
General: IL-1RAcP (IL-1R activating protein) acts as a heterodimer with IL-1R. Oligomerization appears to be
required for activation of several components (such as the complex of 6 MyD88, 4 TRAF4, and 4 TRAF2 that
are recruited in that order to the activated TLR/IL-1 receptors) (1048). RIP1 appears not to be uniquely
required or is redundant for TNFR1 activation of NF-B (1976). IKK2 may only phosphorylate S32 on IB,
leading to its ubiquitination and degradation while IKK1 phosphorylates S36, followed by dissociation from
NF-B without degradation (2028). CD40 can signal through both the alternative and the canonical pathways
but alternative predominates and only this pathway is shown. E2 ubiquitin conjugating enzymes are not shown.
A role for the E3 ligase ZFP91 was recently shown for CD40 signaling (853). aMEKK3 bypasses TAK1
activation but requires p62 (which binds ubiquitin chains on TRAF6 or RIP1) to recruit PKC and MEKK3 to the
signalosome (1277). MEKK3 phosphorylation of IKK1 results in IB phosphorylation on Ser36 by IKK1 (as
opposed to Ser32 by IKK2) that leads to dissociation of IB but without ubiquitination and degradation (2028).
MEKK3 activation is primarily important in IL-1 signaling. bTRAF2 and TRAF5 each appear to largely compensate for absence of the other in NF-B signaling but knockout of both does arrest TNFR1 signaling to NF-B.
They may serve primarily as scaffolds for cIAP1/2. In this regard, TRAF2 appears more active (2075). They
also recruit RIP1 to the complex for K63 ubiquitination by cIAP1/2. This is thought to direct TNFR1 signaling
toward NF-B signaling rather than toward apoptotic signaling through FADD complex formation. TRAF2 E3
ubiquitin ligase activity may only be relevant for ASK1 activation. In TNFR1 signaling, cIAP1/2 is the main E3
ligase for RIP1, decorating it with K63 ubiquitin chains. cTAB2/3 binds only K63-linked Ub chains bringing
TAK1 in juxtaposition with NEMO. TAB1 is regulatory for TAK1 which self-activates. NEMO binds linear Ub
chains much more avidly than K63 chains. LUBAC is thought to bind K63 chains but produce linear chain for
NEMO.
1345
Activating E3
ubiquitin ligases
(targets, chain type)
also serve as E3
targets with self
ubiquitination or
from another E3
CD40 (trimer)
PAUL N. HOPKINS
cific (2079) and whole body (1680) overexpression of SIRT1
decreased atherosclerosis while decreased SIRT1 expression
increased atherosclerosis (1680). Heterozygous deficiency
caused greater endothelial NF-B activity, increased ICAM-1
and VCAM-1 expression, and greater sensitivity to TNF-
(1681). Overexpression of SIRT1 protected endothelial cells
from oxLDL induced apoptosis (2079). Importantly, 1-year
caloric restriction doubled SIRT1 in mouse aorta compared
with ad libitum feeding while 6 mo of high-fat diet decreased
SIRT1 by 50% (2079).
6. Canonical NF-B signaling through the
CBM complex
In T-cell signaling, PI3K recognizes specific scaffold phosphotyrosines produced by Syk and is activated, especially
when the costimulatory receptor CD28 is activated simultaneously, binding and activating additional PI3K. PI3K
generates membrane-bound PIP3, required for PLC activity, whereupon PLC cleaves membrane-bound PIP2 to generate free IP3 and membrane-bound DAG. IP3 opens the IP3
receptor to release calcium from the ER. Free calcium and
DAG together activate conventional and novel PKCs.
One of these PKC enzymes can then phosphorylate the
inactive cytoplasmic adapter CARD 11 (caspase-recruitment domain-containing adapter protein 11, also known
as CARMA1) causing translocation to the membrane,
formation of a trimer and recruitment of two copies of
the adapter Bcl10 which then bind mucosa-associated
lymphoid tissue I (MALT1) as a trimer. MALT1 recruits
and activates TRAF6 while also inactivating A20 (1462).
NIK may also be activated (142). Active TRAF6 deco-
1346
rates MALT1, Bcl10, NEMO, and itself with K63 ubiquitin chains, leading to TAK1 recruitment, activation of
IKK2, IB degradation, and translocation of NF-B.
This activation pathway also explains the long observed
phenomenon that phorbol esters (which are thought to
activate PKCs by mimicking DAG) can activate NF-B
(1616). CARD/CARMA, Bcl10, and MALT1 are together known as the CBM complex.
1347
PAUL N. HOPKINS
PTEN
JNK
Prdx1
Prdx1
inactive
Trx
H2O2
PTEN
inactive
ONOO
PDK1
PKC
Srx
Prdx1
Prdx1
O2
TrxR
Other chaperone
functions
Prdx1
SOK1
14-3-3
NO
JNK
Prdx1
active
inactive
Prdx1
Ras
SH
Higher oxidative stress
PKAc
Cezanne
ROS
Trx
ASK1
ASK1
GST
active
PTP
S
inactive
MEKK1 U U
Src
MAP3K
MEKK2/3
Raf1
ASK1
TAK1
SH
PTP
SH
active
Syk
MAP2K
MEK5
MEK1/2
MKK4/7
MKK3/6
MAPK
ERK5
ERK1/2
JNK
p38
IKK2
LC8
SH
LC8
S
LC8
SH
TRP14
LC8
IB
U
NF-B
U
26S
RSK1/2
PKC
GST
FIGURE 5. Selected MAPK and NF-B interactions with reactive oxygen species (ROS). Peroxiredoxin 1 (Prdx1)
is an effective peroxidase and also forms an inhibitor complex with JNK under low hydrogen peroxide/ROS levels.
However, as ROS increases, Prdx1 is rapidly and reversibly inactivated with formation of higher-order multimers,
allowing suppression of PTEN activity and activation of JNK to proceed. The other pathways are examples of the
many discrete, specific reactions whereby ROS may modulate or amplify some of the effects of MAPK and NF-B
signaling. AIP1, ASK1-interacting protein-1. See text for additional explanation and abbreviations.
1348
1349
PAUL N. HOPKINS
ing). Dimerized ASK1 then activates JNK (1398, 1424). As
depicted in FIGURE 5, ROS-mediated activation of SOK1
(Sterile 20/oxidant stress response kinase 1) may further
contribute to ASK1 activation through 14-3-3 phosphorylation (1314). Additionally, ROS dissociates SENP1 (sentrin/SUMO-specific protease 1 also known as stromal cellderived factor 1) from Trx, whereupon SENP1 moves to the
nucleus and desumoylates the serine/threonine kinase
HIPK1 (homeodomain-interacting protein kinase 1) which
translocates to the cytoplasm and further promotes activation of ASK1 (1234).
1350
In nonstressed cells, JNK and TRAF2 are held inactive, complexed with glutathione-S-transferase 1 (GST or GSTP1).
Upon exposure to ROS, GST dissociates from JNK and
TRAF2 and forms GST oligomers (1997), freeing JNK to
activate c-jun and allowing TNFR1 signaling (see FIGURES 3
AND 4). GST also functions to actively glutathionylate target
proteins when activated by ROS, nitrosative stress (1804), and
tyrosine phosphorylation by EGFR (1340). Interestingly, arsenic appears to require glutathionylation for its detoxification. In a Korean study, the combination of an inactivating
variant (Ileu105Val) of GST with high exposure to arsenic in
well water was associated with a sixfold increase in the risk of
carotid atherosclerosis (1922).
tional activation of APE1/Ref1 are stimulated by ROS, allowing harmonious nuclear reduction of target transcription factors in the face of cytosolic redox stress (139).
elinase activation (FAN). FAN recruits other adapters, receptor for activated C kinase 1 (RACK1) and embryonic ectoderm development (EED), which finally recruits neutral sphingomyelinase, an important source of ceramide on the inner
leaflet of the cell membrane (1228, 1415). Rapid membrane
production of ceramide can be prompted by numerous additional receptors and activating signals. Possibly through redox
signaling, these receptors typically activate acid sphingomyelinase by a process involving translocation of quiescent acid
sphingomyelinase in cytoplasmic vesicles to the outer wall of
the cell membrane where the active enzyme cleaves sphingomyelin (671, 1408). Assembly of dispersed signaling proteins
such as receptors and NOX2 components (p47phox, NOX2
catalytic subunits) is then facilitated by ceramide-induced fusion of small lipid rafts into a functional, large, signaling platform.
The rate-limiting enzyme for de novo ceramide synthesis is
serine-palmitoyl-CoA transferase. Palmitate (the 16-carbon
saturated fatty acid, designated C16:0) is the preferred precursor to ceramide (as opposed to the unsaturated fatty acid
oleate, for example, which is generally incorporated into triglyceride) (839, 1574). Inhibition of serine-palmitoyl-CoA
transferase by the fungal product myriocin decreased atherosclerosis substantially in hyperlipidemic mice (774, 1384) and
even promoted regression (1385). Ceramide can be inactivated by several pathways including deacylation to sphingosine by ceramidase or by further acylation by O-acyl-ceramide synthase, also known as lysophospholipase 3 (LYPLA3).
LYPLA3 deficiency promoted atherosclerosis and sensitized
macrophages to apoptosis (1758) (see TABLE 1).
Interestingly, palmitate can activate JNK by way of mixed
lineage kinase 3 (MLK3). MLK3 activation was due to redundant signaling from multiple activated PKC isoforms (PKC,
PKC, PKC, and PKC), thought to be activated by DAG and
ceramide. Palmitate feeding, as well as obesity due to high-fat
diet, have been reported to cause increased endothelial superoxide production dependent on TLR4, BMP4, and upregulation of NOX4, all promoting NF-B signaling (1131). Release
of PKC from the inhibitory adapter protein 14-3-3 seems
also to be directly promoted by ceramide, allowing PKC to
phosphorylate and inactivate the predominantly anti-inflammatory PKB and ERK5 (535).
1351
PAUL N. HOPKINS
smoke), nitro-alkene derivatives of fatty acids (NO2-oleate,
etc.), prostaglandin by-products such as 15d-PGJ2, and numerous natural products (isothiocyanate, cinnamaldehyde,
etc.). Once considered merely toxic byproducts of cell damage, these reactive electrophiles usually modulate signaling
by targeting cysteine thiolate or sulfenic acid groups, but
their effects are often distinct from H2O2 or other ROS. For
example, some aldehydes, such as malondialdehyde, primarily target lysine. Histidine residues may also be modified
in some instances.
1352
of protective antioxidant genes, illustrating the important balance between pro-oxidant and antioxidant forces
(208). Hitherto, effects of genetic manipulation of GST
enzymes have apparently not been studied with regards
to atherosclerosis (738).
1353
(leukemia associated RhoGEF), p115RhoGEF, and PDZRhoGEF. The subunits released by activated GPCR can,
in turn, contribute to activation of PLC and PI3K, and
have a number of other targets. G protein receptor kinases
(GRKs) phosphorylate the activated GPCR to provide binding sites for various adaptor proteins, such as -arrestins.
Binding of -arrestins causes arrest of G protein signaling
and direction to clathrin-coated pits for endosomal removal
of the GPCR from the cell surface. However, -arrestins can
also serve as docking proteins for entirely different downstream effectors potentially leading to a complete shift in
the initial transduction activities of the GPCR.
PAUL N. HOPKINS
tential effects of NFAT on atherosclerosis remain poorly
understood (1310). Hereafter, AT1R signaling becomes far
more complex.
3. Inflammatory signaling through AT1R is largely
mediated by PYK2
Early studies clearly identified activation of ERK1/2 as a
downstream consequence of certain ligated GPCR, but the
mechanism remained obscure. The effect was seen primarily in Gi and Gq-coupled GPCR (including AT1R) and
required activation of PKC (usually atypical isoforms such
as and ) and/or increased intracellular calcium and ROS,
all potentially attributable to the PLC activated by these
receptors. Merely increased calcium and ROS, seen with
activation of many receptors and ion channels, can also
activate ERK1/2.
1354
1355
PAUL N. HOPKINS
1356
peroxide. NO production is reduced and bioavailability impaired. These endothelial cells do not align with the flow.
They have disorganized cytoskeletons and develop other
features characteristic of lesion-prone areas including increased apoptosis, frequent mitoses, greater permeability
(particularly at sites of mitosis), and a shorter glycocalyx. In
addition, endothelial cells exposed to disturbed flow increase production of subendothelial matrix components
such as fibronectin, resulting in enhanced inflammatory responses and thickening of the basement membrane (282,
693).
A
50
40
Atherosclerosis-prone waveform
30
20
10
0
-10
500
1000
2000
1500
2500
3000
3500
4000
atherosclerosis
Susceptibility to
Shortened glycocalyx
permeability
Monocyte adhesion
Fibronectin synthesis
10
100
0.1
0.01
apoptosis
mitosis
BMP-4
NAD(P)H Ox
superoxide, ROS
NF-B, ERK1/2
Erk, JNK,
Rho GTP
tyrosine kinases
ICAM-, VCAM-1
MCP-1 release
TLR-2
Inflammatory Response
Time (ms)
1000
Hours
50
45
40
35
30
25
20
15
10
5
0
Athero-protective waveform
0
500
1000
2000
1500
2500
3000
3500
4000
0.01
0.1
10
100
1000
Athero-protection
Cyclins decrease
anti-oxidant genes
NF-B continues
Nrf2, Nrf1
Organization of actin
cytoskeleton,
alignment with flow
NO release
Time (ms)
Inflammatory Response
10000
Time (hours)
FIGURE 6. Overview of hemodynamic effects on signaling related to atherogenesis. A: characteristics of a
highly atherogenic waveform (slow flow with directional change during each cardiac cycle) together with
approximate order of resulting signals. B: atheroprotective waveform (rapid flow, high shear stress with
pulsatory but unidirectional flow) and associated signaling. There is an initially pro-inflammatory response,
similar to early signaling in A, followed by prolonged active suppression of inflammation.
1357
PAUL N. HOPKINS
Cilium
Onset of flow or directional
change in shear stress
vWF
ANGPT2
P-selectin
B4 GPCR
P2X4
TRPV4 &
TRPC1
PKD2
CD44
Glypican-1
Caveolin-1
Arg
Kir2.1
PKD1
Ras Ral PLD1
RasGRP RalGDS
Caveola
ATP
PI3K
Vm
PKG
q
Calmodulin
NO
ONOO
O2
CaMKII
DAG
PLC
WPb
Calmodulin
eNOS
PKG
sGC
PKC
PYK2
IP3R
MnSOD
H2O2
cGMP
RyR
Ca2+
IP3
Mitochondria
PKG
PC
DAG
cPLA2
ER
PDK1
COX1
COX2
AA
LPC
PGIS
PGI2
CYP2C/J
MNK1
EETs
p47phox,
p22phox,
eNOS,
Integrins,
VE-cadherin
PKC
Raf1
PYK2
inhibition,
acivation of
MAPK & IKK
PAF
acetylase
PLD
RKIP
LPC
Extracellular matrix
LPA
PAF
ATX
Vasodilation
Inflammation
Thrombosis
FIGURE 7. Early mechanotransduction of shear stress in endothelial cells, characterized by type I inflammatory
signaling through G protein-coupled receptors (GPCR) and ion channels. The downstream signaling from the B2
bradykinin GPCR is typical for many GPCR, but its activation by shear stress without substrate binding is unique to a few
GPCR. Shown in yellow boxes are other mechanical shear-stress transducers. COX-1 is a constitutive but low-capacity
enzyme. COX-2 is induced by proinflammatory signals generally and by prolonged laminar flow specifically in endothelial
cells and has greater overall capacity than COX-1. Endothelial H2O2 has vasodilatory effects on coronary arteries but may
have different effects in different arterial beds. AA, arachidonic acid; LPA, lysophosphatidic acid; LPC, lysophosphatidylcholine; PLD, phospholipase D; ATX, autotaxin; PC, phosphatidylcholine; sGC, soluble guanylate cyclase; Vm, membrane
potential (negative inside, thus decrease in Vm means hyperpolarization); vWF, von Willebrand factor; WPb, WeibelPalade body. Expression of purinergic ATP receptor P2X4 is much greater in veins than arteries; its conductivity for
calcium is markedly increased by flow but also requires ATP. Without the effect of angiopoietin 2 (ANGPT2), released
from WPb to block the highly anti-inflammatory signaling of ANGPT1 through its receptor Tie2 (not shown), endothelial
cells would remain largely unresponsive to subsequent activating signals.
1358
O2
PIP2
1359
PAUL N. HOPKINS
Table 3. Hemodynamic effects on endothelial cells and other early steps in initiation (55 genes tested)
Gene
Model
% A
Function, Comment
153%
PFN1
PFN1/ LDLR/
261%
PECAM-1
PECAM-1/ apoE/
241%
PECAM-1/ LDLR/
284%
VEGFR2
266%
Shc1
(p66Shc)
p66Shc/ apoE/
260%
PLC2/3
PLC2/3/ apoE/
251%
ADAM15
ADAM15/ apoE/
261%
FN
FN EIIIA/ apoE/
253%
nmMLCK
nmMLCK/ apoE/
231%
BMP2
VSCM-specific BMP2 Tg
in apoE/
NS
BMP4
Fc-ALK3 intra-peritoneal
adminstration into
LDLR/ mice
243%
MGP
MGP human Tg in
apoE/
237%
MGP/ apoE/
296%
BMPER/ apoE/
131%
BMPER
Continued
1360
Sdc-1
Table 3.Continued
Gene
Model
% A
Function, Comment
Tie1
ERK5
240%
270%
Tie1 (tyrosine kinase with immunoglobulin and EGF factor homology domains 2) is an
angiopoietin (ANGPT) receptor that is upregulated in endothelium exposed to
disturbed flow. Reduction in plaque shown is progression in aortic regions
exposed to disturbed flow. Little change seen at aortic root (1978). Tie1
activation by shear stress appeared to block the anti-inflammatory signaling of
Tie2.
In the same study, either elevated glucose plus low-dose hydrogen peroxide or
higher hydrogen peroxide activated RSK1/2 binding to ERK5 with inactivating
phosphorylation of ERK5 S496 by RSK1/2 (986).
KLF2
KLF2/ apoE/
131%
Arg2
endothelium-specific Tg
human Arg2 in
apoE/
Arg2/ apoE/
179%
eNOS
(NOS3)
eNOS/ apoE/
180%
nNOS
(NOS1)
nNOS/ apoE/
131%
GTPCH
GTPCH Tg apoE/
228%
Cav1
Cav1/ apoE/
233%
NPR1
NPR1/ apoE/
164%
267%
Continued
1361
SENP2
endothelial-specific
1130%
ERK5 deletion
(tamoxifen-responsive
Cre-Lox) in LDLR/
SENP2/ LDLR/
197%
PAUL N. HOPKINS
Table 3.Continued
Gene
Model
% A
Function, Comment
apoE/
COX-1
COX-2
ER (ESR1)
ER/ LDLR/
CYP7B1
CYP7B1/ apoE/
172%
mPGES-1
mPGES-1/ LDLR/
234%
IP
IP/ apoE/
132%
TP
TP/ apoE/
274%
DP1
DP1/ apoE/
DP1/ LDLR/
FP
FP/ LDLR/
238%
fat-1
fat-1 Tg in apoE/
238%
UCP1
Endothelial protection
Aortic VSMC-specific
1350%
overexpression of
UCP1 in apoE/
UCP2
UCP2/ C57BL/6J
mice fed Paigen diet
LIAS
LIAS/ apoE/
274%
145%
1280%
140%
144%
1321%
143%
Reduction in lesions was even greater in some measures (1167). COX-1 deletion
virtually eliminated plasma TXA2 (more so than complete platelet COX-1 inhibition
with aspirin), suggesting alternate sources of synthesis. Similarity to protection
from TP KO (below) is striking.
Inducible whole body COX-2 KO did not result in blood pressure or lipid changes.
There was no excess atherosclerosis in mice with combined COX-2 and FLAP KO
(2050). Macrophage-only KO by Cre-Lox in LDLR-null mice decreased
atherosclerosis 25% at 6 mo but BMT of COX-2 KO marrow showed no effect in
apoE KO mice (1284).
Shown is the increase in lesion area in ovariectomized ER KO given the larger
estrogen supplement (from subcutaneous pellets). BMT and Cre-Lox endothelialspecific KO studies showed the protective effect of the ER was due endothelial
effects (146). AMPK activation may be involved (2084). Estradiol in
ovariectomized LDLR KO mice increased COX-2 and benefit was dependent on
PGI2 signaling through IP receptor (462).
CYP7B1 converts 27-hydroxycholesterol (27HC) to 7,27-dihydroxycholesterol in the
pathway to chenodeoxycholate synthesis. CYP7B1 deficiency results in accumulation
of 27HC which inhibits both transcriptional and nonnuclear effects of ER and ER
(1833). Increased atherosclerosis also with 27HC administration (1832).
mPGES-1 (microsomal prostaglandin E synthase 1) is an inducible enzyme. It was
primarily expressed in lesional macrophages and KO decreased PGE2 synthesis
mainly in macrophages with subsequent depletion of lesional macrophages. KO
also resulted in increased PGI2 synthesis by VSMC (1906). Data is placed here
rather than table 6 because of the increased VSMC PGI2 and to group this data
with other prostaglandin-related genetic interventions
PGI2 receptor (IP) is present on multiple cell types besides endothelial cells (925).
Similar average increase seen in a study with LDLR KO mice (462).
TXA2 prostanoid receptor (TP) placed in this table because of marked proinflammatory endothelial signaling (925).
The PGD2 receptor, DP1, is the target of the failed drug laropiprant, a DP1
antagonist. Later time points in apoE KO mice were nonsignificant (1688).
Similarly, effect in LDLR KO mice was in females at 24 wk with lesser effects at
later time and males (1654). DP1 KO in apoE deficient mice also increased
aneurysm formation in the setting of prolonged ANG II infusion. Evidence was
presented that the vasodilating DP1 receptor also had anti-platelet effects in
humans (1654)
The F prostanoid receptor (FP), also known as PTGFR (prostaglandin F receptor)
binds PGF2. Deletion of the receptor primarily affected renal sodium/water
handling and thereby lowered blood pressure which appeared to explain the effect
on atherosclerosis (2049).
fat-1 is a round worm gene that converts n-6 to n-3 fatty acids (1893). Markedly
decreased inflammation was seen in lesions.
with prolonged laminar flowrelated to antioxidant defense
Expression of UCP1 (uncoupling protein 1) in aortic smooth muscle unexpectedly led
to increased ROS production (132). There was also a substantial increase in
blood pressure and aldosterone which may have contributed to the increase in
atherosclerosis.
UCP2 (uncoupling protein 2) is upregulated by oxidative stress or inflammation and
dampens stimulated mitochondrial ROS production by an unknown mechanism.
UCP2 KO mice fed a Paigen diet had increased VCAM-1 as well as other signs of
endothelial activation (1240). UCP2/ BMT into LDLR/ resulted in modestly
increased lesions with more macrophage apoptosis and signs of ROS-induced
protein modification (157). UCP2 has since been shown to be crucial for
macrophage efferocytosis as well (1380).
Lipoic acid synthase (LIAS) produces lipoic acid in the mitochondria, a potent
antioxidant which helps regenerate reduced glutathione. Homozygous lipoic acid
synthase (LIAS) deficiency is embryo lethal. Only male LIAS mice had an increase
in lesion area (value given, females 5% increased, NS) and displayed increased
TBARS and decreased erythrocyte GSH/GSSG which correlated strongly and
inversely with lesion area in males (2037).
Continued
1362
COX-1
Table 3.Continued
Gene
Model
Function, Comment
247%
Cu-Zn SOD
human SOD1 Tg in
apoE/ with added
benzo(a)pyrene
MnSOD
SOD2/ apoE/
1167%
EC-SOD
SOD3/ apoE/
238%
HMOX1
HMOX1/ apoE/
Prdx1
Prdx1/ apoE/
170%
Prdx2
Prdx2/ apoE/
125%
Prdx6
Prdx6/ apoE/
NS
GPx1
GPx1/ apoE/
diabetic mice
153%
GPx4
human GPx4 Tg in
apoE/
human catalase Tg in
apoE/
241%
GCLC
GCLC Tg in apoE/
233%
GCLM
GCLM/ apoE/
145%
Trx2
Trx2 Tg in apoE/
265%
TTPA
TTPA/ apoE/
142%
GULO
GULO/ apoE/
SVCT2/
1230%
Catalase
1269%
233%
In the apo E KO mouse Prdx6 KO did not affect atherosclerosis but atherosclerosis
was significantly increased in 1 of the 3 black-6 mouse strains tested (1916).
Aortic sinus at 20 wk in diabetic mice with GPx1 (glutathione peroxidase 1) KO
(1016). Difference in nondiabetic mice was NS. Increased signs of oxidative
damage seen.
Over-expression of GPx4 was also shown to decrease endothelial production of
adhesion molecules (677).
Over-expression of catalase reduced atherosclerosis. A twofold increase in catalase
activity was seen in the aorta (2021). Human SOD1 (superoxide dismutase 1,
also known as Cu/Zn SOD) overexpression alone or with catalase overexpression
showed small, nonsignficant reductions in atherosclerosis in this model. Aortic
sinus lesion area was strongly and directly proportional to aortic F2-isoprostanes.
GCL (glutamate-cysteine ligase) is the rate limiting enzyme in glutathione synthesis.
The enzyme consists of a catalytic subunit, GCLC, and a modifier subunit, GCLM,
encoded on separate genes. GCLM required for full activity. Whole body KO of
GCLC is embryo lethal. Effect of macrophage-specific overexpression is given here
(244).
BMT studies showed more than half the effect of GCLM deficiency could be
accounted for by hematopoeitic cells, presumably macrophages. Increased
apoptosis (caspase 9 activity) of macrophages seen when exposed to acrolein
(244).
Trx2 (thioredoxin 2) is the mitochondrial form in mice. VEGF promoter used for
endothelial-specific over-expression (2070).
TTPA (tocopherol transfer protein, alpha, also -TTP) is a vitamin E transfer protein
involved in loading of VLDL with dietary vitamin E (delivered to the liver by
chylomicrons and chylomicron remnants). Plasma and aortic tissue levels of
vitamin E were reduced in TTPA KO mice and lesions correlated with F2ispoprostanes (1772).
GULO (gulonolactone oxidase) is found only as an inactive pseudogene in humans,
primates, and guinea pigs. Increased atherosclerosis with dietary deficiency of
both vitamin E and vitamin C in GULO KO, apo E-null mice was also shown in this
study together with increased macrophage content. SVCT2 (sodium-vitamin C
transporter 2) is a sodium-dependent transporter mediating vitamin C transport
into many tissues including vascular cells. Prior studies in GULO KO, apo E-null
mice were negative regardless of vitamin C intake, suggesting more severe tissue
deficiency of vitamin C or combined vitamin C/vitamin E deficiency is required to
promote atherosclerosis (93)
Continued
1363
% A
PAUL N. HOPKINS
Table 3.Continued
Gene
SVCT2
Model
% A
Function, Comment
242%
CTH
CBS
with zincinducible human CBS
Tg apoE/
apoE/ mice treated
with CTH inhibitor
propargylglycine
163%
1124%
A mechanism for exocytosis has been proposed which provides a useful illustration of G protein signaling (1506). The
Ca2-CaM complex binds to Ral-GDP dissociation stimulator (RalGDS, a GEF), inducing a conformation change
that relieves autoinhibition and frees RalGDS from its cytoplasmic sequestration by -arrestin. RalGDS moves to
the membrane and binds active (GTP-bound) Ras, allowing
RalGDS to exchange GDP for GTP on membrane-bound
RalA (one of the Ras family of GTPases). Active RalA then
mediates exocytosis of the WPB by coordinating the exocyst
complex through enhancing activity of phospholipase D1
(PLD1). PLD1 cleaves choline from phosphatidylcholine to
yield phosphatidic acid which promotes fusion of the vesicle
and plasma membrane (1506).
3. Cytosolic phospholipase A2, platelet activating
factor, lysophosphatidylcholine, and
lysophosphatidic acid
Cytosolic PLA2 (cPLA2 or group IVA phospholipase A2) is
also depicted in FIGURE 7. Activation of cPLA2 depends on
calcium-induced translocation to cellular membranes (especially the nuclear envelope and Golgi) and interaction with
membrane PIP2 and/or ceramide-1-phosphate. Various kinases, such as PKC, CaMKII, or MAPK interacting kinase
1 (MNK1), can phosphorylate cPLA2 and thereby increase
1364
Because of drug-related controversies, a great deal of attention has focused on COX-1 and COX-2 even though their
substrate and product are the same. Meta-analyses do show
a dose-dependent increase in risk of coronary events among
patients taking COX-2-selective inhibitors, presumably because the greater inhibition of the endothelial-predominant
COX-2 decreases protective PGI2 while leaving platelet
TXA2 synthesis relatively unaffected (618, 1651, 1806).
Recent advances in inducible knockouts show COX-2 deficiency usually does increase atherosclerosis while COX-1
deficiency resulted in a substantial reduction in lesions (see
TABLE 3). In the meantime, relatively little attention has
been paid to cell-specific signaling in relation to the individual prostaglandin receptors.
Effects of TXA2, or more specifically, TP signaling are not
simply restricted to platelets. Various endothelial stressors
such as age, diabetes, and hypertension can lead to increased ROS, elevated intracellular calcium, and decreased
NO production, all of which impair endothelial IP and increase TP expression and signaling. Furthermore, both of
the endoperoxides (PGG2 and PGH2) and even PGI2 (particularly when produced at high but physiologic levels) can
act as agonists for TP, causing endothelial-dependent arterial contractions and possibly promoting inflammation
(1866). Although PGIS predominates, endothelial cells
can express all the prostaglandin synthases. Their levels
of expression can vary by animal variety, with age, intracellular calcium, ROS, and NO exposure. Exposure of
endothelial cells to high glucose caused increased peroxynitrate formation, nitration and inhibition of PGIS,
and diversion of PGH2 to inflammatory TP signaling
with adhesion molecule expression (2123). Importantly,
endothelial TXAS expression and TXA2 synthesis increase with age and inflammation generally (albeit accompanied by increased PGI2 synthesis) regardless of
whether the precursors come from COX-1 or COX-2
(1866). In human embryonic kidney (HEK) cells and aortic VSMC, ROS production caused by TP ligation promotes intracellular TP maturation and a doubling of cell
surface TP expression in a positive-feedback pathway
(1966). In human vascular endothelial cells, TP ligation
also causes changes in cell-surface fractalkine (CX3CL1)
expression and release that promotes leukocyte adhesion
(1798). Such changes also have been associated with progressing atherosclerosis with increased TXAS and TP
seen in more advance lesions of LDLR KO mice (367).
1365
lets is coupled primarily to G12/13 (leading to RhoA activation which mediates shape change) and Gq (causing
PKC activation and subsequent aggregation). IP, DP1, EP2,
and EP4 are relaxant receptors and activate AC (adenylate
cyclase) through Gs coupling (1286). IP in platelets also
causes cAMP generation through Gs coupling which inhibits platelet activation (614).
PAUL N. HOPKINS
Genetic mouse models have clarified a number of additional
issues regarding prostaglandin synthesis, signaling, and atherosclerosis, although questions remain (662). Details of
prostaglandin-related studies are summarized in TABLE 3.
Whole body COX-1 KO did decrease atherosclerosis, but
COX-1 deficiency in hematopoetic cells only actually increased atherosclerosis, probably because of a compensatory increase in macrophage COX-2 production. Consistent with this notion, transplant of bone marrow or fetal
liver cells deficient for COX-2 mostly decreased atherosclerosis.
1366
Arachidonic acid is metabolized by CYP2C and CYP2J subfamily enzymes to 5,6-EET or 11,12-EET and 8,9-EET or
14,15-EET, respectively. These EETs increase cAMP (like
PGI2), but have additional strong anti-inflammatory effects,
including NF-B inhibition. They promote endothelial proliferation as well. The relevant intracellular signaling pathways are poorly understood. Some of their anti-inflammatory effects appear to be due to competitive inhibition of the
TP receptor (121). Soluble epoxide hydrolase (sEH), encoded by Ephx2, converts EETs to much less active dihydro derivatives (410, 411). KO of sEH decreased injuryinduced neointima formation in the femoral artery but not
in the carotid artery of ApoE-deficient mice (1476). Inhibitors of sEH, some of which are undergoing clinical trials,
appear to inhibit atherosclerosis in ApoE KO mice, particularly in the setting of infused ANG II (which downregulates CYP2C and CYP2J and increases sEH expression)
(411, 1923).
Importantly, n-3 fatty acids substituting for arachidonate in
most prostanoid pathways yield less active chemotactic,
proinflammatory, or vasoconstrictive products (391). The
n-3 fatty acids also appear to play a particularly important
role in synthesis of recently recognized inflammation-resolving lipid mediators named lipoxins, resolvins, and maresins. These novel prostanoid pathways involve lipoxygenases, cytochrome P-450 enzymes, and acetylated COX-2.
Typically, one cell type synthesizes a precursor which is
acted on by a different cell type to yield the final product
(1593, 1666). Lipoxin A4 is derived from arachidonate,
resolvin E1 from the n-3 eicosapentaenoic acid (EPA), resolvins D1 and D2 from docosahexaenoic acid (DHA), and
maresins from DHA. These resolving lipids will be discussed further in section VIIC5.
Most animals, including all mammals, lack the ability to
convert n-9 or n-6 to n-3 fatty acids. Transgenic expression
in mice of the roundworm fat-1 gene from Caenorhabditis
elegans, capable of conversion of n-6 to n-3 fatty acids,
increased n-3 levels five- to eightfold in ApoE-deficient mice
and decreased atherosclerosis by nearly 50% (1893).
1367
PAUL N. HOPKINS
Xanthine
Urate
XDH
Onset of flow
XO
Heparan
sulfate
fibers
Hyaluronan
O2
HSP60
Tight junction:
Occludins
Claudins
JAMs
Syndecan-1
Actin
cortical web
ZO1
Linkers
Linkers
SMADs
O2
ZO1
BMP4
Myosin
Actin
stress
fibers
Src
PKC
MLCK
Dense
peripheral
actin bands
JNK, p38,
MKK,
ASK1
PPI
MYPT1
ROS
Cx43
VE-cadherin
NF-B
ERK1/2
MEK1/2
Raf1
Adherens
junction
MAP4K4 Nck1
Rac1
PAK Rac1
Cdc42 PIX Git
RhoGDI
Dbl
Ack1
FAK
Rho
PIP2
Src
Rho
FAK
Talin
Shc
Paxillin
PI3K
PLC
Src
21
Extracellular matrix
Syndecan-4
Collagen
PI3K
PECAM-1
p130CAS
FAK
Paxillin
PKC
Ras
Crk Grb2 Sos
Fibronectin,
fibrinogen, osteopontin
Grb2
Shc
Talin
NF-B
Talin
PKA
Raf1
VEGFR2
Dock180
ROCK2
Src
Shc
Src
Integrins
41, 51, V3
large arrays called focal adhesions which can also contain syndecans (particularly syndecan 4). Integrins are
unique in their ability to make switchable (on/off) links
with extracellular proteins (that is, the binding affinity of
1368
NIK
1369
PAUL N. HOPKINS
flow-induced JNK activation, but not ERK activation, demonstrating at least two separate MAPK activation pathways
emanating from PECAM-1, consistent with the model in
FIGURES 8 AND 9.
Activation of tyrosine kinases through PECAM-1 mediates
the activation of ERK with onset of flow. The mechanism is
instructive and is relevant for subsequent flow-mediated
signaling. Tyrosine phosphorylation of the integrin cytoplasmic tail is also presumably required for talin binding
and regulates competitive binding of inhibitors (66, 67), but
the connection between PECAM-1 activation and integrin
tyrosine phosphorylation is unclear.
1370
Endothelial cell
Endosomal removal
of VE-Cadherin
Arf6
ARNO
Src activation
SH4
ulin
SH3
Vin
c
Raf1
MEK2
ERK1/2
Csk
SHP2
SH4
Catenins
P
VE-cadherin
P
Shc
Src
Inhibitory filamin
binding
SH3
SH4
Kinase
SHP2
VEGFR2
Activated integrins
talin, kindlin bound
Shc
PLC
PI3K
SHP2
Pro
Targets
Csk
Sos Grb2
Ras
Kinase
Src
Yes
SH3
ADAM15
PECAM-1
Fyn
Targets
PKC
Actin
B2
cul
Vin
Filamin
Talin
Shc
Kindlin
Extracellular matrix
Filamin
in
Targets
Talin
Kindlin
PIP2/3
PI3K
Integrins
V3 or 51
FIGURE 9. Detail of endothelial integrin inside-out activation (right) by signaling through VE-cadherin,
VEGFR2, and PECAM-1. Steps (13) in mechanotransduction are indicated by black circles with white numbers. Fyn (Fgr/Yes related novel protein) is a Src-family tyrosine kinase. For a more detailed structural model
of integrin-linker protein-actin interactions, see Legate KR and Fassler R. J Cell Sci 122: 187, 2009. Also
shown is a model for activation of Src (left). Note that Src is constitutively bound to membranes.
cell membrane with disturbed flow while occludin expression actually decreases (1796).
1371
PAUL N. HOPKINS
the focal adhesion to actin microfibers, intermediate filaments, and microtubules.
An important theme illustrated by integrin signaling is the
action of tyrosine kinases creating switchable scaffolds
(1389). Scaffolds or adapter proteins recruited into these
pathways are reversibly tyrosine phosphorylated, allowing
subsequent signaling molecules to attach and generate
downstream signals. Not shown in FIGURE 8 are the various
inhibitors and scaffold phosphatases which turn off focal
adhesion signaling.
The pathway for PAK activation may be outlined as follows. High-affinity binding of activated 51 or V3
integrins to fibronectin or vitronectin promotes binding
of paxillin to the integrin cytoplasmic tail and FAK to
talin and paxillin. FAK then self-phosphorylates, provid-
1372
ing docking sites for proteins with SH2 and PTB domains
including Src, PI3K, PLC, Grb2, and p130CAS. Src and
FAK further phosphorylate each other. Shear stress subtly stretches p130CAS and progressively exposes additional phosphorylation sites for Src (1544). Extensive
p130CAS tyrosine phosphorylation is measurable within
300 ms (1265).
1373
In FIGURE 8, activation of PKC by ROS is shown. Specifically, disturbed flow was found to increase peroxynitrite,
which then promotes activation of PKC (by several possible direct or indirect means). Activated PKC can, in turn,
bind to and activate the SUMO ligase PIAS4 (protein inhibitor of activated STAT4), thereby promoting sumoylation
of nuclear p53, with translocation of sumoylated p53 to the
cytoplasm where it binds Bcl-2 and Bcl-xL and promotes
apoptosis (750, 1738). The sumoylation state of p53 may
thus determine whether the net effects of p53 accumulation
are cytoprotective or apoptotic. PIAS4 also promotes sumoylation of NEMO with subsequent greater activation of
NF-B (1103). Disturbed flow also promotes sumoylation
and inhibition of ERK5, impairing its protective effects
(749). The SUMO deconjugating enzyme SENP2 acts on
p53 and ERK5 and has protective effect on atherosclerosis
(749).
PAUL N. HOPKINS
BMP4 expression and release is greatly upregulated by oscillatory, disturbed flow, whereas BMP2 is not. As such,
BMP4 may be an important means of maintaining oxidative
stress and inflammatory activation of endothelium at atherosclerosis-prone sites. In contrast, BMP2 is produced in
response to inflammation (such as in response to TNF-) or
high pressure (278). Both BMP2 and BMP4 were induced by
JNK1 in mice at atheroprone sites (285). Also, incubation of
human aortic endothelial cells in high glucose increased expression of both BMP4 and BMP2; their type I receptors
ALK1, ALK2, ALK3, and ALK6; the type II BMP receptor
BMPRII; and at least two extracellular inhibitors of BMP
receptor binding, Noggin and MGP. Similar effects were
seen in aortic endothelium of diabetic mice or rats accompanied by vascular calcification (185). BMP2 was also upregulated in endothelial cells incubated with oxLDL (413).
Interestingly, prolonged laminar shear leads to SMAD6 and
-7 phosphorylation which leads to greater SMAD6/7 inhibition of signaling by SMAD1, -5, and -8, thereby blocking
the effects of BMP4 and BMP2 (855). In contrast, disturbed
flow upregulates endothelial production of BMP ligand
traps listed above (though not proportionately as much as
BMP4), possibly as means to partly mitigate the effects of
increased BMP4 production (278).
1374
Recent manipulations of BMP signaling suggest a potentially promising means of mitigating atherosclerosis. MGP
overexpression modestly decreased atherosclerosis (2029).
Yet, surprisingly, MGP deficiency almost entirely blocked
endothelial production of ICAM-1, VCAM-1, and E-selectin and almost entirely abrogated atherosclerosis (2029).
These results suggest MGP may be acting in some role beyond a mere BMP ligand trap. In contrast, BMPER hemizygous deficiency clearly increased atherosclerosis (1416).
Specific inhibition of BMP type I receptors (such as ALK2)
using a small molecule (LDN-193189) reduced atherosclerosis 43% in LDLR-deficient mice with a similar reduction
by use of recombinant ALK3-Fc which bound and inhibited
BMP2 and BMP4 (413). Both interventions were well tolerated by these mice, and clinical trials may be warranted
after further testing. Interestingly, VSMC-specific transgenic overproduction of BMP2 in ApoE-deficient mice did
not increase atherosclerosis but greatly increased intimal
calcification of arteries, demonstrating separability between
atherogenic and calcific effects of BMPs (1276).
7. ER stress in endothelial cells
ER stress, also called the unfolded protein response, occurs in
cells with increased or excessive protein synthesis demands
and is aggravated by increased ROS. In general, ER stress is
triggered by a relative deficiency of ER chaperones which direct normal folding of newly synthesized proteins within the
ER. In times of lower demand, plentiful chaperones occupy
binding sites of several ER membrane-bound receptors. These
receptors transmit stress signals when not occupied as in times
of increased utilization of chaperones. These signals can help
compensate for the demands of increased protein synthesis or,
if extreme, they can promote apoptosis. ER stress is discussed
in more detail in section XC3.
Endothelial ER stress was found in areas of disturbed flow
in both swine (329) and mice (506). BiP (immunoglobulin
heavy chain-binding protein, also known as GRP78), the
best studied ER chaperone and mediator of ER stress, could
be induced by exposing cultured endothelial cells to disturbed flow with upregulation dependent on MAPK p38
and integrin 21 signaling (506). BiP upregulation is generally a protective, compensatory response to ER stress.
X-box binding protein 1 (XBP1), another key regulator of
the ER stress response, was also found to be upregulated in
atherosclerosis-prone branch points in mice. Prolonged
XBP1 signaling promoted apoptosis which was counterbalanced by overexpression of VE-cadherin (2060).
AMP-activated protein kinase (AMPK) is the master energy
sensor and regulator of the cell. It is activated by an increase
in the ratio of AMP/ATP as well as by hypoxia, ROS, peroxynitrite, hyperosmolarity, and certain drugs including
metformin and thiazolidinediones. Activation of AMPK
has numerous effects with multiple protein targets, but
among these is suppresion of ER stress, possibly by decreas-
ies showed more extensive effects in regulation of development, cell proliferation, differentiation, and apoptosis in
various cell types. BMPs are involved in promotion of angiogenesis and can promote osteoblast-like differentiation
in dedifferentiated VSMC and other cells. BMP2 and BMP4
have been most extensively studied for their vascular effects.
Extracellular BMP2/4 binds dimers of type II receptors, such
as BMPRII, and instigates juxtaposition of type I receptor
dimers such as activin receptor-like 1 (ALK2) followed by
serine/threonine phosphorylation by the constitutively active
type II receptor of the cytoplasmic tails of ALK2. Activated
ALK2 then serine phosphorylates SMAD1, -5, and -8, which
form active transcription factors after dimerizing with
SMAD4. BMP signaling is also characterized by the existence
of a series of ligand traps or extracellular endogenous inhibitors which sequester BMPs and block interaction with the
cognate receptors. These inhibitors have varying affinity for
the different BMP proteins and include Noggin, follistatin,
chordin, BMP endothelial cell precursor-derived regulator
(BMPER, called cross-veinless 2 in Drosophila), Cerberus,
gremlin, twisted gastrulation homolog 1 (TWSG1), and matrix Gla protein (MGP). Further controls are imposed by intracellular E3 ubiquitin ligases, SMAD ubiquitination regulatory factors (SMURFs), and phosphorylation by MAPKs.
BMP signaling in endothelial cells clearly induces NOX1 (not
NOX2) and promotes proinflammatory changes including expression of adhesion molecules, primarily attributable to the
induction of ROS production (310, 855). BMP4 signaling can
also induce HMOX1, thus providing some antioxidant balance (278).
100000
Integrin 3 mRNA
Integrin V mRNA
2048
350
p65 mRNA
10000
1000
Percent of baseline
Percent of baseline
Percent of baseline
1024
300
250
200
150
100
50
P-FAK
P-p38
P-MSK1
P-p65
10
Hours of Flow
15
20
256
128
64
32
16
8
4
2
1
100
512
Hours of Flow
10
12
14
16
18
Hours of Flow
FIGURE 10. Measured changes in endothelial signaling molecule activation and cytokine production with
exposure to flow. A: marked changes as a result of slow flow in mRNA of integrins V and 3 (one of the integrin
partners that recognizes fibronectin) and mRNA for the p65 (RelA) subunit of NF-B in human vascular
endothelial cells (HUVEC). Increase in activated phospho-3 was also seen. Treatment with anti-3 antibody
partially blocked subsequent increase in cytokine production (not shown). B: changes in phosphorylated
(activated) signaling molecules seen after onset of slow flow in HUVEC. The initial change was in FAK (P-FAK
signifies active, phosphorylated FAK), followed by p38, then MSK1, and finally the p65 (RelA) subunit of NFB.
C: changes in mRNA of chemokines in HUVEC as a result of slow or high flow. [Data from Shaik et al. (1600).]
1375
The net effect of slow flow on endothelial signaling and activation was elegantly demonstrated by Shaik et al. (1600) (see
FIGURE 10). Slow flow was seen to rapidly activate first FAK,
then the terminal MAPK p38, then the downstream effector
MSK1, and finally phosphorylation of the p65 unit of NF-B
(FIGURE 10A). Levels of these activated, phosphorylated signaling molecules diminished over time, suggesting negative
feedback. At the same time, mRNA encoding the 3 integrin,
and especially the p65 unit of NF-B, were greatly increased.
The 245-fold increase in p65 mRNA was reflected only partially in the much lesser increases of active p65 or nuclear
NF-B. Nevertheless, the large pool of p65 mRNA illustrates
the heightened sensitivity of the pathway, poised for markedly
enhanced activation in areas of slow flow. A marked increase
in cytokine and chemokine production was also seen, both as
tracked by cellular mRNA and as cytokines and chemokines
released into the medium (see FIGURE 10C). Conversely, rapid
flow resulted in a virtually complete suppression of these same
cytokines and chemokines. Besides the changes in IL-8 and
GRO illustrated in FIGURE 10C, a whole host of cytokines
and chemokines were found to be similarly regulated, resulting in a net proinflammatory signal induced by slow flow and
an anti-inflammatory response to rapid flow (1600).
PAUL N. HOPKINS
type with frank unresponsiveness to even potent inflammatory cytokines such as TNF- (1390, 2012) or IFN-
(1812). The molecular mechanisms which mediate this
transition have been the subject of intensive investigation
(see FIGURE 11).
1. Endothelial quiescence through PKA and PKB
activities
Phosphorylation by PKA is one of the major negative controls on PAK, thereby suppressing NF-B activation (568).
PKA is activated when integrins containing 1 bind to collagen and other normal basement membrane proteins
(568). PKA inhibits BMP4 signaling and its subsequent activation of NOX1 (358). Upregulation of COX-2 by laminar flow with generation of PGI2 and autocrine signaling
through the IP receptor activates PKA by generating cAMP.
PKG, activated by cGMP (after NO activates soluble guanylate cyclase), can also phosphorylate PAK, disrupting PAK
association with Nck1 (noncatalytic region of tyrosine kinase adapter protein) and altering inflammatory signaling
through MAP4K4 and other pathways dependent on Nck1
(556).
Akt/PKB inhibits forkhead box protein O3a (FoxO3a),
FoxO1, as well as GSK-3 by serine/threonine phosphorylation as noted above under INSR signaling. Active GSK-3
phosphorylates cyclin D1, marking it for degradation and
causing cell cycle arrest with increased susceptibility to apoptosis. Both these effects were blocked when GSK-3 was
phosphorylated by PKB in response to VEGF2 signaling.
Furthermore, PKB blocked endothelial susceptibility to apoptosis as well as induction of IL-1 and TNF- in response
to IL-1 administration, demonstrating an anti-inflammatory, prosurvival effect (1057).
Endothelial PKB activity stimulated by VEGFR2 signaling
is protective in yet another way. HDAC3 can shuttle in and
out of the nucleus and serve as a scaffold for assembly of
1376
PKB phosphorylation also excludes HDAC7 from the nucleus. In the nucleus, HDAC7 blocks transcription of protective MEF2C (989). In the cytoplasm, phosphorylated
HDAC7 binds -catenin and prevents its translocation to
the nucleus (2100).
2. Endothelial quiescence through Tie2 signaling
Tie2 (tyrosine kinase with immunoglobulin and epidermal
growth factor homology domain 2) is a receptor tyrosine
kinase structurally similar to VEGFR2 (86). Tie2 signaling
between adjacent cells, stimulated by angiopoietin 1
(ANGPT1), strongly supports endothelial quiescence and
stability. Laminar shear stress promotes Tie2 phosphorylation and activation in direct proportion to the speed of flow
(991). VEGFR2 can further promote Tie2 activation by a
complex mechanism involving cleavage of Tie1 (1635).
Remarkably, in isolated endothelial cells, ANGPT1 binding
to Tie2 promotes activation and angiogenesis, whereas only
in confluent cells (with intact cell-cell junctions) does
ANGPT1 promote quiescence. The difference depends on
NOTCH signaling and, apparently, the juxtaposition of
vascular endothelial protein tyrosine phosphatase (VEPTP), both of which only occur at intact cell-cell junctions.
VE-PTP limits ERK1/2 activation by Tie2 signaling (1967).
Active Tie2 does bind Grb2-Sos which could activate the
ERK1/2 pathway (86). In confluent endothelial layers,
ANGPT1 ligates and bridges Tie2 receptors on adjacent
At the onset of flow, the adherens junction PECAM-1VEGFR2-VE-cadherin complex was introduced as the
proximate instigator of integrin inside-out signaling (with
resultant JNK and NF-B activation). With continued unidirectional flow, subsequent binding of Gab1 to the complex (through Grb2) may mark one of the earliest steps
toward the transition to quiescence. Gab1 directs assembly
and activation of a signaling complex that includes Src,
PI3K, SHP2, PLC, and Akt/PKB (854, 982). This complex
is also critical for protective ERK5 activation (see below)
(1627) and is inhibited by cigarette smoke which inactivates
VEGFR2 (460). Gab1 and subsequent SHP2 binding is critical for activation of PKA and activation of eNOS by flow
(428). While its signaling is complex (1968), the net effect
of Gab1 appeared protective as deletion of Gab1 led to a
significant increase in atherosclerosis in ANGII-stressed,
ApoE-deficient mice (765) (TABLE 1).
While excess or prolonged superoxide production promotes cellular damage and apoptosis (1345, 1686), controlled production of superoxide and other ROS is critical
for adequate induction of cellular antioxidant systems (FIGURE 11). Physiological ROS sources include NOX enzymes
and mitochondria as reviewed above. Mild oxidant stress
activates a host of antioxidant defense genes which share an
antioxidant responsive element (ARE) in their promoters.
The newer term electrophile response element (EpRE),
rather than ARE, reflects the importance of electrophilic
reactions in triggering this system (208). The transcription
factor Nrf2 is the major inducer of genes containing promoter ARE sequences. Importantly, nuclear Nrf2 accumulation is equally upregulated by laminar and slow oscillatory (back and forth) flow, but subsequent signaling appears to be impaired in cells exposed to oscillatory flow,
resulting in greatly diminished expression of ARE-containing genes.
Nrf2 is bound in cytoplasm by Keap1 (Kelch-like erythroidderived Cap-N-Collar-Homology-associated protein 1) in a
complex that includes a Keap1 dimer together with the E3
ligase Cullin 3 (528, 1722). This complex directs ubiquitination of Nrf2, targeting it for proteosomal degradation.
Covalent modification of key cysteines on Keap1 by reactive electrophilic compounds disrupts the Keap1 complex
with release of Nrf2. Relevant electrophiles include 4-HE
and other oxidized membrane lipids, 15d-PGJ2, nitrated
fatty acids, peroxynitrite, NO, and numerous naturally occuring substances such as cinnamaldehyde (see sect. IIIC1)
(208). Recall that all these electrophiles also promote mitochondrial ROS production. Laminar flow induces COX-2
(1336) followed by increased 15d-PGJ2 availability. In turn,
15d-PGJ2 can covalently bind and inactivate Keap1, allowing nuclear accumulation of Nrf2 (788, 1329). Remarkably, 15d-PGJ2 also covalently binds to and activates the
PPAR receptor as well as covalently modifying NF-B to
prevent its DNA binding (1517). Enhanced eNOS activity
and NO availability leads to NO nitrosylation of Keap1
and Keap1 inactivation (709). Excessive inhibition of ROS
formation or blocking at least modest accumulation of electrophilic byproducts results in reduced Nrf2 availability
and inadequate induction of cellular antioxidant defenses.
Nevertheless, if ROS or reactive electrophile concentrations
rise excessively, then proinflammatory and proapoptotic
signaling is stimulated through ASK1 and other signaling as
reviewed in section IIIC.
Nearly all cellular antioxidant defenses are induced by
Nrf2. These are only alluded to in FIGURE 11. Many of these
antioxidant proteins are required for transition to a quiescent state. For example, glutaredoxin (Grx) was essential
for flow-mediated upregulation of eNOS (1899), and
HMOX1 induction decreased TNF- release by endothelial
1377
Tie2 signaling through RhoA leads to downstream activation of mammalian diaphanous (mDia) which sequesters
Src, thereby limiting endothelial permeability by blocking
VEGFR2-mediated internalization of VE-cadherin. Active
Src can also reduce expression of KLF2 (1907). Tie2 also
inhibits NF-B translocation by promoting A20 activity
through A20-binding inhibitor of NF-B (ABIN2) (86).
PAUL N. HOPKINS
O2
NOX1/2
SOD3
ONOO
NO,
electrophiles
NO
Caveolin-1
p66Shc
O2
Arg
HSP90
O2
eNOS
SIRT1
TXNIP
NO
PKB
H2O2
Trx
CaMKII
PKA
O2
SOD1
Prdx, Gpx,
Cat
PI3K
Pr-SH, H2O
AMPK
Ras
Raf1
MEKK2/3
ASK1
TAK1
MEK1/2
MEK5
MKK4/7
MKK3/6
lipoic acid
H2O2
H2O
Pr-SOH, Pr-SS
PAK
SOD2
Trx
Grx
TrxR
GSR
Pr-SSG
Pr-SH
+
GSH
NO,
electrophiles
IKK
SHP2
Grb2 Gab1
Grb2 Sos
Grb2 Gab1 VEGFR2
signaling
mDia
ERK1/2
ERK5
JNK
Rho
IB
p38
A20 ABIN2
NF-B
PI3K
PI3K
Bad
NO, electrophiles
MKP1
HDAC5/7
Keap1
Cul3
U U
FOXO1,3
15d-PGJ2
PKB
Nrf2
Keap1
SIRT1
SMRT
HDAC3
FOXO1,3
CBP
HDAC5/7
PPAR
NF-B
GSK3
miR-92a
TXNIP
CBP
PCAF
KLF2/4
MEF2A/C
-catenin
PCAF
HDAC3
CBP
KLF2/4
Nrf2, inflammation, ET-1, TF, vWF, PAI-1, PAR1, ANGPT2, ACE
CBP
Maf
1378
NOTCH signaling,
vascular quiescence
DLL4
-catenin
Bcl2
Tie2
signaling
Src
Gadd45
FIGURE 11. Pathways leading to a quiescent, anti-inflammatory, antioxidant state after prolonged exposure to unidirectional, rapid laminar
flow (high shear stress). Besides catalase (Cat), a number of the antioxidant systems have a net effect of converting H2O2 to water. Most
important for quiescence are induction of KLF2/4 and release of Nrf2 with subsequent transcription of genes induced by KLF2/4 and Nrf2.
Electrophiles can covalently bind cysteines on target proteins. Electrophiles include 15d-PGJ2 (which inactivates Keap1 and NF-B while it
activates PPAR), 4-hydroxy-2-nonenal (4-HNE), peroxynitrite, and others. H2S can act as an electrophile and also induces rapid release of Nrf2.
In excess, electrophiles can be toxic. CREBP binding protein (CBP) represents p300 or CBP. Both are histone acetylases (HATs) and therefore
promote transcription. ERK1/2 can phosphorylate CBP to facilitate Nrf2 signaling as well (not shown for clarity). FoxO1, FoxO3a, and HDAC3
are phosphorylated by PKB causing translocation to the cytoplasm. ABIN2, A20-binding inhibitor of NF-B; Cul3, Cullin 3; GSR, glutathione
reductase; mDia, mammalian diaphanous; SMRT, silencing mediator for retinoid and thyroid hormone receptors; SOD1, copper-zinc superoxide
dismutase, the cytosolic SOD; SOD2, manganese SOD, the mitochondrial SOD; SOD3, extracellular SOD.
1379
Among the most important antioxidant defense mechanisms is the Trx-TrxR system. Trx has a dual role as an
antioxidant enzyme (capable of regenerating reduced cysteine from protein cysteine disulfide bonds and reducing
sulfenic acid) and as a redox-dependent inhibitor of ASK1
(289). Importantly, in oxidizing conditions, Trx is bound
by thioredoxin-interacting protein (TXNIP) through disulfide linkage, allowing ASK1 to activate the JNK pathway.
TXNIP is a scaffold protein belonging to the -arrestin
family. TXNIP can shuttle from the nucleus to either cytoplasm or mitochondria where it can inhibit different isoforms of Trx. TXNIP is markedly increased in endothelial
cells in areas of disturbed flow (1918). In the nucleus during
conditions of disturbed flow, TXNIP acts as a corepressor
of KLF2 expression (possibly by recruiting transcription
repressors like HDAC1/3 to the KLF2 promoter). Indeed,
endothelial KO of TXNIP completely reversed the proinflammatory expression of ICAM-1 and VCAM-1 and inhibited leukocyte adhesion in areas of disturbed flow while
overexpression abrogated the upregulation of KLF2 activity seen with laminar flow (1918). Furthermore, overexpression of TXNIP promotes apoptosis. In pancreatic beta
cells, TXNIP was dramatically induced by high glucose and
was a major mediator of glucotoxicity through a TXNIPTrx2-ASK1 pathway (1546). TXNIP expression and nu-
PAUL N. HOPKINS
1380
2000
1800
Nitric oxide
Superoxide
NF-B expression is generally low in atherosclerosis-resistant regions, while it is elevated in predisposed segments in
mouse (696, 864) and pig (1391), and it increases with age
in humans (432). Nitrosylation of NF-B by NO directly
inhibits IKK, stabilizes IB, induces IB mRNA, decreases
NF-B transport to the nucleus, and inhibits NF-B binding
to DNA (1477). Various electrophiles, particularly nitrofatty acids, can also inhibit NF-B activation. Increased
SIRT1 expression with flow promotes deacetylation of the
p65 subunit of NF-B, making it more susceptible to degradation. SIRT1 deacetylation of p300 results in sumoylation of NF-B which may selectively decrease transcription
of a subset of NF-B target genes (197).
eNOS transcription (294). In addition, increased expression of the transcription factor Erg was found in quiescent
endothelium with downregulation by inflammation. Erg
was found to bind promoter regions of multiple inflammatory genes and block NF-B action at these sites (447).
1600
1400
1200
1000
800
600
400
200
0
0
24
ulin binding domain of eNOS. When these lysines are acetylated by an as yet unidentified acetyltransferase, there is
decreased calmodulin binding and diminished eNOS activity. Cigarette smoke inhibits this effect of SIRT1 and
increases eNOS acetylation (80). Laminar flow clearly
increases SIRT1 activity. Furthermore, there is a physical
association of SIRT1 with eNOS which is enhanced by
flow-dependent phosphorylation of eNOS (at S633 and
S1177) by AMPK2 (299). Increased SIRT1 activity with
laminar flow may be stimulated by the modest pro-oxidant environment and increased NOX2 activity which
lead to increased NAD utilized by SIRT1 in deacetylation reactions. Generally, NAD/NADH is a major determinant of SIRT1 activity. Other protective activities
of SIRT1 with laminar flow include upregulation of mitochondrial biogenesis through activation of PGC-1
and increased activities of Nrf1 and liver X receptor
(LXR) (299) as well as repression of p66Shc (2106) and
inhibition of pro-inflammatory NF-B signaling. Aging
leads to diminished SIRT1 and increased eNOS acetylation in mouse arteries and in human endothelial cells
isolated from the brachial artery (433). Interestingly,
low-dose aspirin appears to nonenzymatically acetylate
another lysine residue (K609) in the calmodulin autoinhibitory domain of eNOS leading to enhanced calmodulin binding. The deacetylase HDAC3 reverses this acetylation (869).
1381
Cross-talk between different MAPK pathway kinases appears to be another important mechanism whereby prolonged laminar flow induces a protective endothelial phenotype. ERK1/2 is increased by flow, and although it can
support inflammation, its antiapoptotic effects (such as activating Bcl-2 and eNOS by phosphorylation) may be relevant for transition to a quiescent state. Nevertheless, activation of the ERK5 module is considered most important in
this regard. As depicted in FIGURE 11, activation of MEK5
by flow inhibits activation of the pro-apoptotic, inflammatory JNK (130, 1029). MEK5 activates ERK5 which interferes with signaling downstream of JNK (130). Of primary
importance is the upregulation of KLF2 and KLF4 by ERK5
signaling through phosphorylation and activation of the
transcription factor MEF2C (MADS box transcription enhancer factor 2C) (330, 1332, 1881). Laminar flow further
supports MEF2C action by calcium/CaM-dependent export of inhibitory HDAC5 from the nucleus (1912). Inhibitory HDAC7 can be excluded from the nucleus by PKB
phosphorylation (989). ERK5 also phosphorylates and activates eNOS, inactivates the proapoptotic factor Bad, and
displaces the corepressor SMRT (silencing mediator for retinoid and thyroid hormone receptors), thereby allowing
transcription mediated by PPAR (154). PKC, which is
activated by TNF-, binds and inhibits ERK5 by phosphorylation (1306). ROS and advanced glycosylation end-products (AGE) appear to promote ERK5 sumoylation and inactivation, a potentially important mechanism for diabetesrelated endothelial dysfunction (1977). High blood sugar
and mild hydrogen peroxide exposure together or high hydrogen peroxide alone increased direct physical binding of
RSK1/2 to ERK5. RSK1/2 then phosphorylated ERK5 on
S496 and inhibited ERK5 transcriptional activity followed
by increased VCAM-1 and E-selection expression while
eNOS was decreased. Inhibition of RSK1/2 activity with the
specific inhibitor fluoromethyl ketone-methoxyethylamine
resulted in reduced atherosclerosis in ANG II-treated ApoEdeficient mice (986). In contrast, endothelial-specific, tamoxifen-induced conditional KO of ERK5 led to a 130%
increase in atherosclerosis in LDLR/ mice (986).
PAUL N. HOPKINS
VSMC phenotype (472). These microRNAs are packaged in
vesicles which are secreted by endothelial cells and taken up by
VSMC, followed by induction of a more quiescent, contractile
phenotype in VSMC. Infusion of such vesicles into ApoEdeficient mice resulted in a reduction of atherosclerosis (752).
1382
A number of human intervention trials are underway utilizing various H2S donors (1028, 1098). Interestingly, naturally occuring sulfur compounds in garlic yield biologically
meaningful levels H2S by interacting with GSH in red blood
cells (124). These insights may help explain positive cardiovascular outcomes of several smaller, randomized controlled trials that utilized garlic or garlic extracts. Results
were largely independent of any effects on standard risk
factors (222, 223, 247, 939).
V. INITIATION OF ATHEROSCLEROSIS:
ENDOTHELIAL ACTIVATION BY
DYSLIPIDEMIA
Striking electron micrographs reveal monocytes adhering to
endothelium in atherosclerosis-prone areas just days after
inducing severe hypercholesterolemia by feeding animals
high saturated fat diets (with butter, lard, or coconut oil)
enriched with added cholesterol (492, 493, 1511, 1577).
Such hyperlipidemia clearly provides one or more signals
for activation of already primed endothelial cells found
in atherosclerosis-prone areas (26, 365, 696, 1023). Furthermore, lowering serum cholesterol, even if by only dietary means, rapidly decreases endothelial activation and
1383
In one recent study in ApoE KO mice (2067), high early mortality seen in prior studies of CBS/ mice was avoided by
utilizing a zinc-inducible human CBS transgene in otherwise
CBS-deficient, apoE/ mice. During pregnancy and weaning,
zinc was added to the drinking water, the transgene was activated, and Hcy levels were only slightly elevated, preventing
early mortality. After weaning, zinc was removed from drinking water and the mice developed severe hyperhomocysteinemia and accelerated atherosclerosis, confirming prior observations in conventional CBS/ apoE/ mice (1896). Furthermore, lesion size and elevations in TNF-, MCP-1, and
Ly-6Chi monocytes/macrophages were all proportional to
Hcy levels. However, insights from other studies suggest that
impaired H2S was likely the underlying causal factor. Thus in
vitro studies showed that elevated plasma Hcy inhibited CBS
and impaired H2S production while H2S supplementation
completely reversed inflammation caused by hyperhomocysteinemia (1586). Chemical inhibition of CTH in ApoE KO
mice caused a marked increase in atherosclerosis. This excess
lesion formation was decreased (by 38%) by sodium hydrosulfide supplementation (1921). In another study, sulfur dioxide supplementation markedly decreased lesion development
in atherogenic diet-fed rats (1037). Thus much of the prior
work on Hcy in animal models of atherosclerosis needs to be
reconsidered in light of newer understanding of H2S signaling.
exposed to rapid, unidirectional pulsatile flow. For example, after exposure to disturbed flow there was increased
mRNA for IL-1a, IL-1a receptor, IL-6, IL-8, Cx43,
VCAM-1 (with enhanced response to IL-1 stimulation), Eselectin, MCP-1, fibronectin, vWF, NADPH oxidase,
MMP-1, and MMP-10 while some protective genes such as
eNOS, IL-10, and chemokine CXCR4 were downregulated. A host of antioxidant genes were upregulated by the
concerted actions of laminar flow, and these changes were
associated with greater expression of KLF2 and Nrf2 (528,
529). Besides confirming effects on genes with known links
to inflammation, these studies also provided many new candidate genes to be considered for their potential roles in the
already complex molecular biology of endothelial responses to hemodynamic factors and subsequent atherosclerosis susceptibility.
PAUL N. HOPKINS
myeloperoxidase in activated macrophages or neutrophils
also results in characteristic tyrosine modifications. These
molecular signatures are found in abundance in LDL from
advanced human plaques, whereas modifications characteric of metal-catalyzed oxidation are not (740, 996, 997).
Furthermore, myeloperoxidase-generated reactive nitrogen
species convert LDL into a form avidly taken up by macrophages with conversion to foam cells (1427).
Not only do lipoproteins preferentially accumulate in atherosclerosis-prone areas, but hyperlipidemia itself clearly
increases the permeability of the endothelium and total area
of susceptibility, suggesting direct activation (1577, 1578).
Mechanisms for increased permeability include alterations
in adherens junction function with increased paracellular
permeability, greater susceptibility to apoptosis with increased cell turnover resulting in physical gaps in the endothelium, diminished glycocalyx height, and other factors
(1843). Activation of endothelial RhoA by multiple mechanisms is known to assist in opening endothelial paracellular junctions (1172). Even modest hyperlipidemia induced
by a high-cholesterol diet in pigs (increasing total serum
cholesterol from 74 to 187 mg/dl) tripled endothelial labeled thymidine uptake, an index of endothelial cell turnover (531). Several possible mechanisms for endothelial activation by hyperlipidemia are presented below, and relevant genes are presented in TABLE 4.
The question then arises whether preformed oxLDL activates endothelium to summon the macrophages and/or neutrophils initially or whether oxLDL are formed mainly after
macrophages and neutrophils have arrived and become activated. Yet even if formation of tyrosylated, chlorinated, or
nitrosylated oxLDL requires the presence of activated macrophages or neutrophils, once formed these modified LDL
could then perpetuate endothelial activation and otherwise
promote atherosclerosis (1303, 1304). Unfortunately, the
near absence of myeloperoxidase in experimental mouse
atherosclerotic lesions will make clarification difficult, although humans with myeloperoxidase deficiency are apparently protected from atherosclerosis (1303).
1384
inflammation (1023). As early as 2 h after injection of human LDL into rabbits, grapelike clusters of aggregated LDL
could be seen enmeshed in focal areas of the subendothelial
matrix (1305). VCAM-1 and MCP-1 were expressed by
endothelial cells within 3 wk of starting a high-cholesterol,
coconut oil-based diet in rabbits (1023). The events occuring during these weeks have been the object of intense investigation.
Table 4. Endothelial activation by hyperlipidemia, other risk factors (20 genes tested)
Gene
Model
% A
Function, Comment
TLR1/ LDLR/
NS
TLR2
TLR2/ LDLR/
232%
TLR4
TLR4/ apoE/
255%
TLR6
TLR6/ LDLR/
NS
RP105
RP105/ BMT in
LDLR/
243%
CD14
CD14/ apoE/
NS
TRIF
TRIFLPS2/LPS2
LDLR/
221%
LOX-1 (OLR1)
OLR1/ LDLR/
250%
RAGE
RAGE/ apoE/
DN-RAGE apoE/
252%
278%
AGE-R3
LGALS3/ in fat-fed
C57BL/6J
155%
AR
AR/ apoE/
1180%
Human AR Tg in
diabetic LDLR/
115%
G2A
(GPR132)
G2A/ LDLR/
243%
LPA1/3
240%
Continued
1385
TLR1
PAUL N. HOPKINS
Table 4.Continued
Gene
Model
% A
Function, Comment
LPL
LPL
GPIHBP1
GPIHBP1/
PON1
PON1/ apoE/
PON2
PON2/ in C57BL6/J
fed Paigen diet
human PON cluster Tg
in apoE/
PON1, 2, 3
in aged mice
11,000%
1933%
142%
1166%
221%
ANGPTL2/ apoE/
238%
PARP-1
PARP1/ apoE/
251%
With regard to the mechanism whereby mmLDL might activate endothelial cells, several investigators have suggested
that arachidonic acid in the sn-2 position of phospholipids
in trapped LDL becomes oxidized, resulting in abnormal
phospholipids with structures similar to PAF (1288). This
simplistic model is complicated, however, by findings that
anti-inflammatory electrophilic compounds are also formed
during LDL oxidation and that the above PAF-like lipids
probably do not act through the PAF receptor (131, 196,
871).
1386
IFN- production (apparently a delayed feature which becomes active after TLR4 uptake into endosomes). The activation of IRF3/7 is analogous to the NEMO-IKK1/2 complex activation of NF-B (284). Upon binding to TRIF,
TRAF3 acts as an adapter to recruit E3 ubitin ligases (including Mib1/2) as well as a complex consisting of the scaffold TRAF family member-associated NF-B activator
(TANK) and IKK-related kinases, IKK or TANK-binding
kinase 1 (TBK1). Other scaffolds, NAP1 (NF-B activating
kinase-associated protein 1) and SINTBAD (similar to
NAP1 TBK1 adapter) can act like TANK to bind IKK or
TBK1. K63 ubiquitination of TANK and TBK1 are involved in activation of TBK1, while IKK may be activated
somewhat differently (1032). Such ubiquitination may allow cross-talk between the IRF3/7 and NF-B activation
pathways. Once activated, IKK and TBK1 serine phosphorylate IRF3 or IRF7 causing their activation and nuclear
translocation, followed by transcription of the type I interferon IFN- (which also requires binding of NF-B and
ATF2 to other domains of the promoter). Interest in this
pathway has recently been piqued with findings that knockout of IKK or chemical inhibition of IKK and TBK1 elevates energy expenditure and results in resistance to obesity
or weight loss in fat-fed mice (1470).
ANGPTL2
1387
PAUL N. HOPKINS
RAGE
WBC, platelets,
bacteria
CRP, HSP70,
apoptotic
bodies
oxLDL, oxHDL
electronegative LDL
carbamylated LDL
TGRL remnants
AGE, lyso-PC
TLR2
TLR4
HSP60/70/90 RP105
oxLDL
NOX1/2
TLR4
mmLDL
PEIPC
LPS, SFA,
FN-EDA
MD1
MD1
MD2
MD2
LBP
CD14
LOX-1
MMP1
Rho
Rac1
Ras
Rac1
ARHGEF1
TNF
TIRAP
BMP4
SMADs
MyD88
PKC
ERK1/2
ROCK2
CASP8
TRIF
IRAK1
RIP1 TRAF3
FADD
TRAF6
U
U
U
U
TANK
UU
U
IKK
U TBK1
ROS
Src
TRAM
IRAK2
NF-B
PKC
Syk
IRAK4
MIB1/2
Oct1
PI3K
Bcl2
Bcl-xl
Bad
PKC
Bax
JNK
SOD1
TAK1
IKK2
p38
Apoptosis
JAK
JNK
STAT3
IB
NF-B
IRF3/7
NF-B
STAT3
IFN / etc.
IRF3/7
NF-B
1388
JNK
U
U
ECSIT MEKK1
eNOS
PKB
Early studies with oxLDL demonstrated powerful proinflammatory and apoptotic effects when incubated with endothelial cells. Subsequent studies identified lectin-like oxidized LDL receptor 1 (LOX-1), coded by the OLR1 (oxidized LDL receptor 1) gene, as a scavenger-type receptor
expressed on the surface of endothelial cells which likely
mediated these effects (1545). LOX-1 is also expressed on
monocyte/macrophages and platelets, but unlike macrophages, LOX-1 is the only scavenger-type receptor expressed highly on endothelial cells. LOX-1 is yet another
PAMP-recognizing receptor with a key role in innate immunity (728), being activated by certain gram-positive and
gram-negative bacteria, apoptotic bodies, senescent red
blood cells, activated white blood cells (WBC) and platelets,
advanced glycation end-products (AGEs), lysolecithin, and
LOX-1 ligation also results in powerful proapoptotic and inflammatory responses due to inactivation of PI3K and PKB
(1040, 1082). A major mechanism for these effects appears to
be formation of a complex on the receptor cytoplasmic tail
which includes ARHGEF1 and ROCK2. ARHGEF1 and
MMP1 together activate RhoA (not found in the complex)
which then activates ROCK2. Direct phosphorylation by
ROCK2 inhibits eNOS and PI3K activity (1158). RhoA
may also suppress PI3K through an effect on the actin cytoskeleton. Serine phosphorylations by ROCK2 (and
ROCK1) directly activate MLC while inhibiting MYPT1,
leading to increased myosin phosphorylation, endothelial
contraction, and opening of paracellular junctions to facilitate leukocyte transmigration. Importantly, ROCK2 is
known to activate NF-B in endothelial and other cells.
FIGURE 13. Activation of endothelial cells through LOX-1, RAGE, and TLR4 signaling. 1-Palmitoyl-2-epoxyisoprostane-sn-glycero-3-phosphorylcholine (PEIPC) is one of several oxidized phospholipids that can activate TLR4. Comparable pathways are present in macrophages which can
lead to activation and foam cell formation. CASP8, caspase-8; FN-EDA, fibronectin extra domain A, also termed EIIIA. FN-EDA is an alternatively
spliced FN seen accumulating in artery walls and even plasma of aging mice which can bind and activate TLR4. See text for other abbreviations
and additional explanation.
1389
More complete understanding of LOX-1-mediated intracellular signaling has been frustrated by the lack of usual cytoplasmic domains (1081). Some of the identified pathways
are depicted in FIGURE 13. Octamer binding transcription
factor 1 (Oct1) is an apparently proatherogenic transcription factor activated by LOX-1. PKC activation appears
key in activating Oct1 (1792). Overlapping DNA binding
sites between Oct1 and Nrf2 may lead to suppression of
antioxidant responses generally (627). Oct1 also appears to
repress several CYP enzymes (CYP1A1, CYP2A6/7,
CYP2B6, CYP2C8, CYP2C9, CYP2J2) which can synthesize vasodilatory products such as 11,12-EET. Oct1 may
also be the means through which PKC increases CD40 and
CD40L expression (1019).
PAUL N. HOPKINS
sclerosis progression (829). Generalized KO of LOX-1 reduced atherosclerosis 50% in a high cholesterol/fat fed
LDLR KO model and preserved endothelial function
(1185). Overexpression of LOX-1 in coronary arteries in
ApoE KO mice leads to an atherosclerosis-like vasculopathy (825).
Other modification of LDL may also be sufficient for endothelial activation. Thus exposure of LDL to a proteinase
and cholesteryl-esterase resulted in LDL that stimulated
IL-8 release by incubated endothelial cells, an effect that the
investigators attributed to free fatty acids (1716). Possibly,
the treatment caused conversion of LDL to an electronegative LDL and activation of LOX-1. On the other hand, these
findings may be consistent with recent studies demonstrating that exposure of endothelial cells to saturated fatty acids
leads to a somewhat delayed activation of endothelial cells
through a mechanism dependent on TLR4 and NADPH
oxidase as well as downstream activation of NF-B (1131).
Feeding mice a diet high in saturated fat resulted in similar
activation (905). Incubation with LDL at concentrations of
220 mg/dl (LDL-C), isolated with scrupulous care to avoid
oxidation or other modification, also activates endothelial
cells in culture (2114). However, whether such high concentrations may have included nonoxidatively modified
LDL, such as electronegative LDL, is not clear.
LOX-1 can also mediate inflammatory responses to triglyceride-rich lipoproteins (TGRL) and their remnants (1382,
1626). Interestingly, pioglitazone, through PPAR, blocked
LOX-1 signaling and the LOX-1 mediated increase in
monocyte binding to endothelial cells (1183).
RAGE is generally considered in the context of diabetesrelated modifications to proteins or LDL. AGE accumulates
as products of nonenzymatic reactions between glucose and
1390
Thus LPA binding to its G13-coupled GPCR on endothelial cells activates a RhoGEF followed by RhoA activation
of ROCK2. ROCK2 then phosphorylated its immediate
substrates within 5 min, followed by peak Ser536 phosphorylation of the p65 subunit of NF-B at 30 min and
ICAM-1 and VCAM-1 expression within 4 h (1623).
1391
sclerosis was actually decreased in AGE-R3-deficient animals. However, there was a trend in the opposite direction
in younger animals.
PAUL N. HOPKINS
diated by adenylate cyclase-inhibiting LPA5 (with Gi coupling), probably together with effects from other LPA
receptors. However, in mouse, the predominant effect of
LPA is inhibition of platelet aggregation by LPA4 (with Gs
coupling).
These findings have important implications and should redirect therapeutic investigation to the LPA receptors. LpPLA2 actively converts oxidized phospholipids to LPC
(1129). An inhibitor of Lp-PLA2, darapladib, inhibited atherosclerosis in prior animal studies and was found to reduce
in necrotic core size in a human study (1594). Darapladib is
currently being tested in larger phase III clinical trials
(1509). A more direct approach may prove to be blocking
LPA formation by inhibiting ATX or development of specific LPA receptor inhibitors.
1392
sclerosis (see TABLE 5). CD73-deficient endothelial cells express more adhesion molecules (665).
Anti-inflammatory signaling by adenosine may largely be
through A2BR, with upregulation of CD73 expression on
the endothelial cell surface as well (1551). Deletion of A2BR
increased catheter-induced intimal VSMC hyperplasia, together with increased TNF- expression, effects probably
mediated by bone marrow-derived platelets (2019); yet increased expression of adhesion molecules was seen on endothelium of whole body A2BR KO mice (16). A2BR activation increases cAMP, which is generally anti-inflammatory
and also specifically downregulates expression P2Y1 ADP
receptors in platelets (2018).
In contrast to generally anti-inflammatory A2BR signaling, whole body A2AR deficiency resulted in a 24% reduction of atherosclerosis largely due to a marked reduction in macrophage numbers. Plaques, although smaller,
displayed markers of greater inflammation, with impaired PKA activity (1898). Other effects of A2AR signaling include increased ABCA1 transporter expression and
cholesterol efflux (149) and increased ANG II-induced
NOX2 activation (1779).
P2Y12
ADP
RBC
hypoxia
low pH
Platelet
Monoc
Monocyte
q
Monocyte
Cx37
P2Y1
i
cAMP
Ca2+
cAMP
ATP
Px1
NO
q
i
TPs
PARs
13
P2Y22
P2Y
P2X1
ATP
CD39
ATP
ADP, AMP
ADP
Adenosine
CD73
P 1& 2
P2Y
A2BR
PI3K
Flow
P2Y2
s AC
eNOS
PKB
ATP
VCAM-1
P2X4
eNOS
Cx40
Ca2+
cAMP
EET
PGI2
NO
NO
Cx43
Vasodilation
Vasoconstriction
Ca2+
Cx43
Cx43
Vascular
smooth muscle
K+
P2Y2
P2Y6
P2Y12
P2X1
FIGURE 14. Examples of purinergic signaling. CD39 is also known as NTPDase1 (nucleoside triphosphate
diphosphohydrolase 1). Endothelial P2Y receptors are Gq coupled and stimulate PLC and IP3 release, which
activates IP3R to promote release of ER-stored calcium. Calcium release activates cPLA2 with liberation of free
arachidonate followed by prostacyclin and EET synthesis and secretion. Px1, pannexin 1.
1393
PAUL N. HOPKINS
Model
% A
158%
CD39
CD39/ apoE/
1108%
CD73
Cx40
CD73/ apoE/
endothelial-specific Cre-Lox
Cx40/ in apoE/
mice
1150%
1117%
Cx43
Cx43/ LDLR/
250%
A2AR
232%
A2BR
A2BR/ apoE/
167%
P2Y1
P2Y1/ apoE/
224%
P2Y12
P2Y12/ in C57BL/6
1394
Cx37
Function, Comment
1395
PAUL N. HOPKINS
Monocyte rolling
cAMP
ERM
PSGL-1
E-selectin
Fibronectin
51
P-selectin
Rho
41
ERK1/2
?
Src
Filamin ERM
PYK2
JAMs
Src
Rho
PKC
CysLT1
PLC
Leukocyte
focal adhesion
MyD88
BLT1
ICAM-1
VCAM-1
NF-B
JAK2
-arrestin STAT1
L2
41
NF-B
MAPK
ICAM-1
Rho
Src
PI3K
PLC
Chemokines
i/q
LTC1
12-HETE
LTC4S
LTA4H
13-HODE
LTA4
MAPK
5-LO
12/15-LO
Ca2+
LDL
FLAP
AA
LTB4
PI3K
PKC
Mast cell
Cytokines
PLC
cAMP
MMPs
cPLA2
T-cells
Histamine,
Renin, IL-6,
TNF, PAF
AA
BLT1/2
Macrophage
TNF, IL-1,IL-6,
IL-12, IL-12,
IL-15, IL-18
FIGURE 15. Leukocyte recruitment to the endothelium. While monocyte recruitment is depicted, neutrophils
and several other leukocytes follow similar steps. After entry into the intima, monocytes undergo differentiation
into macrophages, represented by the large cell on the left. Other leukocytes and some of their contributions
to the inflammatory milieu are shown. Leukotrienes and 12/15-LO products act as lipid amplifiers of early
inflammation, much like cytokines. ERM, ezrin, radixin, moesin proteins.
Activated endothelial cells secrete CCL2 (MCP-1) which captures passing monocytes. T-cell receptor (TCR) recognition of
antigens presented by endothelial cells can also incite T-cell
integrin activation and T-cell capture. After transmigration
and transformation, activated macrophages or foam cells
themselves secrete MCP-1, providing a positive feedback signal for additional macrophage recruitment. Th1 cells express
CCR5 and CXCR3 and adhere to activated endothelium expressing CCL5, CXCL9, CXCL10, and CXCL11 (without the
need for TCR activation). Th1 cells enter plaque and secrete
1396
Cathepsin-G
Azurocidin,
Proteinase-3,
TNF, INF,
IL-8, MPO,
LL37, NETs,
Chemerin
Neutrophil
Endothelial
signaling
GPR31
BLT1
IL-6, TNF,
MCP-1, MMPs
Chemokines
Outside-in
signaling
Cdc42
ESL-1
MCP-1
(CCL2)
Diapedesis and
transmigration
: Recptor
: CCR1
: CCR2
: CCR5
: CX3CR1
Rac1
LTB4
Syk
CCR2
Inside-out
signaling
Rh
o
BLT1
Chemokine
CCL5
CCL2 (MCP1)
CCL4
CX3CL1
Table 6. Ligands and corresponding cognate receptors in endothelial-leukocyte interactions (43 genes tested)
Endothelium
Model
% A
Leukocyte
E-selectin
(SELE)
224%
245%
ESL-1, PSGL-1,
KN1A
P-selectin
P-selectin/ LDLR/
(assessed at 37 wk, NS)
(862).
P-selectin/ apoE/ (343).
P-selectin/ in C57BL/6
(1272).
BMT confirms a modest role
for platelet P-selectin (232)
220%
PSGL-1 (CD24,
CD34)
Model
% A
270%
234%
Tethering or rolling
(SELP)
245%
265%
FucT-VII
PSGL-1 (CD24,
CD34)
265%
L-selectin (on
multiple cell
types)
250%
278%
298%
279%
260%
CCR2
262%
247%
267%
286%
262%
Continued
1397
C2GlcNAcT-I
265%
PAUL N. HOPKINS
Table 6.Continued
Model
% A
CCL3
(macrophages,
activated
platelets,
neutrophils,
and mast
cells)
CCL5 (RANTES)
(also in VSMC,
platelets)
231%
CCR1, 4, 5
25%
258%
CCR1, 3, 5
1140%
238%
CCR6
230%
145%
lymphorganogenic
chemokines (also CXCL13).
Secreted by cells in lymph
nodes to promote homing
of DC and naive T-cells
(1077)
CXCL1/ LDLR/ (167).
Effects primarily seen for
macrophage accumulation
in established fatty streaks,
not earlier lesions.
CXCR2
(neutrophils,
mast cells,
monocytes)
262%
CXCR2
see above
CXCR3B
CXCR2
see above
CCL20
(activated
VSMC)
CCL19, CCL21
(activated
VSMC)
CXCL1 (GRO)
CXCL2 (GRO)
CXCL3 (GRO)
CXCL5-8
CXCL4 (platelet
factor 4in
platelets)
CXCL8 (IL-8)
222%
280%
263%
Leukocyte
CCR7
Model
% A
234%
Continued
1398
Endothelium
Table 6.Continued
Endothelium
CXCL9,10,11
CXCL12 (SDF-1)
CXCL16 (also on
VSMC,
monocytes)
CXCL19, 20
CX3CL1
(fractalkine
expressed
primarily in
VSMC rather
than EC)
% A
240%
Leukocyte
Model
% A
CXCR3
(monocytes,
Th1, mast
cells)
0%
1320%
CXCR4
136%
CXCR5
NS
CXCR6 (Th1,
NKT, VSMC)
CXCR7
CX3CR1
157%
285%
250%
222%
250%
Continued
1399
CXCL13
Model
PAUL N. HOPKINS
Table 6.Continued
Endothelium
M-CSF (also
known as
CSF1.
Secreted by
activated
macrophages
as well.)
ICAM-2
Model
M-CSF/
M-CSF/
M-CSF/
M-CSF/
apoE/ (1648).
apoE/ (1443)
apoE/ (397)
LDLR/ (1454)
% A
Leukocyte
286%
284%
271%
299%
CSF1R
(monocytes)
MAC1, LFA1
Model
% A
272%
Slow rolling and firm adhesion (generally after integrin activation), further activation through adhesion molecules
VCAMD4D/D4D LDLR/
(VCAMD4D/D4D has 2-8% of
VCAM/ VCAM-1.
VCAM/ is embryo lethal)
(366).
242%
fibronectin
ICAM-1
E-selectin
ICAM-1, ICAM-2,
fibrinogen,
collagen, iC3b,
LPS
vWF
See Table 10
collagen,
fibronectin
collagen
266%
253%
41 integrin
4 or 1 KO IS embryolethal. An 4
(VLA4,
binding peptide (connecting
CD49D/CD29)
segment 1 of fibronectin) was
infused into LDLR/ mice to block
VLA4 binding and signaling (1622).
Decreased neointimal growth with
VLA4 antibody (581). (Note: 47
also binds VCAM-1.)
51
see above for 1
0%
230%
260%
L2 integrin
(LFA1,
CD11a/CD18)
245%
11 integrin
NS
224%
NS
238%
242%
Continued
1400
VCAM-1,
fibronectin
Table 6.Continued
Endothelium
collagen
Model
% A
Leukocyte
21 integrin
IIb3 integrin
(platelets)
vitronectin (VN),
Thbs1
V3 integrin
Thbs4
MADCAM1
NS
232%
M2 integrin
47 integrin
% A
NS
220%
1160%
Transmigration
JAM-A/ apoE/
Decreased neointimal
formation after wire injury
(2063).
JAM-A
JAM-B
JAM-C (also in
VSMC)
CD99
PECAM-1
LFA1, JAM-A
VLA4, JAM-C
MAC1, JAM-B
238%
CD99
PECAM-1
see Table 3
Ligands and receptors may be shared by several leukocytes and platelets, but monocyte interactions are
generally emphasized. Several of the chemokine interactions are also involved in leukocyte/VSMC cross-talk in
advanced lesions as indicated. Reference numbers are given in parentheses.
have downstream signaling effects that, in a number of instances, are directly opposite those of Gs-coupled receptors
and cAMP (1950). The subunits released by these receptors
have a number of effectors of their own including PI3K and
1401
fibrinogen, vWF,
fibronectin
Model
PAUL N. HOPKINS
PI3K (followed by Akt/PKB activation and downstream signaling) and PLC (releasing IP3 and DAG, followed by calcium release and activation of conventional PKCs). PLC is
further activated directly by active Gq/11 binding while
G12/13 directly activates several Rho GEFs (1950). PI3K
production of PIP3 can lead to activation of downstream tyrosine kinases with phosphorylation of Shc followed by Grb2SOS binding to Shc and then ERK1/2 activation. PYK2 activation by Gq/11 receptors has not been examined extensively
in chemokine receptors, at least in relation to atherosclerosis.
PYK2 increases endothelial permeability after leukocyte binding to endothelial ICAM-1 (54). Most chemokine receptors
are found on the leading edge of migrating cells, and the PIP3
formation helps provide polarization and directionality to the
cell.
The global effect of raising cAMP on expression of inflammation-related proteins was recently examined in an insightful
study (1672). Human neutrophils were activated by a cocktail
including LPS, formyl-methionyl-leucyl phenylalanine, GMCSF, TNF- and IL-1. Then, either an A2AR agonist, PGE2,
or a phosphodiesterase inhibitor were added separately to elevate cAMP. Results were remarkably similar for all three
cAMP elevating agents. Transcription of several anti-inflammatory proteins was increased by cAMP elevation, including
A20, COX-2, SOCS3, phosphatases DUSP1 and 2 (dualspecificity phosphatase 1 and 2, which can inactivate MAPK
by dephosphorylating both tyrosine and serine residues),
NR4A3 (nuclear receptor 4A3), and IER-2 (immediate early
response 2, a GAP for G proteins). Downregulated proteins
included pro-inflammatory transcription factors Egr2/3 as
well as TNF-, CCL3, and endothelin-1. While only neutrophils were examined, a variety of anti-inflammatory effects
can be expected as a result of elevating cAMP in other cells as
well. NR4A3 is a transcription factor whose expression is
induced by NF-B. NR4A3, in turn, can induce endothelial
VCAM-1 and ICAM-1 expression but may also provide feedback inhibition in macrophages or other leukocytes (175,
2096). Whole body KO of NR4A3 decreased atherosclerosis,
suggesting the endothelial effect may be predominant (2096).
3. Leukocyte transmigration
The mechanism for inside-out signaling in leukocytes induced by chemokines was reviewed previously. Firm ad-
1402
hesion and diapedesis requires leukocyte integrin activation as shown in FIGURE 15. Briefly, GPCR binding by
chemokines frees subunits to activate PI3K, PI3K,
and PLC followed by generation of DAG and IP3 which
releases ER calcium after binding IP3R. Mobilized calcium can activate the sequence CalDAG-GEF1-Rap1RIAM-talin- integrin (66, 1605). This signaling occurs
in 1 s. Integrin clustering and up-down polarization are
established within 30 s, while spreading, lamellipodia
formation, and crawling with formation of a clear leading and trailing edge occur in 30 300 s (1783). At the
leading edge of the leukocyte, oriented toward the
chemokine gradient, activated G proteins Rac1 and
Cdc42 mediate lamellipodia and filopodia extension by
serving to both orchestrate the machinery of actin microfiber polymerization, which drives membrane protrusion, and to anchor the growing chains to the cell membrane. Toward the rear, active RhoA mediates uropod
retraction. Firmly adherent integrins anchor the cell to
endothelial adhesion molecules in the middle. As the cell
advances, integrins are detached toward the rear of the
cell and recycled by cellular motors to the front.
1403
While chemokines are often considered a subtype of cytokines, the following discussion focuses on cytokines other
than chemokines. During the ongoing recruitment of macrophages and other leukocytes to the subendothelial space,
numerous cytokines amplify and modulate the inflammatory response. In the context of T-cell activation, the T-cell
receptor recognizes a specific antigen, which, when combined with costimulator activation leads to proinflammatory cytokine production. The expression of the costimulator receptors are often influenced by the cytokines present.
Many cytokines promote leukocyte activation, differentiation, or proliferation while actively inhibiting apoptosis.
Some have inhibitory effects. Cytokines may be secreted or
presented on the cell surface. They may lead to autocrine,
juxtacrine, and/or paracrine signaling. Cytokine classification schemes may be based on structure, post-receptor signaling, proinflammatory or anti-inflammatory effects, or
physiological function. In one notable review, cytokines
were designated as pro-atherosclerotic or anti-atherosclerotic (1766). Others operate in innate or adaptive immunity
pathways or both (1). Results of numerous genetic interventions are reviewed in TABLE 7.
kinases seem also to be able to activate JAK/STAT pathways. Typical JAK/STAT receptors are dimeric. Ligand
binding activates bound JAK tyrosine kinases which
phosphorylate tyrosines on the receptors cytoplasmic
tail. Several signaling molecules can then bind to these
phosphotyrosine sites (such as Grb2-Sos with downstream MAPK activation), but in particular there is binding of STAT transcription factors. JAK then phosphorylates the STAT followed by STAT dimerization and
translocation of the active STAT dimers to the nucleus.
Suppressors of cytokine signaling (SOCS) proteins are
negative feedback inhibitors that can both block the catalytic domain of JAK and ubiquitinate the receptor for
proteolytic degradation.
PAUL N. HOPKINS
Table 7. Initiation and perpetuation of inflammation through cytokine and immune cell signaling (91 genes tested)
Gene
Model
% A
Function, Comment
Cytokines or proteins primarily related to macrophages (for TNF-, IL-1, and related, see Table 1)
/ apoE/
255%
FcRIII (FCGR3,
CD16)
Fcgr3/ LDLR/
230%
FcRIIb
FcRIIb/ apoE/
140%
1318%
IL-6
228%
185%
173%
127%
gp130 (IL6ST
in humans)
liver-specific
gp130/ in apoE/
243%
IL-12
IL-12/ apoE/
252%
IL-18
IL-18/ apoE/
235%
IL-20
intramuscular plasmid
cDNA injection into
apoE/
122%
KO of chain affects both the FcRI (CD64) and FcRIII receptors. Each
have ITAM domains in their cytoplasmic tail. Both are activating IgG
receptors responsible for antibody-dependent phagocytosis and other
signaling in macrophages and other leukocytes. FcRI can act as a
co-receptor with platelet GPVI. Intermediate reductions in
atherosclerosis were seen in apoE/ mice (757). While
opsonized LDL may be a target, these KO mice showed generalized
reduction in inflammation and NF-B signaling which is triggered
through a CARD9-Bcl10-MALT1-TRAF6 pathway (714). Also
confirmed in mice receiving Fc chain deficient BMT (1298).
An increase in IL-10 as well as IFN- production by CD4 cells was
seen. Lipids were slightly higher in double KO mice despite reduced
atheroslerosis (895).
FcRIIb is an inhibitory IgG receptor with a cytoplasmic ITIM domain on
monocyte/macrophages, DC, B-cells.
Only male mice were affected. Shown is effect at 34 wk; earlier effects
were stronger. Inflammatory cytokines in aorta were higher as were
plasma IgG directed against modified LDL (1191)
Only abdominal aorta area at 12 wk assessed. Increase in anti-oxLDL
IgG also seen (2093).
IL-6 is induced by IL-1. Synthesized by macrophages, activated EC,
VSMC, and mast cells in response to PAMPs, IL-1, and TNF. In liver,
IL-6 binding results in release of acute phase reactants including CRP
and SAA. IL-6 signaling, with STAT3 activation, combined with TGF
leads to Th17 differentiation which may be protective. Though IL-6
infusion appears to increase atherosclerosis, effects of KO on
atherosclerosis are variable. Effects were NS for both models in one
study (228%) (918), but atherosclerosis was signficantly increased
in an apoE knockout by 85% (468) and 73% (1552) in other studies.
IL-6 signal transducer (IL6ST) is the human homolog of gp130, a
transmembrane subunit present in all IL-6 related receptors and also
a component of other IL receptors. Marked reductions in serum SAA
seen. Greater relative reduction of plaque earlier. Association of
variants with human disease also reported (1085). IL-6 transsignaling involves complex formation between IL-6 and soluble IL-6R in
plasma followed by binding and signaling through gp130. Transsignaling may be responsible for the most pro-inflammatory aspects
of IL-6 signaling, including suppression of Treg differentiation. A
soluble form of gp130 acts as a natural inhibitor of trans-signaling
and decreased atherosclerosis when infused as a sgp130-Fc fusion
protein. EC ICAM-1 and VCAM-1 were decreased with decreased
macrophage infiltration (1569).
Produced largely by macrophages and DC in plaque and promotes Th1
differentiation and proliferation. Can upregulate CCR5 expression.
Other studies confirm pro-atherosclerotic effects. Potentiates IFN-
production. Shown is atherosclerosis reduction at 30 wk. Effect NS
by 45 wk (918).
IL-18 induces both Th1 and Th17 cells to produce IFN- (together with
IL-12) (471, 918). IL-18-deficient mice have much higher cholesterol
than wild-type mice, and this may modulate the expression of
atherosclerosis. Another group reported more atherosclerosis in IL18 knockout, but this was only true in mice fed the high cholesterol
diet (1400). Overexpression decreases collagen content and
promotes rupture (1300).
IL-20 clearly increased angiogenesis (see Table 6) and may promote
T-cell recruitment to lesions (296).
Continued
1404
FcRI/III
(FCGR1/3)
Table 7.Continued
Gene
Model
Function, Comment
MIF
MIF/ LDLR/
278%
DLL4
anti-DLL4 antibody in
LDLR/
221%
OPN (SPP1)
267%
GDF-15
269%
CB2
113%
(NS)
Ntn1
Ntn1/ BMT to
LDLR/
245%
GRK5
GRK5/ apoE/
150%
NFAT5
NFAT5/ apoE/
BMT into apoE/
mice
286
S1PR2/ apoE/
282%
S1PR3
S1PR3
NS
apoE/
Continued
1405
% A
PAUL N. HOPKINS
Table 7.Continued
Gene
Model
Function, Comment
NS
250%
EP2
EP4
EP2/ LDLR/
EP4/ LDLR/
FLAP
(ALOX5AP)
FLAP
5-LO (ALOX5)
5-LO/ LDLR/
5-LO/ ApoE/
NS
BLT1 (LTB4R1)
BLT1/ apoE/
236%
12/15-LO
(ALOX15 in
human, 12LO in mouse)
12/15-LO/ apoE/
12/15-LO/
LDLR/
12/15-LO/ in
apobec/ LDLR/
12/15-LO/ BMT
into apoE/
human 15-LO Tg in
WHHL rabbit
263%
277%
macrophage-specific
12/15-LO Tg in
apoE/ mice
231%
12/15-LO/ BMT
into apoE/
12/15-LO/ apoE/
170%
ApoE/
macrophage-specific
12/15-LO Tg in
apoE/ mice
12/15-LO/ BMT
into LDLR/ mice
fed high PUF or SAT
NS
246%
256%
245%
227%
NS
262%
BLT1 (B leukotriene receptor 1) is a major LTB4 receptor. Shown is 12week difference at aortic root; no difference in aortic area by this time.
Earlier effects were stronger (744). Others similarly show early effects
only (1693). BLT1 KO macrophages expressed BLT2 and still showed
chemotaxis to LTB4 (1693). Specific BLT1 inhibitor decreased
atherosclerosis in apoE- and LDLR-deficient mice (21). Intermittent
hypoxia greatly accelerated atherosclerosis in apoE-null mice and BLT1
KO essentially eliminated the excess lesion development (1031).
Assessed at 15 wk (aortic sinus) (368)
Assessed at 9 wk (aortic sinus) (605)
Assessed at 8 mo (aortic surface area) (2089). Reduced urinary
isoprostanes in 12-LO KO which correlated with lesions. Marked
reduction of IL-12 by 12/15-LO KO macrophages.
Assessed at 12 wk (aortic surface area). All atherosclerosis effect was
due to bone marrow cells (807).
WHHL rabbits were fed a high-fat, high-cholesterol diet and assessed at
13.5 wk. Enzyme was expressed essentially only in monocytemacrophages (1613)
In chow-fed mice, overexpression of human 12/15-LO in macrophages
increased production of LXA4 (sixfold), protectins (PD1), and resolvins
(RvD1) thought to help resolve inflammation. RvD1 and PD1 also
showed strong anti-inflammatory effects in cultured endothelial cells
(1197).
BMT of 12/15-LO null cells increased atherosclerosis in chow-fed mice
(1197).
Mice fed high fat Western diet for 10 wk (1198). Became
nonsignificant at 14 wk. Similar results with BMT (less
atherosclerosis in recipients of 12/15-LO deficient bone marrow).
Animals fed high fat Western diet in contrast to chow feeding in prior
studies by this group (1198).
In mice fed a high-PUF diet, BMT of 12/15-LO null cells clearly
decreased atherosclerosis (1507). Whole body KO of 12/15-LO was
also clearly protective in mice fed either high PUF or high SAT.
Continued
1406
% A
Table 7.Continued
Gene
Model
% A
Function, Comment
CD28
237%
1133%
B7-1/2
LDLR
1107%
143%
PD-1
PD-1/ LDLR/
173%
PD-L1/2
PD-L1/2/ LDLR/
190%
CD40
CD40/ LDLR/
CD40L
(CD154)
CD137
(4-1BB)
OX40L
(TNFSF4)
NS
CD40/ ApoE/
242%
245%
CD137/ apoE/
239%
CD137/ LDLR/
231%
OX40L/ apoE/
259%
282%
Continued
1407
ICOS
Whole body KO of both B7-1 (CD80) and B7-2 (CD86). B7-1/2 are
constitutively expressed on antigen presenting cells and were found
on plaque macrophages. Their common ligands are CD28 (activating)
and CTLA4 (inhibitory) on T cells. Decrease in atherosclerosis shown
was for intimal area at 20 wk but surface area was NS. Impaired
IFN production by triple KO mice suggested maily Th1 cell reduction
(230).
Opposite results seen with BMT (33). In this model, mostly Treg cells
appeared to affected (647).
Again, Treg proliferation seemed to be most impaired by CD28
deficiency in BMT. Tregs transfer reversed the increase in
atherosclerosis and markedly decreased Th cells in plaques (33).
ICOS (inducible co-stimulator), a positive co-stimulator receptor on
differentiating T-cells that binds ICOSL on antigen presenting cells.
Promotes Treg proliferation especially (less so Th cells). ICOS-deficient
mice had fewer Treg cells and increased Th cells with greater IFN-
and TNF- production (646). No genetic atherosclerosis model
reported for ICOSL.
PD-1 (programmed death 1 receptor) is the receptor for PD-L1/2.
Both CD4 Th and CD8 CTL cells were more activated in PD-1 KO
mice (221).
PD-L1 (programmed death ligand 1) and PD-L2 are coinhibitory ligands
to PD-1 expressed on antigen presenting cells and Treg that bind
their receptor PD-1 expressed on activate Th cells. Aortic root at 19
wk shown. KO mice had many more active T-cells and macrophages
in lesions and higher serum TNF- (645)..
On EC, VSMC, T-cells, B-cells, DC, and macrophages. CD40L
interaction with Mac1 may explain the lack of effect with CD40/.
Treatment of LDLR/ mice with anti-Mac1 antibodies resulted in a
50% reduction in lesion size with fewer macrophages (2119).
Reduction in atherosclerosis quite variable (not significant in thoracic
aorta, only abdominal). Plaques had fewer macrophages and T-cells
and more collagen (1093).
Slightly more significant reduction in atherosclerosis than whole body
CD40 knockout (1093).
Mostly on activated T-cells and platelets but also DC, macrophages, EC,
VSMC. Absence of CD40L had the greatest effect on advanced
lesions and resulted in a more stable plaque phenotype (1092).
PAUL N. HOPKINS
Table 7.Continued
Gene
Model
% A
Function, Comment
Related to neutrophils
/
IRF8
IRF8
BMT into
apoE/
1142%
MPO
1136%
152%
FAAH/ apoE/
221%
CRAMP
FAAH
252%
Ly49A Tg
(overexpressing) BMT
into LDLR/
270%
KLRK1
KLRK1/ apoE/
280%
CD1d1
CD1d1/ apoE/
267%
CD1d1/ LDLR/
247%
J18/ LDLR/
224%
J18
HDC
HDC
HRH1
HRH1/ apoE/
260%
HRH2
Kit
HRH2/ apoE/
KitWsh/Wsh LDLR/
NS
283%
apoE
240%
Continued
1408
Table 7.Continued
Gene
Model
% A
Function, Comment
GM-CSF
GM-CSF/ LDLR/
GM-CSF/ apoE/
Rag1
Rag1/ apoE/
Chow fed
Western diet
Rag2
Rag2/ apoE/
27 wk
40 wk
PRKDC
scid/scid apoE/
CD4
CD4/ apoE/
TCR
TCR/ apoE/
249%
TCR
TCR/ apoE/
249%
235%
1100%
Related to Th1 cells. Listed cytokines are increased after activation, typically by antigen presentation (DC or macrophage),
reenforced by CD40-CD40L costimulation.
T-bet (Tbx21)
T-bet/ LDLR/
STAT4
STAT4/ in BALB/c
fed Paigen diet
IFN (IFNG)
IFN/ apoE/
260%
IFN/ LDLR/
243%
IFNGR1
IFNGR1/ apoE/
258%
IL-2
220%
229%
NS
IL-4
IL-4
apoE
IL-5
227%
or NS
150%
Continued
1409
270%
NS
PAUL N. HOPKINS
Table 7.Continued
Gene
Model
% A
Function, Comment
IL-13
191%
IL-33
IL-33 infusion in
apoE/
262%
ST2
189%
STAT6
STAT6/ in BALB/c
fed Paigen diet
1400%
250%
162%
IL-17R
IL-17R/ BMT
LDLR/
246%
SOCS3
T-cell-specific SOCS3/
(Cre-Lox) BMT
LDLR/
250%
anti-CD3 antibody in
LDLR/
259%
CD25
anti-CD25 antibody in
apoE/
150%
Foxp3
134%
The vaccination reduced Treg cells specifically by about 30% in various tissues
by sensitizing CTL with 1.ninefold increase in splenic T-cells (1852)
220%
TGF-1
TGF- receptor
II (TGFBR2)
IL-10
CYP24
1200%
135%
14336%
Continued
1410
IL-17A
in C57BL/6
fed high fat diet and
infected with C.
pneumoniae
IL-17A/ apoE/
Table 7.Continued
Gene
MHCI
CD8
TAP1
PRF1
GZMB
Secreted IgM
Id3
BAFFR
% A
Function, Comment
Related to dendritic cells (DC). Can be defined as CD11c, MHCIIhi and includes conventional DC derived from nonmonocyte, preDC
and nonconventional DC which include monocyte-derived DC and plasmacytoid DC.
CD74
CD74/ LDLR/
274%
FLT3
FLT3/ LDLR/
168%
Continued
1411
B-cell receptor
IgM
Model
PAUL N. HOPKINS
Table 7.Continued
Gene
Model
Function, Comment
CCL17
CCL17E/E apoE/
250%
CCR4
NS
CCL22
in vitro anti-CCL22
antibody
NS
IFN-
peritoneal injection of
IFN- into LDLR/
165%
IFN
178%
IFNAR1
234%
BST2
Anti-BST2 antibody: in
apoE/ in LDLR/
with carotid collar
250%
186%
1412
now a guiding principle in immunology (1). Innate immunity refers to a system of inherited defenses activated by
pattern recognition receptors (PRR) which bind pathogenassociated molecular patterns (PAMPs) that are invariant
features of invading microbes. Innate responses need no
prior exposure or learning or memory to be fully functional. Innate immune responses are found (at least to some
degree) in virtually all multicellular organisms (836). Adaptive immunity is found only in jawed vertebrates and essentially involves activation of mature T and B lymphocytes.
For an adaptive immune response to be mounted, a protein
or peptide derived from a perceived invader must be recognized as foreign, and this signal must be paired with a pro-
% A
PRR that activate the transcription factors NF-B, interferon regulatory factors (IRFs), and MAPK pathways can
help trigger an adaptive immune response by stimulating
both cytokine release and expression of transmembrane costimulatory receptors. Thus, when LPS binds TLR4 on dendritic cells (DC), macrophages, or endothelial cells, NF-B
is activated, TNF- is secreted (peaking at 1 h), followed
by IL-1 release (peaking at 2 h) and chemokine secretion.
Endothelial cells are stimulated to display selectins and adhesion molecules, while chemokines are trapped in their
glycocalyx. Leukocytes are captured and move toward the
site of inflammation, guided by a chemokine gradient. All
this is in the realm of innate immunity. Macrophages summoned and activated by such an inflammatory stimulus, or
activated DC, typically display costimulatory receptors and
will activate T-cells if the antigens presented match the TCR
of the T-cells present, thereby triggering an adaptive immune response.
Belonging to both innate and adaptive immunity are Fc
receptors that can recognize antibodies bound to antigen.
Some Fc receptors stimulate phagocytosis and cell activation while others mediate inhibition. The response depends
on whether the receptor is linked to a coreceptor that contains an activating ITAM motif (such as FcRI/III) or contains an inhibiting ITIM motif in its cytoplasmic tail (e.g.,
FcRIIb). As reviewed in TABLE 7, ITAM-containing FcRI
and FcRIII were associated with increased atherosclerosis
while FcRIIb, with its ITIM linkage, was protective.
1413
PAUL N. HOPKINS
marrow to splenic red pulp. These cells give rise to M1 type
macrophages and eventually ingest lipid to become foam
cells (1493).
2. Macrophage phenotypes: artificial but useful
constructs based on in vitro cytokine, PAMP, and
DAMP exposure
Incubation of macrophages with different PAMPs, DAMPs,
cytokines, scavenger receptor ligands, and various lipids is
followed by characteristic changes in expression of surface
markers, cytokine and signaling molecules, as well as
changes in lipid handling and propensity toward foam cell
formation. The resulting macrophage phenotypes may have
apparently opposite functions (such as pro-inflammatory
versus anti-inflammatory, or tissue repair promoting).
While recognition of macrophages with widely differing
functions or phenotypes is useful, macrophages in vivo actually display plasticity in modulating between not only
these polarized phenotypes but activation states with intermediate features, depending on stimuli encountered (1237).
1414
1415
PAUL N. HOPKINS
after HbHp-CD163 uptake or erythrocyte phagocytosis
with lysosomal degradation. Mild to moderate oxidative
stress ensues, apparently activating Nrf2 and possibly the
transcription factor ATF1. PKA activation may also be involved. Active PKA can activate ATF1 as well. Additionally, heme or even the iron-free protoporphyrin ring of
heme can inactivate a transcription repressor, Bach1 (Btb
And Cnc Homology 1), which blocks ARE sites activated by
Nrf2 (1144). Both Nrf2 and ATF1 then induce HMOX1,
ferritin, and ferroportin. Active ATF1 additionally increases expression of LXR, followed by LXR and PPAR
activation, with downstream induction of ABCA1 and
ABCG1 and presumably other markers of Mhem such as
IL-10 (199, 201).
phages. Such segregation avoids an otherwise highly inflammatory setting since both heme and hemoglobin are
strong oxidizers of LDL and cellular lipids as are the ROS
generated by M1 macrophages, especially in the presence of
iron. Excess lipid oxidation would generate ligands for
TLR, LOX1, and RAGE as well as cause direct damage to
cellular proteins and DNA, further exacerbating the inflammation.
1416
Fe3 O2 Fe2 O2
Mhem is a macrophage specialized in taking up HbHp complexes and effete red blood cells while rapidly excreting
accumulated free iron through ferroportin without excess
production of ROS. At the same time, it suppresses surrounding inflammation and promotes repair by secreting
IL-10. Such polarization would be particularly advantageous in areas of hemorrhage or traumatic wounds. Furthermore, this Mhem phenotype actively mobilizes intracellular cholesterol to HDL through upregulated ABCA1 and
ABCG1, thereby elegantly dealing with the cholesterol derived from phagocytosed erythrocytes while being resistant
to additional lipid accumulation by downregulation of
SR-A and CD36. Thus handling of heme and iron by Mhem
is physically segregated from the lipid accumulation and
superoxide production that occurs in M1-type macro-
Recently, the effect of hepcidin reduction by pharmacological means was tested for its affect on atherosclerosis in
ApoE-deficient mice (1526). A small molecule inhibitor of
SMAD1/5/8 activation (LDN 193189) was used to reduce
hepatic expression of hepcidin. Macrophage ferroportin expression was increased and atherosclerosis area in the aortic
sinus and aorta were decreased 4350%. Furthermore, lesional macrophages had less lipid, expressed more ABCA1
and much more ABCG1. In vitro, peritoneal macrophages
from the treated mice demonstrated greater cholesterol efflux together with reduced hydrogen peroxide production.
These in vitro macrophage findings were reversed by incubation with hepcidin. Certainly, before such a compound
could be considered for human use, longer term testing for
effects on iron homeostasis in vulnerable tissues such as
liver, pancreas, and myocardium would be necessary.
3. Other cytokines associated with macrophages
As suggested by the above discussion, certain cytokines are
classically associated with specific leukocytes. Those studied for effects on atherosclerosis are included in TABLE 7.
1417
Reduced inflammatory responses by iron-depleted macrophages have also been reported. In mice with hepcidin deficiency due to KO of the hemochromatosis gene (HFE), an
oral Salmonella challenge resulted in much less intestinal
inflammation (1901). Macrophages from these HFE/
mice showed increased ferroportin expression and decreased intracellular iron with reduced inflammatory responses when challenged in vitro with Salmonella or LPS.
These diminished inflammatory responses to Salmonella or
LPS were reproduced in wild-type macrophages by iron
depletion with iron chelators or by overexpression of ferroportin.
PAUL N. HOPKINS
Besides atherogenic TNF- and IL-1, macrophages secrete
IL-6, IL-12, and IL-18 in response to LPS (3 h peak). IL-6
causes synthesis of acute phase reactants (and hepcidin) in
the liver but was only variably associated with atherosclerosis in genetic models (see TABLE 7). IL-12 and IL-18 were
atherogenic. They stimulate T-helper 1 cells (Th1) to secrete
IFN-. KO of IFN- or its receptor IFNGR1 decreased atherosclerosis (see TABLE 7 under Th1 cells). IL-12 also stimulates cytotoxic CD8 T lymphocytes (CTL) and NK cells
to increase their cytotoxic activity. Other interleukins secreted by macrophages include IL-23 and IL-27, but their
effects on atherogenesis have apparently not been studied.
Growth differentiation factor 15 (GDF-15) is an atherogenic cytokine in the TGF- superfamily. GDF-15 is secreted by macrophages and promotes macrophage accumulation in a CCR2-dependent manner (392).
1418
M-CSF is a chemokine-like cytokine whose cognate receptor is CSF1R (see TABLE 6). M-CSF production is induced
by oxLDL in endothelial cells and VSMC. It appears to be
key for growth and survival of macrophages. M-CSF also
stimulates production of other cytokines and may induce
transformation of VSMC into foam cells (918). Deficiency
of M-CSF results in macrophage-deficient mice which also
lack osteoclasts and are osteopetrotic (op). The op/op
mouse is homozygous for a naturally occurring base pair
insertion in M-CSF leading to a premature stop codon with
marked reduction in M-CSF. Op/op mice are highly resistant to atherosclerosis. M-CSF/ also showed substantial
reduction of atherosclerosis as did KO of its receptor (see
TABLE 6). Almost all the effect of M-CSF on atherosclerosis
was attributable to vascular wall production or other cells
other than bone marrow derived (1602).
Atherosclerosis was clearly reduced by blocking BLT1 either by genetic deletion (as in the intermittent hypoxia
model noted above as well as in other studies of genetically
hyperlipidemic mice), or by pharmacological inhibition of
BLT1. Surprisingly, however, most of these studies showed
effects only at early time points with little or no atherosclerosis reduction at later time points (1428) (see TABLE 7).
Inhibition of CysLT1 with montelukast also decreased atherosclerosis in hyperlipidemic mice in a relatively shortterm (16-wk) study (1428). Follow-up of a very large Swedish drug registry found only suggestive effects for prevention of stroke or MI associated with montelukast use (824).
Why inhibition of BLT1 (or possibly CysLT receptors) does
not result in more durable inhibition of atherosclerosis remains difficult to explain. Certainly, redundancy of chemokine signaling, with effects restricted to early progression,
may explain findings in mice. Nevertheless, 5-LO is increased in unstable human lesions, and further studies in
human populations may still be fruitful (1428).
Since FLAP facilitates AA loading onto 5-LO, inhibition or
deletion of FLAP and/or 5-LO might be expected to decrease atherosclerosis. Nevertheless, recent studies with genetic deletion of 5-LO or FLAP consistently resulted in little
or no effect on atherosclerosis (see TABLE 7) (253, 1428).
1419
PAUL N. HOPKINS
BLE 7),
The above paragraphs briefly summarize the apparent proinflammatory, proatherogenic effects of 12/15-LO. Thus, in
activated cells, this enzyme appears to produce 12-S-HETE
which may act through a specific GPCR to activate MAPK
and NF-B with markedly increased endothelial expression
of adhesion molecules. KO of 12/15-LO reduced atherosclerosis in most studies while nonphysiological transgenic
overexpression, especially in endothelial cells, markedly increased atherosclerosis. If these were the only effects, the
discussion of this intriguing enzyme could have been much
abbreviated.
1420
To summarize findings regarding 12/15-LO and atherosclerosis, the major effect of the gene may be protective in the
setting of a low-fat diet due to the role of 12/15-LO in
lipoxin production. The fact that 5-LO can supply a precursor for lipoxin production may help explain the generally
negative results from 5-LO and FLAP deletion studies as
well. However, at least early in the course of rapidly progressing atherosclerosis produced by high-fat feeding in hyperlipidemic animals, the pro-inflammatory signaling effects from 12-HETE produced by 12/15-LO seem to predominate. This assessment would suggest that in the human
setting, where hyperlipidemia is generally much less severe
and plaques develop over decades, one might expect a protective role for 12/15-LO activity. Indeed, this seems to be
the case as manifest by a 1.3- to 1.6-fold increase in risk of
CAD associated with carrying a single copy of a low-activity 12/15-LO variant in two large epidemiological studies
(84). Yet even here, the ambiguity continues since the rare
homozygotes were, if anything, protected.
6. Neutrophils are involved in atherosclerosis: at
least in early stages
Many of the same receptors and chemokines reviewed in
relation to monocyte recruitment and transmigration also
mediate neutrophil recruitment into atherosclerotic
plaques. CXCR2 is more strongly related to neutrophil recruitment and was strongly atherogenic as was at least one
of its ligands, CXCL1 in hyperlipidemic mice (see TABLE 6).
In ApoE-deficient mice, neutrophil recruitment to larger
arteries early in atherogenesis seemed to depend on CCL5
from platelets being deposited on endothelial cells and then
being recognized by CCR5 on neutrophils. A high-fat diet
increased neutrophil production in the marrow and mobilization into the bloodstream while also decreasing removal
rate from the circulation. Furthermore, neutrophil depletion inhibited plaque development in these mice at 4 wk but
not at later time points (445).
Earlier studies often lacked the ability to specifically detect
neutrophils in plaques but more recent techniques clearly
disclose the presence of neutrophils in early plaques of hyperlipidemic mice (1649, 1858). Neutrophils were strongly
associated with unstable plaques in coronary arteries in a
human autopsy series (1285). Similar evidence was presented for human carotid plaques (827). Neutrophil extracellular traps (NETs) have recently been observed in and on
human plaques (1179). Neutrophils recruited to early
plaques are found in shoulders near macrophages, but undergo apoptosis and efferocytosis relatively rapidly after
they move into plaques (1649). Neutrophils adhering to
endothelium release azurocidin which facilitates monocyte
adhesion. Furthermore, proteins released from neutrophil
granules, such as proteinase 3, directly activate chemokine
1421
PAUL N. HOPKINS
production by endothelial cells and appear capable of
greatly increasing the affinity of certain chemokines for
their receptors on monocytes. Neutrophil cathelicidin
(LL37) promotes differentiation of monocytes into more
inflammatory macrophages (1846); surprisingly, LL37 also
promoted endothelial recovery after injury and limited neointimal hyperplasia (1650). Neutrophils also help promote
macrophage activation by secreting TNF-, IL-8, and
IFN- as well as myeloperoxidase (444). Neutrophil phagocytosis is triggered by innate cues followed by prodigious
release of MPO (myeloperoxidase).
7. Natural killer T-cells and related
1422
1423
PAUL N. HOPKINS
macrophages and DC present antigens through the MHCII
receptor, secrete cytokines TNF-, IL-6, IL-12, and produce large amounts of NO through iNOS and ROS through
NOX2 (286).
In contrast to Th1, Th2 develop in response to IL-4 stimulation and go on to produce more IL-4, a cytokine important in promoting B-cell antibody production (with permission from antigen-specific activated Th2 cells). A major function of Th2 cells and IL-4 appears to be local
stimulation of macrophage proliferation (846). IL-4, as well
as IL-13, also cause alternative macrophage activation to
M2 cells. M2 were often considered anti-inflammatory.
However, M2 are induced to express MHCII as well as
phagocytic receptors and different chemokines (CCL2,
CCL7), all in addition to the anti-inflammatory IL-1 receptor inhibitor IL-1Ra. In M2, arginine is diverted by arginase-1 away from NO production to reparative ornithine
and polyamines (286). Th2 can produce IL-5 which seems
to be important to activate eosinophils in helminth infections, but importantly for atherosclerosis, IL-5 also promotes B1a production of natural antibodies.
1424
Rag1 and Rag2 form a tetramer known as V(D)J recombinase with Rag1 making a nick in the DNA at coding/noncoding junctions of variable (V), diversity (D), and joining
(J) gene segments of immunoglobulins or corresponding
receptors in developing lymphocytes. DNA rearrangements
and annealing follow. Rearranged DNA then directs transcription and translation of both mature Ig and T-cell receptors (TCR). Without normal surface expression of Ig or
T-cell receptors, developing lymphocytes undergo apoptosis. Rag1 or Rag2 deficiency thus leads to complete absence
of both T and B lymphocytes and immunodeficiency. However, knockout of Rag1 or Rag2 would have also eliminated
the anti-inflammatory T-regulatory (Treg) cells together
with Th cells.
Later, other models clearly showed that suppression of
adaptive immunity (particularly Th cells) led to protection
from atherosclerosis (e.g., scid/scid mice, CD4 KO, TCR
subunit KO see TABLE 7). CD8 CTL seem to have less of
a role as CD8 KO had little effect on lesions in ApoE or
LDLR-deficient mice (TABLE 7) (469, 470). Adoptive transfer, transplant, and several knockout studies in mice further
demonstrate that both arms of the immune system are major contributors to atherosclerosis (61, 580, 1130, 1933).
Indeed, current models of atherosclerosis depict not only a
variety of immune cells in the plaque (particularly macrophages and T-cells), but an inflammatory lesion richly endowed with numerous B-cells in adjacent adventitia as well
(580).
Th1 cells appear strongly proatherogenic given the marked
reduction in atherosclerosis in mice with KO of the costimulatory CD40L as well as models of IFN- signaling interruption (see TABLE 7). Deletion of T-bet, a transcription
factor for differentiation of the Th1 subset, reduced atherosclerosis signficantly but modestly. Markedly decreased
atherosclerosis was seen in another study in which ApoE
KO mice were treated with a chemical agent that inhibits
Th1 differentiation (984).
IL-4 is a key Th2 cytokine that promotes M2 macrophage
activation. Effects of genetic manipulation or infusion have
been quite mixed. Some studies show some reduction in
atherosclerosis with blocked IL-4 signaling, while a more
recent study using multiple models (IL-4 KO in ApoE- or
LDLR-deficient mice, with or without ANG II infusion, and
IL-4 infusion). None of the models showed an effect on
atherosclerosis (913). Other Th2 signaling, namely, release
Macrophage deactivation occurs in response to Treg cytokines TNF- and IL-10 with downregulation of MHCII
receptors. Importantly, ingestion of apoptotic cells (if these
targeted cells do not themselves express or contain danger
signals) also leads to macrophage deactivation and production of TNF- and IL-10 (286). Macrophages and neutrophils can recognize targets coated by antibody by their
Fc receptors, while other receptors recognize complement
or lectin-coated invaders. Receptor binding is followed by
phagocytosis and killing. Thus macrophages and neutrophils are effectors for both innate and adaptive immunity.
In contrast to the above study showing suppression of atherosclerosis by a bolstered Treg response, adoptive transfer
of bone marrow that was deficient in Treg into irradiated
mice led to increased atherosclerosis (33). The transcription
factor Foxp3 is critical for differentiation to and maintenance of the Treg phenotype. Artificially inducing a cytotoxic response toward Foxp3-bearing cells (by vaccination
with DC transgenically modified to over-express and present Foxp3) decreased Treg and increased atherosclerosis
(1852). Multiple additional genetic manipulations that impair Treg activity also promote atherosclerosis (see TABLE
7). More recently, tolerance to specific human APOB-100derived peptides was induced in ApoE-deficient mice by
subcutaneous delivery of relatively low doses of the peptides subcutaneously without adjuvant. This was achieved
by an osmotic minipump implanted for 14 days. This
tolerogenic protocol led to increased Treg activity and a
reduction in atherosclerotic plaque development as well as
arrest of further progression of plaques. Treg from APOB100 tolerant mice specifically suppressed proliferation of
Th1 cells from mice that had been sensitized to the same
APOB-100 peptides. Thus tolerogenic Treg suppress cells
that actively present the specific antigen recognized by the
Treg. Depletion of Treg abrogated the antiatherosclerotic
effect of the treatment (751). These studies also identify
APOB-100 as a likely target of proatherogenic adaptive
immune responses.
Increasing the number and activity of Treg appears to be the
main mechanism whereby repeated oral or nasal administration of low to moderate doses of various antigens (induction of mucosal tolerance) leads to a reduction in atherosclerosis. Targets for tolerance induction have included oxLDL (with decreased atherosclerosis after an oral tolerance
protocol), malondialdehyde treated LDL (no effect) (1859),
HSP60 (reduced atherosclerosis) (1860), HSP65 (reduced
atherosclerosis) (1142), and 2-glycoprotein I (reduced
atherosclerosis) (606) in addition to the APOB-100 studies
1425
The role of CTL in atherosclerosis may need to be reassessed based on recent data. Consistent with older evidence
that CTL are less important for atherosclerosis, deletion of
TAP1 or CD8 in ApoE-deficient mice had no effect on atherosclerosis (932). However, complete absence of CTL or
TAP1 during development may lead to compensatory increases in Th, NK, or T cell responses or absence of a
strongly anti-inflammatory CD8 regulatory CTL (972).
CD8 CTL are said to be the predominant leukocyte in
advanced human plaques, constituting up to 50% of such
cells (972). In ApoE-deficient mice, antibodies against either the or isoform of CD8 decreased atherosclerosis
while reconstitution of lymphocyte-deficient mice with
CTL increased atherosclerosis. Transfer of CTL deficient in
either perforin, granzyme B, or TNF-, however, resulted in
much less of an increase in atherosclerosis compared with
transfer of wild-type CTL. In this study, CTL appeared to
target macrophage, VSMC, and endothelial resulting in increased indexes of apoptosis, necrosis, several markers of
inflammation (such as MCP-1) and increase necrotic core
size (972). The authors suggest that in developing plaque,
MHCI may present peptides derived from oxLDL (although no MHCI antigenic peptides promoting atherosclerosis have been identified directly) to CTL promting release
of their granules containing perforin and granzyme B, the
main cytotoxic molecules utilized by CTL. The resulting
necrotic cells could promote sterile inflammation and increased apoptosis of surrounding macrophages and other
cells.
PAUL N. HOPKINS
above. In addition to these self targets which had been
hypothesized to be involved in the atherogenic sensitization
of Th1 cells, mucosal tolerance protocols directed against
pneumococcus, or even measles virus envelope resulted in
reduced atherosclerosis, suggesting there may be multiple,
unsuspected potential antigenic targets as well the possibility of nonspecific effects from simply increasing Treg numbers with increased secretion of IL-10 (30, 241). Indeed, the
ovalbumin sensitization study reviewed above would suggest that the Treg stimulus need not be directly involved in
atherosclerosis.
2. Effects of costimulatory and coinhibitory
receptors on Treg and atherosclerosis
1426
1427
PAUL N. HOPKINS
Th2 cytokines is repressed while TGF- and IL-10 are induced. Although IL-2 production is supressed, CD25 (the
IL-2R subunit) is increased along with STAT5 activation,
facilitating Treg survival and proliferation. Expression of
the coinhibitory receptor CTLA4 is upregulated as is Cbl-b.
The micro-RNA miR-155 is increased which suppressed
SOCS1 and thereby increases STAT5 signaling (1083).
STAT5 signaling supports Foxp3 production as well as
many other functions including hematopoeitic cell proliferation.
1428
1429
Utilizing a very different approach, transgenic mice expressing human APOB-100 were immunized with human oxLDL followed by generation of T-cell hybridomas. The investigators were surprised to find that none of the hybridomas recognized oxLDL epitopes, but only native LDL or
intact APOB-100 peptide. Nevertheless, the sensitized Th1
cells could apparently license mature B-cells to produce a
much wider array of antibodies against oxLDL as well as
native LDL. Inhibition of Th1 cells sensitized to LDL with
an antibody specific to the sensitized T-cell receptor decreased atherosclerosis by 65% in APOB-100 expressing
LDLR/ mice, suggesting but not proving a proatherogenic role for anti-LDL IgG or anti-oxLDL IgG (756). At
the very least, these results clearly contrast with the protection seen in studies using immunization protocols that actually ended up promoting production of anti-oxLDL IgM
natural antibodies. Possibly related to this finding, a proteolytic fragment of native APOB-100 present in human
atherosclerotic plaques was recently identified that could
activate macrophages and may lead to activation of both
innate and adaptive immune responses directed against
APOB-100 or a naturally derived proteolytic fragment
(897).
PAUL N. HOPKINS
deficient only in B cells due to absence of IgM chains
(MT/). This study showed B2 cells can increase atherosclerosis in the absence of T cells, but that a much greater
increase occurs when T cells are present (969).
Deletion of the BAFF receptor (BAFFR), necessary for B2
development, was also recently shown to be protective in at
least two studies (968, 1528). Like the anti-CD20 antibody
infusion studies, BAFFR deficiency resulted in marked selective reduction of B2 cells with little effect on B1a cells.
However, as noted above, these data cannot resolve the
issue of IgG secretion separately from other B-cell activities.
1430
At least some additional functions of B-cells remain unexplained with apparently protective effects from mature Bcells in certain contexts. Id3 (inhibitor of DNA binding 3,
also known as inhibitor of differentiation 3) is a protein
than can form heterodimers with various transcription factors and inhibit their binding to DNA. In B-cells Id3 apparently limits proliferation and Id3 mutations are frequently
found in Burkitts lymphoma. Deletion of Id3 resulted in
impaired homing of B-cells to the aortic adventitia, an increase in macrophage content of plaques, and increased
atherosclerosis (437). This effect was not due to a reduction
in B1a function or lower IgM. The mechanism of protection
from these adventitial B-cells remains to be explained.
alternatives include RAGE and LOX-1 previously reviewed. In addition, hyperlipidemia, diabetes, infection,
and injury can stimulate expression of various ligands of the
KLRK1 receptor on endothelial cells and macrophages,
among other cells. Activation of the KLRK1 receptor causes
vigorous production of cytokines by NK, NKT, and other
cells while KO of KLRK1 markedly reduced atherosclerosis
as noted previously (1118, 1989).
In many animal studies, modest caloric restriction with reduced insulin signaling increases life span and is associated
with improved mitochondrial biogenesis as well as greater
removal of defunct or impaired mitochondria by autophagy
(or mitophagy) (695). In 18 humans attempting to follow a
calorically restricted but nutrient-rich diet, carotid atherosclerosis appeared to be reduced compared with 18 controls
(532). The apparent benefit of caloric restriction, however,
may be most relevant to relatively overfed animals (or humans) as recent data suggest no survival benefit for caloric
restriction in already lean monkeys (1160).
2. Cell senescence: in part aggravated by excess
ROS and TNF- signaling
While still a controversial topic, cell senescence associated
with multiple cell divisions may be relevant. After several
replications, porcine endothelial cells showed increased
NF-B activity, increased oxidative stress, reduction of several antioxidant enzymes, and increased p53 and apoptosis
(993). Aging arterial VSMC tend to be modulated toward
the secretory phenotype (1903). In general, aging appears to
lead to marked increases in TNF- signaling (particularly
from membrane-bound TNF-), NF-B activation, and
ROS production in endothelial as well as other cells of the
arterial wall (1538, 1834). Even aging skin demonstrates a
gene expression pattern characterized by increased NF-B
signaling (14). One of the major contributors to the increase
in NF-B signaling with age may be accumulation of fibronectin (as documented especially in hypertensive rats)
(826, 976). Fibronectin might be expected to accumulate
particularly in vascular areas hemodynamically prone to
atherosclerosis. As noted above, high glucose, as seen in
diabetes, promotes increased ROS and mitochondrial dysfunction. Endothelial cells from Zucker diabetic rats were
strongly prone to premature senescence with evidence of
marked improvement after administration of the peroxynitrite scavenger ebselen (210). In general, evidence that endothelial senescence may contribute to vascular disease is
1431
PAUL N. HOPKINS
accumulating, but its relevance to human disease has yet to
be demonstrated (488).
The above considerations regarding age-related changes in
the vasculature led to the hypothesis that there may be
greater susceptibility to atherosclerosis initiation, promotion, and progression in older versus younger arteries exposed to the same standard risk factors such as dyslipidemia
and hypertension. Recent evidence suggests this may indeed
be the case.
1432
B. Hypertension
Pressure and flow are the first risk factors to which the
endothelium is exposed. Blood pressure in the arterial range
is an essential requirement for the development of atherosclerosis. Venous atherosclerosis does not occur even in
patients with homozygous familial hypercholesterolemia
(226). The relatively rapid progression of atherosclerosis in
saphenous veins used in coronary artery bypass suggests
there is nothing uniquely resistant about veins themselves.
Normally, the pulmonary arterial circulation with its systolic pressures of 1222 mmHg is another entirely protected
site. Nevertheless, with pulmonary hypertension, atherosclerotic plaques are commonly seen (620). While other
mechanisms leading to altered permeability cannot be excluded, including pressure- or stretch-induced activation of
the proinflammatory protein BMP2 (359), the major mechanism whereby higher blood pressure promotes atherosclerosis appears to be through increased pressure-driven convection of LDL and other lipoproteins into the intima
(1713). LDL accumulation in the intima of pressurized rab-
C. Exercise
Even modest exercise serves as a potent stimulus to improve
endothelial function as measured by vasodilation in response to acetylcholine, particularly in older animals and
humans (415, 457, 1580). Exercise increases eNOS expression in animals and reduces restenosis induced by catheter
injury (823, 1979). Exercise stimulates phosphorylation of
eNOS by PKB (2078). Aged mice had much higher levels of
nitrotyrosine (a marker of chronic oxidative stress) in their
aortas compared with younger mice, and this was markedly
reduced by modest amounts of voluntary running on an
exercise wheel (457). FIGURE 11 illustrates several of the
mechanisms thought to mediate these benefits. Exercise
acutely increases flow, and hence the shear-stress experienced by endothelial cells in many vascular beds, not just
the exercising limb. Furthermore, the NO released alters
transcription of eNOS and leads to increased expression of
eNOS protein. Thus inhibition of eNOS activity abrogates
the exercise-induced increase in eNOS protein expression
(1335).
Recently, exercise was found to increase telomerase activity
in mice. Telomerase then synergized with eNOS to confer
impressive resistance to endothelial stress. In exercised
mice, LPS induced far less endothelial apoptosis, and this
effect was dependent on the presence of both telomerase
1433
PAUL N. HOPKINS
and eNOS. Exercise also decreased p53 and associated
checkpoint kinases and increased the p70 component of the
DNA repair enzyme Ku in this study (1943). High shear
stress induces increased SIRT1 expression, suggesting exercise may potentially have anti-aging effects in arteries as
well. Other effects of exercise on endothelial function including increased PGI2, thrombomodulin, and plasmin production also accrue (1145).
E. Diabetes
D. Hyperlipidemia
Extensive effects of hyperlipidemia to initiate atherosclerosis are reviewed above. Not only does hyperlipidemia activate the endothelium and otherwise promote atherosclerosis, but effects on circulating white blood cells are also apparent. Thus monocyte expression of CCR2, the cognate
receptor for MCP-1, was increased twofold when incubated
in serum from hypercholesterolemic patients with average
LDL of 167 mg/dl compared with persons with LDL of 80
mg/dl (706). This upregulation was thought to be due to
uptake of LDL through the LDL receptor and may have
been due to an increase in membrane cholesterol. Subsequent studies suggest calcium transients may be involved in
this activation with sensitization of monocytes so that they
have increased production TNF- and IL-8 when subse-
1434
Not only does elevated glucose provide a substrate for oxidation and ROS production, but in some way, metabolism
of glucose prior to pyruvate entry into mitochondria causes
a major shift in mitochondrial morphology from elongated
tubelike structures to smaller fragments. Mitchondrial fission and fusion are fundamental features of mitochondrion
biology, being intimately involved in distribution of mitochondria into dividing cells, mitochondrion turnover, apoptosis, mitophagy, and ATP generation (583, 1615, 2044).
Fission is triggered by phosphorylation events on the cytoplasmic GTPase dynamin-like protein 1 (DLP1) causing
binding to its target Fis1 (fission 1) which is anchored on the
outer mitochondrial membrane. Inhibition of mitochondrial fragmentation by various measures prevents the increase in ROS produced by cells exposed to excess glucose
(1615, 2047, 2048). Glucose-induced mitochondrial fragmentation appears to involve rapid early calcium entry into
the cell leading to ERK1/2 activation, followed by phosphorylation of DLP1 and mitochondrial fission. Pyruvate
supplies the metabolic fuel to generate ROS by the fragmented mitochondria (2047, 2048). Mitochondrial fission
and ROS production occur within 1530 min after exposure of cells to high glucose, followed by a rapid reduction
in mitochondrial fission and ROS production only to be
followed by a more prolonged secondary rise in mitochondrial fragmentation and ROS production peaking at 16 h
(2048).
Remarkably, incubation of endothelial cells in high glucose
(2530 mM) for 16 h then returning the cells to normal 5
mM glucose resulted in elevated ROS production that persisted for as long as 6 days. With some cells, only 6 h
exposure was sufficient to cause persistent ROS production
(203, 467). This elevated ROS production was accompanied by altered histone methylation that favored induction
of the p65 component of NF-B and endothelial activation
(203, 467). The mechanism(s) responsible for this hyperglycemic memory is under intense investigation and may
include the following: mitochondrial dysfunction perpetuated by mitochondrial fission (583, 2044); activation of
PP2A by ROS, RNS, and ceramide (especially with increased availability of palmitate) followed by PP2A-mediated dephosphorylation and inhibition of Akt with activation of apoptotic proteins such as Bad (inhibited by Akt), as
well as inhibition of AMPK (450, 944); overactive GSK3
blocking assembly of a mitochondrial complex which consists of voltage-dependent anion channel (VDAC), hexokinase (HK), and adenosine nucleotide transporter (ANT)
which is responsible for normal shuttling of ADP back into
the mitochondria after phosphorylation of glucose (1392,
1495); mitochondrial localization of p66Shc after its phosphorylation by PKCII (246, 1377, 1712); and impaired
SIRT1 activity (2098) with impaired suppression of p66Shc
1435
An elegant, unified model to explain the seemingly disparate aspects of endothelial dysfunction in diabetes has
been forwarded (215, 216, 612). The endothelial cell is
one of a handful of cell types that are unable to regulate
glucose entry. Glucose moves into the cells through
GLUT1 transporters at a rate proportional to external
concentrations. Hyperglycemia thus leads to high intracellular glucose, a rapid flux of substrate through the
glycolytic pathway, and an abundance of pyruvate for
transfer into mitochondria. Synthesis of acetyl CoA ensues followed by ample generation of reducing equivalents (NADH, FADH2) by the citric acid cycle. The transfer of high energy electrons from NADH and FADH2 to
complexes of the electron transport chain provides energy to pump hydrogen ions from the matrix into the
mitochondrial intermembrane space, thereby generating
the hydrogen ion gradient which subsequently drives
ATP synthesis through F0F1 complexes. The high NADH/
NAD ratio would be expected to inhibit protective
SIRT1. In addition, with an overabundance of energy
substrate, the hydrogen gradient increases to a critical
level, at which point the final electron transport to complex IV is impaired (where water is normally formed
from controlled electron transport to oxygen). The
backed-up electrons begin to be transported from coenzyme Q10 (ubiquinone) to oxygen directly, forming
superoxide anion. Paradoxically, a similar phenomenon
occurs with hypoxia. This formation of superoxide anion
can be blocked by dissipating the hydrogen ion gradient
(for example, by overexpression of uncoupling proteins),
by preventing transfer of electrons into the transport
chain, or by inhibiting transfer of pyruvate into the mitochondria in the first place (2048). In larger arteries,
uncontrolled endothelial uptake and oxidation of free
fatty acids, which are excessively abundant in type 2
diabetes, further exacerbates the energy surfeit with even
greater generation of superoxide anion and the presence
PAUL N. HOPKINS
by SIRT1 (2106) and decreased FOXO1-mediated induction of MnSOD (1236). Clearly, excess ROS production by
mitochondria and probably other sources such as various
NOX enzymes is a critical step in triggering endothelial
dysfunction in diabetes, but further discussion of its mechanisms is beyond the scope of this review.
2. Consequences of ROS overproduction: genetic
damage
While superoxide anion does not penetrate membranes, a
substantial amount of the excess superoxide formed on the
outer side of the mitochondrial inner membrane can be
transferred to the cytosol through the VDAC which traverses the outer mitochondrial membrane (2068). The large
increase in endothelial ROS production in diabetes is sufficient to overwhelm SOD antioxidant defenses and results in
both nuclear and mitochondrial DNA damage such as single-strand breaks.
1436
1437
PAUL N. HOPKINS
flammatory activation during efferocytosis can increase
both autoimmunity and atherosclerosis.
Several older studies illustrated synergism between immune
injury and hyperlipidemia. Thus immune injury to even
mildly hypercholesterolemic rabbits by horse serum aggravated atherosclerosis while antihistamines were protective
(1215). Increased antibodies to heat-dried cows milk and
boiled eggs were found in patients with CHD in an older
study (385). The display of HSP60 on endothelial cells in
association with activation of endothelial cells from a variety of initiating factors has been proposed to lead to autoimmune reactions, endothelial damage, and increased atherosclerosis, especially when coupled with hyperlipidemia
and other risk factors (1957).
G. Cigarette Smoking
Smoking has long been considered an endothelial cell stressor and activator. Cigarette smoke has 1017 free radicals
and many other ROS-producing chemicals per puff. Cigarette smoke extract can cause HDAC2 nitration, nitrosylation, and carbonylation, all leading to reduced HDAC2
activity which impairs the normal inhibition of inflammatory gene transcription by HDAC2. Greater NIK activation
is also seen (623).
Recently, the effects of exposing cultured endothelial cells
to cigarette smoke extract on expression of 35,000 genes
was reported (747). There was massive upregulation of
genes related to the unfolded protein response, thought to
be due to immense oxidative stress. Generation of free radicals was apparently mediated by metals as well as reactive
species in the extract. This was accompanied by mitochondrial dysfunction, upregulation of HSF1 and heat shock
proteins (including HSP60), and cell cycle arrest. These ob-
1438
servations are consistent with prior reports of the pro-oxidant effects of smoking leading to activation of endothelial
cells (58). The rapid reversibility of cardiovascular risk after
smoking cessation suggests this activation not only affects
early endothelial activation but also precipitating factors
such as clotting and stability of advanced plaques as well
(probably also through inflammatory effects) (946). Nevertheless, some evidence suggests persistent inflammatory effects can last for years after smoking cessation (1927). More
recently, the stimulating effects of nicotine on vaso vasorum
development seem to contribute to atherosclerosis as well
(992) (see below).
Table 8. Genes related to promotion of atherosclerosis by foam cell formation (72 genes tested)
Gene
Model
% A
Function, Comment
ACAT2 (SOAT2)
ACAT2/ apoE/
298%
SREBP-1
SREBP-1 / LDLR/
271%
APOC1
human APOC1 Tg in
apoE/
142%
APOC3
human APOC3 Tg in
LDLR/
1165%
CETP
1517%
1504%
Native lipoprotein affinity for the intimal ECM (extracellular matrix)lipoprotein and matrix properties
APOB
LEPR
Perlecan
perlecan/ apoE/
248%
Decorin
Decorin Tg apoE/
247%
265%
1290%
Continued
1439
APOBEC-1
PAUL N. HOPKINS
Table 8.Continued
Gene
Model
% A
Function, Comment
1125%
sPLA2 (group V)
238%
sPLA2 (group X)
SMS2
SMS2/ apoE/
252%
ASM (secreted)
ASM/ LDLR/
255%
157%
1107%
Macrophages from Tg group IIA sPLA2 overexpressing mice had 2.5fold greater rates of LDL oxidation, dependent on 12/15 LO
expression and increased foam cell formation. Urinary isoprostanes
were also increased (1793). Confirmatory of prior observations
(833, 1931).
Secreted phospholipase A2 (sPLA2) enzymes modify LDL and promote
foam cell formation. Transgenic over-expression of human GIII sPLA2
caused increased lysophosphatidyl choline in LDL and increased
atherosclerosis in animals fed an atherogenic diet (1540).
sPLA2 is thought to be largely secreted from activated macrophages.
Overexpression of the GV sPLA2 in this same study resulted in a
170% increase in lesion extent as well (186).
The main target of this isoform is phosphatidyl choline. Unexpectedly,
deficiency of the group X sPLA2 resulted in a marked increase in Th1
activity, IFN-, and macrophage necrosis while over-expression of
human PLA2G10 reduced plaque size, and enhanced Th2 and
macrophage IL-10 production (29).
Sphingomyelin synthase 2 (SMS2) KO mice had lower sphingomyelin
content in nonHDL lipoproteins, less aggregation of these
lipoproteins upon exposure to sphingomyelinase, and decreased
lipoproteins in the aortic wall (497). Similar results with
macrophage-only SMS2 deficiency in LDLR/ mice (1058).
Adenoviral delivery of SMS2 to apoE KO mice increased
atherosclerosis but also increased non-HDL-C and decreased HDL-C
(1914). Similarly, adenoviral expression of SMS1 and SMS2
increased sphingomyelin content and aggregation of APOB-containing
lipoproteins after sphingomyelinase treatment (434).
Acid sphingomyelinase (ASM [causes Niemann-Pick disease, type A
when deficient]) also has a secreted form besides the lysosomal
form. Secreted ASM cleaves sphingomyelin to ceramide, promotes
lipoprotein binding and retention, and greatly increases uptake and
foam cell formation for a number of lipoproteins (417).
Macrophage scavenger receptors, bridging proteins, extracellular peptides affecting lipoprotein uptake
LPL
233%
Fhx2
297%
ANGPTL3
ANGPTL3/ apoE/
269%
ANGPTL4
ANGPTL4/ apoE/
275%
Continued
1440
Table 8.Continued
Gene
Model
% A
LIPG/ apoE/
270%
APOE4 (human E4
isoform versus
E3)
152%
HSP27
235%
SR-AI/II/ apoE/
258%
237%
CD36
CD36/ apoE/
245%
DGAT1
DGAT1/ apoE/
224%
VLDLR
VLDLR/ LDLR/
LDLR
NS
1170%
NS
266%
227%
GIP
239%
Salusin-
250%
Salusin-
190%
Continued
1441
LIPG (EL)
Function, Comment
PAUL N. HOPKINS
Table 8.Continued
Gene
Model
% A
Function, Comment
Macrophage and VSMC intracellular signaling affecting scavenger receptor expression or lipoprotein uptake
LXR ( and )
LXR (NR1H3)
LXR (NR1H2)
LXR/ LDLR/
LXR/ LDLR/
1230%
173%
NS
241%
Arg1
241%
FXR
254%
1104%
PXR
PXR/ apoE/
240%
PPAR
125%
PPAR
262%
Bcl6
157%
Continued
1442
iVP16LXR LDLR/
Table 8.Continued
Gene
Model
% A
PGC-1
PGC-1/ apoE/
NS
AEBP1
1280%
Nrf2 (NFE2L2)
Nrf2/ apoE/
227%
ABCB4
181%
RIP2
137%
130%
The PPAR induces LXR and CD36 transcription while LXR induces
ABCA1 (287). Similar results in LDLR/ mice (97). PPAR also
promotes macrophage efferocytosis (286). PPAR signaling also
normally suppresses atherosclerosis-associated vascular calcification
by a newly recognized Wnt5a pathway in VSMC (1969).
No change in lesions despite more macrophages and increased ICAM-1
expression in plaques (1679). The PGC-1/ mice were lean and had
lower serum triglycerides but had much lower aortic PPAR and
PPAR expression.
AEBP1 (adipocyte enhancer-binding protein 1) is a transcriptional
repressor that downregulates LXR and PPAR activity in
macrophages (1116). Parallel studies including deletion of AEBP1
and studies in LDLR KO animals were confirmatory (165).
Nrf2, also known as NFE2L2 (nuclear factor erythroid type 2-like 2),
supports expression of CD36 and foam cell formation in
macrophages (1717). Chow-fed Nrf2 KO mice again showed less
CD36 in macrophages, fewer macrophages in plaques, but also
lower plasma lipid levels and reduced hepatic lipid production (106).
ABCB4 (also known as P glycoprotein 3) Foam cell formation was
greatly accelerated upon incubation with acetylated LDL due to
increased activity of scavenger receptors. Neither cholesterol nor
phospholipid efflux to HDL were affected (1403)
Appeared to promote macrophage spreading and macropinocytosis
(1012).
NPC1
NPC1/ apoE/
1230%
ACAT1
1119%
Elovl6
236%
ADRP (PLIN2)
225%
ATGL
243%
NCEH1
NCEH1/ apoE/
170%
CEH
macrophage-specific NCEH1
transgenic LDLR/
250%
250%
Continued
1443
PPAR
Function, Comment
PAUL N. HOPKINS
Table 8.Continued
Gene
Model
% A
HSL
120%
GPR109A
GPR109A/ LDLR/
133%
Function, Comment
APOA1
LDLR/
APOBEC1/
1110%
ROR
1504%
APOA2
human APOA1/AII Tg in
11367%
C57BL/6 fed atherogenic
diet
256%
ABCA1
158%
TRPV1
TRPV1/ ApoE/
1300%
ABCA5
150%
ABCA7
NS
Continued
1444
APOA1
Table 8.Continued
Gene
ABCG1
% A
Function, Comment
1141%
120%
ORP1L
1108%
SR-B1 (SCARB1)
SR-B1/ apoE/
15000%
SR-B1/ BMT
apoE/
186%
PDZK1
PDZK1/ apoE/
126%
GNMT
GNMT/ apoE/
APOM
APOM adenoviral
transduction in LDLR/
278%
HL (LIPC)
HL/ apoE/
273%
LCAT
LCAT/ LDLR/
LCAT/ apoE/
235%
251%
PLTP
248%
1500%
Reference numbers are given in parentheses. Not included are gene modifications which lead to changes in
lipoprotein plasma levels (see text).
1445
Model
PAUL N. HOPKINS
The notion of postprandial atherogenesis (2117, 2118) remains a strong possibility, particularly in light of the recent
report that much more APOB-48 was found in human
plaque than APOB-100 (1867). Furthermore, transgenic
overexpression of SREBP-1c in LDLR-null mice increased
VLDL production rate, VLDL size, and postprandial
plasma triglyceride levels, with greater overall exposure to
TGRL remnants. These animals suffered increased atherosclerosis. Conversely, double KO of SREBP-1 and LDLR
resulted in lower VLDL and less atherosclerosis compared
with LDLR KO controls (882). Other genes that may affect
lipoprotein size and composition due to effects on synthesis
or residence time are apo CI, apo CIII, and CETP with
proatherogenic effects demonstrated for all these except
CETP, which showed mixed effects (see TABLE 8).
An intriguing additional hypothetical mechanism related to
fat intake and chylomicrons is the possibility that these
intestinal lipoproteins carry sufficient LPS from intestinal
bacteria to promote inflammation in both adipose and the
artery wall (237, 1919). However, ApoE-deficient animals
had more atherosclerosis when raised in germ free conditions, at least when fed a low cholesterol diet, though there
was essentially no difference when the mice were fed a highfat diet (1683).
Somewhat related is the important recent observation that
the intestinal microflora decomposes dietary choline to the
gas trimethylamine which is efficiently absorbed and metabolized in the liver by FMO3 (flavin monooxygenase 3
and possibly other related enzymes) to a proinflammatory
agent, TMAO trimethylamine N-oxide (TMAO) which
1446
promotes inflammation and macrophage foam cell formation. ApoE KO mice fed a high-choline diet had three times
the atherosclerosis compared with either antibiotic-treated
mice or mice fed a regular diet. Atherosclerosis susceptibility also seemed to correlate with FMO3 activity in mice
with different genetic backgrounds. Data demonstrating a
strong positive association between CAD and TMAO in
human subjects was also presented (1924).
1447
On the basis of recent work, prelesional, intimal DCs include similar numbers of conventional and monocyte-derived DCs. Plasmacytoid DCs seem to enter into the intima
later as lesions progress (314). In fact, all studied DC subsets seem to proliferate or enter the plaque in increased
numbers as lesions advance. Conventional DCs proliferate
in response to the cytokine FLT3L which binds the DC
receptor FLT3 (FMS-related tyrosine kinase 3 where FMS
refers to feline McDonough sarcoma). Conventional DCs
do not respond to M-CSF. In contrast, monocyte-derived
DCs proliferate in response to M-CSF but not FLT3L. Thus
administration of FLT3L in test animals only promoted
proliferation of prelesional conventional DCs, and these
could be tracked by cell-specific markers. Importantly, KO
of FLT3 increased atherosclerosis along with a reduction of
Treg (314).
PAUL N. HOPKINS
protein cathelicidin antimicrobial peptide (CAMP) (981).
CAMP is the human protein; also known as LL37, it is
referred to in mice as cathelicidin-related antimicrobial protein (CRAMP). Neutrophil granules contain large amounts
of CAMP. CAMP also acts as a bactericidal protein on
human skin where it is expressed by keratinocytes. CAMP
and to a lesser extent other cationic proteins found in neutrophil granules bind the negatively charged phosphodiester backbone of DNA and protect it from rapid degradation by DNAses normally found in the extracellular fluid
(980). Nevertheless, specificity for nonmethylated CpG and
the C-G order persists even after CAMP binding (336).
When incubated with CAMP, even human leukocyte DNA
produced a substantial IFN- response in pDCs (980), but
this response was only about one-sixth as great as bacterial
DNA complexed with CAMP, since unmethylated CpG
motifs are relatively infrequent in mammals (336).
1448
Perlecan, the most abundant heparan sulfate glycosaminoglycan in the artery wall, also appears to promote LDL retention,
at least acutely. It may also suppress VSMC proliferation
(1805). Heterozygous deficiency of perlecan (1880) or excision of perlecan exon 3 (1805) reduced atherosclerosis significantly. In contrast, transgenic overexpression of decorin, a
smaller proteoglycan, actually decreased atherosclerosis in
1449
PAUL N. HOPKINS
nase is released by activated endothelial cells and other
lesional cells and has been implicated in LDL modification.
Importantly, sphingomyelinase produces ceramide in the
lipoprotein which promotes greater retention by proteoglycans and greatly promotes uptake and foam cell formation
for several lipoprotein types. Deficiency of acid sphingomyelinase in LDLR KO mice resulted in a 55% decrease in
atherosclerosis (417). Not only might sphingomyelinase be
important within the subendothelial space, but a fraction of
plasma LDL appears to carry substantial sphingomyelinase
activity. These LDL were also electronegative and carried
PAF-AH (also referred to as Lp-PLA2). Electronegative
LDL, when incubated in vitro, were shown to degrade their
own phospholipids, rendering them highly subject to aggregation and macrophage uptake. Such LDL could also activate inflammatory responses from various cell types (103,
1535).
seem unlikely to be an initial cause of macrophage activation (and perhaps not even a necessary one). Furthermore,
the LDL could be oxidized after it was taken up into the
macrophage and only become available for (re)uptake after
apoptosis or necrosis of the primary phagocyte. Unfortunately, standard mouse models of atherosclerosis are not of
much help in resolving the issue of myeloperoxidase involvement as mice express much lower levels of myeloperoxidase in their macrophages than seen in human lesions.
Furthermore, knockout of mouse myeloperoxidase does
not decrease atherosclerosis. However, overexpression of
human myeloperoxidase in the mouse macrophages does
increase atherosclerosis (1304). These considerations support the notion that myeloperoxidase is potentially
proatherogenic but redundant, as mouse atherosclerosis
can progress extensively in its absence.
5. Other chemical modification of LDL
1450
Carbamylation is a recently recognized form of LDL modification that may be particularly important in renal disease
and smoking. A small fraction of urea can spontaneously
and reversibly form highly reactive cyanate, which can then
bind to lysine residues in proteins forming homocitrulline.
Many proteins are altered in uremia, but carbamylated LDL
from uremic patients or prepared in vitro were found to
activate endothelial cells primarily by way of LOX-1 (70
72). Furthermore, carbamylated LDL binds macrophage
SR-AI receptors and causes foam cell formation (1926).
Importantly, thiocyanate, which is markedly increased in
the plasma of cigarette smokers, can be converted to cyanate by myeloperoxidase, also leading to carbamylation.
Protein homocitrulline levels were found strongly related to
cardiovascular risk with an odds ratio of 7 8 in the top
versus bottom quartile (1926). Indoxyl sulfate is another
uremic toxin that can modify LDL which then may cause
endothelial activation and inflammation (832).
In addition to traditionally cited scavenger receptors as mediators of modified lipoprotein uptake, recent evidence
points to non-scavenger receptor uptake as potentially a
quantitatively more important pathway. LDL treated with
group V sPLA2 is taken up by macropinocytosis which is
mediated by syndecan 4. This process is accelerated by macrophage activation and can lead to foam cell formation
(318). Unbound native LDL can also be taken up in substantial quantities by this process. In general, macropinocytosis is upregulated by pro-inflammatory signaling, such as
TLR4 and Syk activation which can be mediated by oxLDL,
acetylated LDL, mmLDL, LDL treated by 15-LO, PMA
activation of PKC and PKC, cholesteryl ester hydroperoxides, oxysterol-binding protein-related protein 2
(ORP2), and even viruses. In contrast, LXR agonists, which
are anti-inflammatory, block macrophage fluid-phase uptake of LDL. Unlike uptake of oxLDL through scavenger
receptors, macropinocytosis is blocked by inhibition of
PI3K (956). Thus foam cells can clearly be formed independent of scavenger receptors. In a sense then, scavenger
receptor uptake of modified LDL may be considered yet
another example of redundant pathways.
1451
PAUL N. HOPKINS
tein (PLTP), SCD-1, LPL, Mertk, IRF8, and SREBP-1c.
All of these genes (with the exception of SREBP-1c and
LPL) either facilitate cholesterol efflux or are otherwise
protective against atherosclerosis (see TABLE 8). IRF8 is a
transcription factor that increases expression of protective Arg1 (arginase 1) that helps form inflammation-resolving polyamines (see TABLE 8). Overexpression of
LXR increased net cholesterol transport from macrophages to the intestine, promoted intestinal non-bile
transintestinal cholesterol excretion (TICE), and reduced
liver fat while reducing atherosclerosis by 41% (1066).
Of note, LXR was not protective apparently because
when it is not bound by agonist it represses protective
ABCA1 and AGCG1, although it also decreases SREBP-1
expression (152).
1452
ABCB4 (also known as P glycoprotein 3) transports phospholipids into bile, thereby facilitating biliary cholesterol
secretion, but is also present on macrophages. Foam cell
formation in ABCB4-deficient macrophages was greatly accelerated upon incubation with acetylated LDL due to increased activity of scavenger receptors. Neither cholesterol
nor phospholipid efflux to HDL was affected (1403). These
findings raise the question of whether other cholesterol and
phosphlipid acceptors might be equally important as HDL
in macrophage cholesterol homeostasis.
Finally, despite less inflammatory signaling, RIP2 deficiency
increased atherosclerosis apparently because RIP2-deficient
macrophages took up more lipid and increased foam cell
formation due to constitutively active TLR4/Syk signaling
ified lipoproteins and enhanced activity of ACAT to esterify free cholesterol derived from engulfed lipoproteins.
Several classically proatherosclerotic cytokines including
IFN-, TNF-, and IL-1 promote foam cell formation in
this manner. They also contribute to enhanced foam cell
formation by downregulation of cholesterol efflux
(1174). To this list of well-known proinflammatory cytokines was recently added the TNF-family cytokine
TNF-like protein 1A (TL1A) which binds to its receptor
death receptor 3 (DR3). Activation of DR3 signaling in
macrophages results in increased uptake of acetylated
LDL (AcLDL) and oxLDL, presumably by the observed
upregulation of SR-A, CD36, SR-B1, and LPL. Simultaneously, cholesterol efflux was diminished together with
suppressed secretion of ApoE and downregulation of
ABCA1 and ABCG1 transporters (1173). This coordinated induction of scavenger pathways together with
downregulation of cholesterol efflux pathways has become apparent in a number of studies. Opposed to these
effects (with essentially mirror image activities) are cytokines which block foam cell formation and favor cholesterol efflux. These include TGF- and IL-10 (1174) to
some extent, but more specifically IL-33 binding to its
receptor ST2L on macrophages (1175). Elevation of macrophage cAMP may also be part of this response as glucagon-like peptide 1 (GLP-1) binding to its Gs-coupled
receptor GLP1R as well as gastric inhibitory peptide
(GIP) with its Gs GPCR both decreased foam cell formation and atherosclerosis during prolonged administration (1271). The peptide salusin- also decreased foam
cell formation and atherosclerosis while salusin- had
opposing effects (see TABLE 8).
Macrophage lipid droplets are surrounded by several proteins including perilipin 1, ADRP (also known as perilipin 2
or PLIN2), TIP47 (tail-interacting protein of 47 kDa), and
S312 (1397). Cholesteryl ester stored in these droplets can
be hydrolyzed by several enzymes with neutral cholesteryl
ester hydrolase activity. These enzymes include neutral cholesteryl ester hydrolase 1 (NCEH1), cholesteryl ester hydrolase (CEH), and hormone-sensitive lipase (HSL); interestingly, all three were found to be protective against atherosclerosis (see TABLE 8). Free cholesterol released by these
enzymes appears to be efficiently removed from macrophages through ABCA1 and AGCG1 transporters (1908).
The recently identified niacin receptor GPR109A is a Gi
type GPCR expressed on the surface of macrophages, other
white blood cells, and adipocytes but not endothelial cells.
One physiological function in adipose may be suppression
of free fatty acid mobilization and stimulation of adiponec-
1453
As macrophages take up modified LDL and TGRL remnants, cholesteryl ester in lysosomes is hydrolyzed by lysosomal acid lipase A (LIPA). Deficiency of LIPA promotes
atherosclerosis, possibly due to lysosomal damage and necrosis caused by excess lipid accumulation (448, 2124). The
action of normal LIPA releases free cholesterol which is
transported between cell organelles by NPC1/2. ApoE-deficient mice with NPC1 KO have worse atherosclerosis, and
interestingly, severe, spontaneous thrombosis. The cause of
the pro-thrombotic state was not clear (1940). Released free
cholesterol is esterified by acyl-CoA:cholesterol acyltransferase 1 (ACAT1) and stored in intracellular lipid droplets.
Cholesteryl ester (CE) droplets contribute to foam cell formation, and ultimately the size of plaques but appear to be
less problematic than excess free cholesterol which is cytotoxic. Accordingly, bone marrow deficiency of ACAT1 in
hyperlipidemic mice caused a marked increase in atherosclerosis (see TABLE 8), presumably due to accumulation of
free cholesterol. Nevertheless, deletion of the enzyme
Elovl6 (elongation of very long-chain fatty acids family
member 6) from macrophages in LDLR-null mice decreased
foam cell formation and atherosclerosis (1529). Elovl6 provides the preferred substrate to ACAT1 (mainly oleate) by
elongation of palmitate and palmitoleic acids to stearate
and oleate, respectively. Adipose differentiation-related
protein (ADRP), another protein required for lipid droplet
formation, also proved to be mildly proatherogenic (see
TABLE 8). In contrast, triglyceride droplets do appear to be
benign as bone marrow specific deletion of adipose triglyceride lipase (ATGL) led to massive macrophage triglyceride
accumulation but decreased macrophage activation and
less atherosclerosis (978).
PAUL N. HOPKINS
increased atherosclerosis (see TABLE 8). These signaling effects of HDL are reviewed further in section XE. Macrophages also constitutively secrete ApoE which can act together with phospholipid as a free-cholesterol acceptor,
forming disks much like nascent HDL (2082). ApoE also
acts in efferocytosis as reviewed below.
ABCA1 and ABCG1 (and probably ABCA5) show compensation for one anothers deficiency while macrophage-specific knockout of both ABCA1 and ABCG1 clearly increased atherosclerosis (2054). These insights help explain
variable results seen with knockout of only ABCA1 or
ABCG1 (22). Hepatic ABCA1 expression may be most important in atherogenesis through preservation of newly secreted APOA1. This may be followed by rapid minimal
lipidation of pre- HDL before leaving the liver, perhaps in
the space of Disse (220, 1249). Without such lipidation of
nascent particles, lipid-poor APOA1 is rapidly eliminated
by the kidneys and HDL levels are extremely low, as in
Tangier disease patients (who lack ABCA1).
1454
Another interesting example of potential complexities in reverse cholesterol transport is the role of PLTP (364, 1350).
PLTP is found in plasma associated with HDL and mediates
complex HDL remodeling. Excess surface phospholipid and
unesterified cholesterol that remain on TGRL remnants after
delipidation (as well as some from LDL) are transferred by
PLTP to HDL. In addition, PLTP causes fusion of smaller
HDL3, yielding both larger HDL2 and smaller spherical HDL,
pre- HDL, or free APOA1. ApoM may also interact with
PLTP in regeneration of pre- particles. High plasma or systemic PLTP activity appears generally atherogenic, perhaps
because increased generation of pre- HDL or free APOA1 in
plasma would cause greater renal loss of APOA1, resulting in
low HDL and decreased overall APOA1 availability. Wholebody PLTP deficiency in two models of hyperlipidemic mice
led to reduced production of VLDL and LDL and marked
reduction of serum lipids, together with reduced atherosclerosis (852). Lower systemic PLTP activity predicted by a genetic
score in humans was also associated with reduced coronary
risk (975). In contrast, PLTP released locally by macrophages
appears protective since PLTP physically interacts with
ABCA1 and pre- HDL to promote cholesterol efflux (1350).
Furthermore, PLTP interaction with ABCA1, even when the
lipid transfer activity is eliminated by a PLTP M159E mutation, stimulates JAK2 and anti-inflammatory STAT3 signaling
in macrophages (1891). Thus whole-body KO of PLTP in
LDLR/ mice was atheroprotective (presumably by preserving APOA1 generally as discussed above). However, transplant of bone marrow from PLTP/ LDLR/ mice into
PLTP/ LDLR/ mice further reduced atherosclerosis,
demonstrating the protective role for normal local expression
of PLTP by macrophages (1839). This model resolves a number of seemingly discrepant observations regarding PLTP
(364, 1840, 1856). Interestingly, PLTP also binds and inactivates bacterial LPS, and PLTP-deficient mice tolerate endotoxemia poorly. The anti-inflammatory effect of PLTP may also
be important in regard to multiple sclerosis, as lower PLTP
activity was associated with more active disease (1891).
X. PROGRESSION OF ATHEROSCLEROSIS
A. Features of Unstable Plaques: Challenges
With Animal Models
As noted in the introduction, this phase of atherogenesis has
to do with growth of complex plaques, plaque rupture, and
subsequent thrombosis with precipitation of acute ischemic
Surprisingly, HL KO decreased atherosclerosis in ApoEdeficient mice (1204). HL is most active toward intermediate-sized lipoproteins, hydrolyzing both TG and phospholipid in larger HDL and in IDL (intermediate density lipoproteins). Even though HL/ ApoE/ mice had much
higher total cholesterol and -VLDL, they also had higher
HDL-C. Furthermore, HDL from ApoE/ are severely impaired in promoting efflux from cultured cells and HL deficiency partially restored this capacity.
1455
PAUL N. HOPKINS
Table 9. Genes related to further plaque growth, progression of advanced atherosclerosis (54 genes tested)
Gene
ATM
p53
Rb
Kip1 (CDKN1B)
CDKN2B
MTAP
PDGF-B
LRP1
% A
Function, Comment
Continued
1456
CDKN2A
Model
Table 9.Continued
Gene
Model
% A
LRP5
LRP5/ apoE/
LRP6
PKG
231%
TRAIL
TRAIL / apoE/
126%
TAGLN
VSMC-specific CreLox
TAGLN/ in apoE/
128%
DDR1
DDR1/ LDLR/
260%
CLU
CLU/ apoE/
252%
232%
1350%
11,000%
245%
NLRP3
269%
ASC
270%
CASP1
CASP1/ apoE/
240%
Continued
1457
RAP
Function, Comment
PAUL N. HOPKINS
Table 9.Continued
Gene
% A
Function, Comment
AIF-1
VSMC-specific Tg in C57BL/6
fed Paigen diet
1151%
iNOS
247%
S100A9
S100A9/ apoE/
225%
AIF-1 (allograft inflammatory factor 1) is a calcium- and actinbinding, cytoplasmic scaffold protein that may promote complex
formation with multiple signaling proteins including G proteins
such as Rac1 (1652). Increased expression of AIF-1 also leads to
greater phosphorylation of Akt, Erk1/2, and p38. Endothelial AIF1 promotes angiogenesis. Both macrophage and VSMC
expression of AIF-1 is increased in human plaques and by
incubation with oxLDL. AIF-1 promotes proliferation, migration,
and phagocytic activity in these cells. Whole body AIF-1
transgenic, apoE-null mice had a 20% increase in atherosclerosis
(1216).
Also known as NOS2. Shown are results in males.
Proatherosclerotic effects of iNOS appear to be primarily related
to leukocyte expression. iNOS was shown to produce both NO and
superoxide in the lesions (1430). iNOS deletion in normolipidemic
mice appears to cause hyperlipidemia and increase
atherosclerosis (818).
S100A9 (also known as myeloid-related protein 14) stabilizes
S100A8; S100A9 KO leads to undetectable S100A8 as well.
SA100A8/9/12 are calgranulins which can act as TLR4 and
RAGE agonists. S100A9 KO macrophages had reduced cytokine
production in response to LPS (356) but VSMC (or endothelial)
production may be relevant as well (see S100A12 below). No
effect seen with S100A9/ BMT into LDLR/ (88)
S100A12
140%
Leptin
ob/ob apoE/
244%
FABP4
FABP4
248%
FABP5
236%
CHOP
CHOP/ apoE/
235%
XBP1
Bax
1837%
149%
Continued
1458
Model
Table 9.Continued
Gene
Model
Bcl-2
macrophage-specific Bcl-2
deletion (Cre-Lox) in
apoE/ mice
AIM
AIM/ LDLR/
% A
Function, Comment
NS
273%
Macrophage efferocytosisgeneral
HMGB1 neutralizing antibody
to apoE/
255%
Fas
Fas/ apoE/
170%
FasL
FasL/ apoE/
Mertk
Gas6
Gas6/ apoE/
Mfge8
171%
TG2
122%
APOE
TLR3
TLR3/ apoE/
1207%
166%
NS
1892%
140%
Continued
1459
HMGB1
PAUL N. HOPKINS
Table 9.Continued
Gene
Model
% A
Function, Comment
PTX3
PTX3
CRP
ADIPOQ
human ADIPOQ-adenoviral
overexpression in apoE/
mice
C1qa/ LDLR/
C1qa
apoE
150%
NS
230%
1238%
C3
C3/ LDLR/
C5
C5def/def apoE/
C6
C6def/def cholesterol-fed
rabbits
281%
C3aR1
C3aR1/ apoE/
234%
DAF
DAF/ LDLR/
CD59
CD59/ apoE/
130%
265%
1100%
NS
1159%
149%
1460
transplantation of the vessel may have somehow accelerated macrophage egress in that model, a condition
thought to be much less relevant to the usual human
situation. Thus, rather than coaxing faster macrophage
egress from plaques, lipid normalization leads to marked
reduction in macrophage recruitment as the main means
to effect plaque regression. These findings also suggest
that slowing monocyte/macrophage entry into lesions
would be an important means to facilitate regression.
As alluded to earlier in this review, apoptosis of macrophages early in plaque development seemed to minimize
CFB
MBL
Table 10. Plaque destabilization, thrombosis, and precipitation of acute events (46 genes tested)
Gene
Model
% A
Function, Comment
232%
MMP2
MMP2/ apoE/
249%
MMP3
MMP3/ apoE/
1343%
MMP7
MMP7/ apoE/
27%
MMP9
MMP9/ apoE/
1122%
MMP12
MMP12/ apoE/
252%
MMP13
MMP13/ apoE/
NS
MMP14
NS
TIMP1
TIMP1/0 apoE/
NS
TIMP2
260%
TIMP3
macrophage-specific TIMP3 Tg in
LDLR/
248%
CatK
CatL
CatK/ apoE/
CatL/ LDLR/
242%
277%
CatS (CTSS)
CatS/ LDLR/
230%
Cst3
Cst3/ apoE/
NS
Continued
1461
MMP1
PAUL N. HOPKINS
Table 10.Continued
Gene
Model
% A
Function, Comment
vWF
LDLR
240%
ADAMTS13
ADAMTS13/apoE/
160%
TF
TF/ apoE/
TF/ BMT into LDLR/
TFPI
TFPI/ apoE/
129%
Factor II
F2/ apoE/
244%
Factor V
157%
Factor VIII
PAR1
F8/ apoE/
PAR1/
240%
PAR2
PAR2/
PAR3
PAR3/
PAR4
TM
PAR4/ apoE/
TMPro/Pro apoE/
Protein C
PC/
Fibrinogen
Fbg/ apoE/
NS
Plasminogen
Plg/ apoE/
1620%
NS
NS
167%
Continued
1462
vWF
Table 10.Continued
Gene
Model
Function, Comment
tPA (PLAT)
tPA/ apoE3-Leiden
270%
uPA (PLAU)
uPA/ apoE3-Leiden
NS
2-AP
2-AP/
HCF2
HCF2/ apoE/
FBN1
HPS1
172%
HPS3
265%
HPS4
HPS5
253%
245%
HPS6
259%
164%
1179%
NS
Continued
1463
% A
PAUL N. HOPKINS
Table 10.Continued
Gene
Model
Function, Comment
PlGF
PlGF/ apoE/
241%
VEGFR1
250%
G-CSF
1100%
nAChR1
242%
PAI-1
113%
273%
Hp
NS
HAMP
NS
Col18a1
Col18a1/ apoE/
167%
Endostatin
Endostatin subcutaneous
injections into apoE/
285%
Angiostatin
Angiostatin subcutaneous
injections into apoE/
231%
1464
% A
In advanced lesions, both macrophages and VSMC proliferate. A series of newer studies have demonstrated interesting biological features of pathways controling cell
proliferation, but their direct clinical relevance to human
atherosclerosis remains somewhat uncertain. In general,
genetic manipulations resulting in greater macrophage
and VSMC proliferation typically increase plaque size,
but with regard to VSMC, such plaques may be more
stable. Also, proliferation is often accompanied by protection from apoptotic signals. In early atherosclerosis,
greater apoptosis generally leads to smaller plaques.
However, in advanced disease, increased apoptosis can
overwhelm efferocytosis pathways, promote secondary
necrosis, and lead to increased inflammation with unstable plaques. Pathways related to control of proliferation
are reviewed in the following paragraphs.
Cell replication is initiated by extracellular growth factors that signal to ultimately stimulate the transcription
of cyclin D together with a reduction of specific cyclindependent kinase (CDK) inhibitors (136). In general,
CDKs bind cyclins and then phosphorylate other proteins
to promote specific steps through the cell cycle. Cyclin D
binds CDK4/6 and remains high throughout the cell cycle
while discrete waves of other cyclins peak at specific
phases of the cycle. Presiding over this system is the transcription factor p53, whose elevation leads to increased
production of CDK inhibitors, other proteins that inhibit
the cell cycle, DNA repair enzymes, and proapoptotic
proteins. In its role, p53 has been dubbed the guardian
of the genome.
To replicate, the cell must pass through a series of checkpoints that are controlled by a series of serine/threonine
kinases known as checkpoint kinases. Ataxia telangiectasia
mutated (ATM) is a major checkpoint kinase that is central
to a network of proteins that control cell replication, energy
production, protein synthesis, autophagy, and apoptosis
(126, 1461). The kinase activity of ATM is controlled by
ROS and surveillance proteins which bind double-stranded
breaks in DNA and recruit and activate ATM. ATM then
phosphorylates and activates ChK1/2 (checkpoint kinases 1
and 2). These phosphorylate the transcription factor p53,
thereby inhibiting p53 ubiquitination and degradation.
ATM also directly phosphorylates and protects p53 and
phosphorylates LKB1 (liver kinase B1). Besides AMP or
ADP, LKB1 is the main activator of AMPK. Active phosphorylated LKB1 binds to the AMPK regulatory subunit,
rendering the AMPK subunit susceptible to other activating phosphorylation (45).
Active ATM and increased p53 generally limit cell growth
and mitogenesis while their reduction will promote proliferation. Accordingly, reduction of ATM promoted athero-
1465
A newly recognized function of ApoE relates to proliferation and mobilization of monocytes and neutrophils.
Several models of hyperlipidemia, including cholesterol
and saturated fat-fed pigs, rabbits, and especially ApoEdeficient mice are accompanied by pronounced monocytosis and neutrophilia. Monocytosis was also pronounced in mice with combined deficiency of the ABCA1
and ABCG1 receptors which are involved in export of
free cholesterol from various cell types. The monocytosis
in these mice was related to cell membrane accumulation
of free cholesterol, increased lipid raft formation, and
hypersensitivity to the proliferative effects of the GMCSF (also known as IL-3) receptor (CSF2RB) in hematopoietic stem cells, with CSF2RB ligation leading to ERK
and STAT5 activation (1258). Further studies in ApoE
KO mice fed a Western-type diet showed that the threeto fourfold increase in blood neutrophils and monocytes
was due to impaired cholesterol efflux from hematopoietic stem cells. Quiescence of these cells was normally
maintained by cholesterol efflux and controlled plasma
membrane cholesterol content through LXR-dependent
production of ApoE, ABCA1, and ABCG1. ApoE is normally released from the cell and bound to cell surface
heparan proteoglycans where it can immediately interact
with ABCA1 and ABCG1 transporters to promote cholesterol efflux. Treatment with heparinase impared this
pathway and promoted proliferation through increased
GM-CSF receptor activity leading to intracellular STAT5
and ERK activation. In ApoE-deficient mice, high doses
of a recombinant HDL having high cholesterol mobilization capacity (with APOA1 and phospholipid but no cholesterol) could also suppress proliferation. Interestingly,
transplant of ApoE-deficient bone marrow cells into
LDLR KO mice led to greater hematopoeitic cell proliferation, leukocytosis, and proportionately increased deposition of monocytes into atherosclerotic lesions despite
similar cholesterol levels (1258). Thus the monocytosis
of ApoE-deficient mice may directly contribute to the
rapid development of atherosclerosis in these animals.
These studies may provide additional insights into previously noted increased risk of CAD associated with higher
leukocyte and especially monocyte counts noted in epidemiologic studies (340, 378).
PAUL N. HOPKINS
sclerosis with increased macrophage proliferation (and
JNK activation). In contrast, chloroquin, which activates
ATM, reduced atherosclerosis but only in ATM/
ApoE/ mice (1561), an effect that also required p53
(1465).
Prior to initiation of G1 (growth phase 1), hypophosphorylated Rb (retinoblastoma gene) sequesters the transcription dimer E2F/DP which is needed for transcription
of genes required in the G1 phase of cell replication,
thereby blocking growth and proliferation. At the beginning of the cell cycle, CDK4/6:cyclin D and CDK2:cyclin
E complexes hyperphosphorylate Rb which then releases
E2F/DP, allowing the cell to enter G1 (570). Macrophage
Rb KO resulted in increased lesion size with more macrophages (163).
1466
pression and the gene targets are likely cell- and conditionspecific. Among the targets of these repressive complexes
are the CDKN2A/B loci (349).
VSMC are among the first cells found in the intima in locations destined to become atherosclerotic plaques, presumably in response to PDGF-BB produced initially by activated endothelial cells in regions of disturbed flow. Beyond
being a source for extracellular matrix and collagen, in an
activated, secretory state VSMC can elaborate chemokines
including MCP-1 and CX3CL1 (fractalkine), as well as adhesion molecules (VCAM-1, ICAM-1) which attract and
then capture macrophages within the intima. VSMC can
synthesize and secrete pro-inflammatory cytokines including IFN-, IL-1, IL-6, and IL-18. They can alter the extracellular matrix to further promote inflammation by producing fibronectin. They express various scavenger receptors
and take up modified lipoproteins to become foam cells
which can cause further activation. Thus VSMC can take on
characteristics of endothelial cells in recruitment of leukocytes, some of the functions of T-cells by producing cytokines to amplify the inflammatory response, and the properties of macrophages by conversion to foam cells and elaboration of similar cytokines. More recently, VSMC have
also been observed to recapitulate additional functions generally attributed to macrophages, such as efficient phagocytosis of apoptotic cells with secretion of inflammation-resolving (although pro-fibrotic) cytokines including PDGF
and TGF- (438).
1467
VSMC are normally potent, effective phagocytes of apoptotic VSMC. This capacity of VSMC for avid efferocytosis has
been previously unappreciated. Studies in mice engineered
with inducible, VSMC-specific diptheria toxin receptor
provide new insights into an important role for VSMC in
advanced plaques as active phagocytes. Low-level diptheria
toxin administration to these mice resulted in apoptosis in
1% of VSMC, a rate similar to human plaques. In normolipidemic mice, this degree of apoptosis resulted in no
obvious inflammation. However, in the face of hyperlipidemia, normal phagocytosis of apoptotic VSMC by other
VSMC was severely impaired (332). Failed phagocytosis of
apoptotic VSMC by surrounding normal VSMC resulted in
secondary necrosis and markedly increased production of
inflammatory cytokines by necrotic VSMC (including IL1, IL-1), followed by greater IL-6 and MCP-1 production
by surviving VSMC (332). A twofold increase in atherosclerosis with impaired outward remodeling and increased stenosis ensued (331).
PAUL N. HOPKINS
biguous (63). PDGF actually consists of homo- or heterodimers of four different PDGF gene products (PDGF-A,
-B, -C, or -D) while receptors are homo- or heterodimers of
PDGFR and PDGFR. Frank deficiency of PDGF or
PDGFR genes is generally embryo lethal as these growth
factors are intimately involved in various stages of organogenesis. PDGFR signaling is complex, with multiple adaptors and effectors. PDGFR ligation is generally pro-proliferative and antiapoptotic (in part through PKB and ERK
activation) as well as anti-inflammatory and profibrotic. In
one study, transplant of PDGF-B-null fetal liver cells into
irradiated ApoE/ mice resulted only in a delay of VSMC
accumulation and fibrous cap formation without a change
in lesion volume (945). In addition, macrophages appeared
more inflammed or activated in the PDGF-B-null chimeric
mice.
1468
The LRP1 receptor has major effects on VSMCs. Interestingly, LRP1 can mediate uptake of aggregated LDL and
subsequent cholesterol accumulation in VSMC. Aggregated
LDL seems to stabilize LRP1 and increase its expression by
decreasing ubiquitin-mediated degration (240). VSMC-specific deficiency of LRP1 in LDLR-null mice resulted in thickening of the media, dilation of the aorta, aneurysm formation, and accelerated atherosclerosis (191). Importantly,
feeding VSMC-specific LRP1-deficient mice a high-fat,
high-cholesterol diet led to dramatically accelerated atherosclerosis with massive foam cell formation and occlusion of
mesenteric vessels and even aortic occlusion followed by
death of the animals within 2 mo. LRP1 can bind at least 40
ligands and modulates signaling of other receptors. In liver,
together with LDLR, LRP1 mediates endocytosis of TGRL
into hepatocytes (192). In VSMC, LRP1 physically interacts
with and restrains the activity of both the PDGFR and the
TGF- receptors. Absence of VSMC LRP1 led to increased
cell-surface PDGFR expression, PDGF-BB signaling, and
VSCM proliferation.
A newly recognized contribution of medial VSMC to inflammation in atherosclerotic lesions has recently been reported. VSMC stimulated simultaneously through TNFR1
and LTR ligation can direct lymphorganogenesis in the
adventitia, resulting in a collection of activated immune
cells which may contribute to the inflammation and progression of the nearby plaque (1077).
Finally, an additional, surprising feature of VSMC behavior
in advanced plaques should be mentioned. Culprit plaques
causing acute coronary syndromes in humans have been
consistently shown to rupture just proximal (upstream) to
the point of maximum stenosis. This is the area of most
rapid flow (560). This is true even though the area of slow
flow just downstream of the stenosis is where endothelium
is activated and foam cells accumulate. It appears that while
high shear stress promotes endothelial integrity and higher
NO production in the upstream area, the higher shear may
simultaneously promote plasmin production as well. Furthermore, NO appears to inhibit underlying VSMC proliferation and collagen synthesis as well as promote VSMC
apoptosis. Why this would be the case is not clear since
PKG, activated by NO, actually promotes VSMC proliferation and PKG deficiency led to smaller lesions with fewer
VSMC in hyperlipidemic mice (1972). Perhaps prolonged
1469
PAUL N. HOPKINS
which binds MyD88 with downstream signaling to activate
NF-B which increases production of pro-IL-1 and various chemokines. TLR4 further binds the adapter TRIF with
increased production of type I interferons, IFN-/ (1684).
CD36 can also form a complex with TLR2 with further
activation of NF-B. Src family kinases can also mediate
signaling through a PLC-DAG, IP3-calcium release-PKC
activation pathway, further promoting PYK2 activation.
Src family kinases simultaneously activate the GEF Vav1 to
exchange GTP for GDP on Rac1. Vav1 and Vav3 couple
with CD36 and are required for foam cell formation upon
incubation with oxLDL (1449). Active PKC and Rac1 promote assembly and activation of NOX2 to enhance ROS
production (1210). These pathways provide greater understanding of the marked reduction in atherosclerosis after
CD36 KO or pharmacological CD36 inhibition in mice
(716).
1470
Increased PC synthesis by macrophages in response to excess FC has come to be recognized as part of the unfolded
protein response (UPR). Some elements of the UPR are depicted in FIGURES 16 AND 17. Excellent reviews of the UPR
in relation to cardiovascular disease are available (1213,
1505). Briefly, in unstressed cells, three key transmembrane
sensors (IRE1, PERK, and ATF6) extend through the ER
oxLDL
CD36
SR-A
LOX-1
CD68
Src
Rac1
Vav1/3
FC
NCEH1
ACAT1
PL
C
HSL
Macrophage
CE
IB
FC
CD36
LXR
TLR2/4/6
AEBP1
FAB4/5
PPAR
PKC
mmLDL
Lp(a)
Necrotic cell
Fyn
Inflammatory signaling, cytokines
NF-B
88
Syk
yD
AP1
ERAD, PC, chaperones
XBP1
NF-B
ATF4/6
PYK2
CHOP
JNK
CaMKII
Bcl2
Bim
eIF2
TAK1
ASK1
Bip
PERK
ATF6
Bip
Bip
Ca2+
Tiam1
Bak
Bax
Rac1
ac1
ERO1
ROS
SERCA
IP3R1
Nox2
IL-1
Inflammatory signals
Prior FC or oxLDL loading
Uric acid
DNA
HMGB1
Prior NLRP3 activation
ATP (large release)
Prior TLR activation
Pathogen derived factors
(LPS, flagellin, etc.)
Saturated fatty acids
Nrf2
NLRP3
Inflammasome
Cholesterol
crystals
FIGURE 16. Proinflammatory lipid-mediated signaling and efferocytosis in macrophages. Nrf2 is in some way
permissive for activation of the NLRP3 inflammasome and IL-1 production. ACAT1, acyl-CoA:cholesterol
acyltransferase 1, also known as SOAT1 for sterol O-acyltransferase 1; CE, cholesterol ester; FC, free
cholesterol; nCEH, neutral cholesteryl ester hydrolase; PL, phospholipid; PS, phosphatidylserine. Src refers to
Src or other Src-family protein tyrosine kinases and may include Src, Lyn, Fyn, Lck, Yes, Fgr, Hck, Blk, or Csk-1.
1471
U U U U
TRAF2 IRE1
mito Ca,
JNK, Fas,
STAT1
PAUL N. HOPKINS
appear to deliver both FC and saturated fat to the ER,
promoting ER stress. Additionally, these FABP, in some
way, block desaturation of palmitate by SCD-1 and
block the protective activation of LXR, possibly by direct
physical interaction with and inhibition of PPAR or by
binding and sequestering away natural ligands of PPAR.
KO of FABP4 leads to greater availability of these ligands, greater stimulation of PPAR, and suppression of
atherosclerosis (1123). FABP5 deletion was also protective (95, 484, 800) (TABLE 9).
1472
release from the ER. Besides stimulating the NLRP3 inflammasome as noted above, ER calcium escape can activate
CaMKII leading to serine phosphorylation of STAT1, and
activation of PYK2. ROS production by mitochondria as
well as Rac1 and NOX2 with general promotion of proinflammatory and apoptotic pathways follows (1729, 1731).
After being summoned, the phagocyte must encounter appropriate eat me signals displayed by the apoptotic cell
(such as phosphatidylserine and various opsonin-like proteins) which match a series of receptors on the phagocyte,
thus forming a synapse between the phagocyte and the target cell. Certain inhibitory dont eat me signals, generally
displayed on healthy cells, can strongly block efferocytosis.
In advanced plaques there appears to be an over-abundance
of efferocytosis signals with many awaiting apoptotic cells,
suggesting saturation of efferocytosis mechanisms (1791,
1863). Several efferocytosis promoting ligands and receptors, together with subsequent intracellular signaling, are
discussed below and are reviewed in TABLE 9 and depicted
in FIGURE 17.
Virtually all of the defects of efferocytosis listed in TABLE 9
(as well as those related to Treg function) promote autoimmune disorders as well as atherosclerosis (473, 2120). Interestingly, while Fas (fragment, apoptosis stimulating) is
often referred to as the death receptor, Fas, and its ligand
1473
FasL, are also among the receptors and ligands that control
efferocytosis. Mice that lack functional Fas or FasL have
impaired ability to clear apoptotic debris and display features of autoimmune disease. When crossed with ApoEdeficient mice, these mice have increased atherosclerosis
(73, 517).
PAUL N. HOPKINS
pre- HDL
FC & PL
ABCA1/5
LXA4,RvD/E,
PD1, Ma
LIPA
15-LO
5-LO
FC, oxysterols
EETs
FC
CYP2C/2J
FcR
LXA4
RvD/E
15d-PGJ2, etc.
S1PR1
Desmosterol
CRP
LRP1
+sumo
PI3K
GAS6
PS
Tim1/4
Inflammatory genes
NF-B
Mfge8
Bcl6 PPAR
PS
LPC, LPS
V3
Chaperones, PC
Rheb
mTOR
S6K1
eIF2
S6
PGE2
IRE1
PERK
ATF6
Bip
Bip
Bip
G2A
Efferocytosis
Anti-inflammatory signals
Adenosine, LPC, S1P
PS (phosphatidyl serine)
Lipoxin, resolvin, protectin,
maresin precursors
n-3 fatty acids (EPA, DHA)
RNA (through TLR3)
Lysophosphatidylserine (LPS)
Hemoglobin/haptoglobin 1-1
Compensated ER stress
FIGURE 17. Anti-inflammatory lipid-mediated signaling and efferocytosis in macrophages. Nuclear receptor corepressor 1 (NCoR) functions as an inhibitory deacetylase complex. An important anti-inflammatory function of LXR and PPAR is to block the removal of NCoR from the promoters of proinflammatory
genes. LPC binding to GPCR G2A may promote efferocytosis. Not shown is extensive anti-inflammatory
signaling that occurs with APOA1 binding to ABCA1. PC, phosphatidylserine; FFA, free fatty acids; LPC,
lysophosphatidylcholine; LXA4, lipoxin A4; Ma, maresins; PD1, protectin 1; RvD/E, resolvins of the D and
E series; S1P, sphingosine-1-phosphate.
1474
TSC1 TSC2
Apoptotic
cell
PS
NCor
AP1
Mertk
ABCA1, ABCG1,
LXR
HDL
C1q, MBL,
adiponectin
ApoE
CD36, ABCG1,
PPAR
LXR, CYP27
PKB
PTX3
cAMP,
PKA,
PKB
RXR
Active
NFB, AP1
TGF,
IL-10,
PGE3
ABCG1
CYP27
FFA, FC
ALX
ChemR23
HDL3/2
PL
While generally depicted as a means of promoting cholesterol efflux from macrophages, ApoE secreted by either the
phagocytosing macrophage or apoptotic macrophages promotes efferocytosis and downregulates inflammatory responses. These effects are mediated by binding to LDL receptor-like protein 1 (LRP1) which, in this setting, activates
cAMP, PKA, and PKB signaling followed by greatly reduced responsiveness to TNF- (2016).
An influx of lipids from ingested lipoproteins and efferocytosis may also promote synthesis of lipoxins (such as
LXA4), resolvins, protectins, and maresins with marked
downregulation of inflammatory pathways (1726). These
lipid mediators are analagous to PGI2 and other anti-inflammatory prostaglandins in that they mediate their effects
through GPCR (951). Furthermore, PGE2 and PGD2, produced by COX-2 in macrophages, cause increased expression of 15-LO, leading to a switch from production of proinflammatory prostaglandins and leukotrienes toward production of anti-infammatory lipoxins, resolvins, protectins,
and maresins. This is yet another means whereby selective
COX-2 inhibition may be detrimental. Indeed, NSAIDs
generally seem to slow resolution by blocking prostaglandins that signal the switch and possibly precursors for some
of these active lipid mediators of resolution (1590). Precursors to these lipids may be synthesized by endothelial cells,
platelets, or PMN. Macrophages, which arrive later in acute
inflammatory reactions, may use these precursors to synthesize the resolving lipid mediators. Alternatively, PMNs may
also produce lipoxins and resolvins.
1475
PAUL N. HOPKINS
Emerging understanding of the complement pathway in atherosclerosis points to a protective role for early components
and related molecules which bind or opsonize apoptotic cells
(besides invading bacteria) and promote efferocytosis. In contrast, assembly of later components that promote cell damage
are atherogenic (1663). These findings are summarized in TABLE 9. Only a few orienting comments will be given here.
Pentraxin 3, long pentraxin (PTX3) and C-reactive protein
(CRP) are both acute phase reactants of the pentraxin family which act as part of the innate immune response to coat
or opsonize invaders such as certain bacteria. They are capable of subsequently triggering the classical complement
pathway when the opsonized invaders encounter the first
component of the complement pathway, C1q. Thus, in addition to the Fc portion of antigen-bound antibodies, C1qa
also recognizes and binds PTX3, CRP, the phosphatidylserine on apoptotic cells, and the membrane of certain bacterial cells, and can thereby activate the classical complement
cascade.
1476
Besides activating the classical complement pathway, invaders coated with IgG or IgM antibodies can be phagocytosed by interacting with Fc receptors, or when C1q is
bound by the calreticulin/CD91 complex on phagocytes
(1431, 1663). Apoptotic cells opsonize by C3b and C4b can
be phagocytosed through recognition by other macrophage
receptors (1663). C1q KO was atherogenic (TABLE 9). Importantly, both PTX3 and CRP can also bind apoptotic
cells and promote efferocytosis. CRP in particular recognizes oxidized phospholipids. PTX3 appears to be anti-atherosclerotic while most CRP manipulations resulted in little
or no change in atherosclerosis. The few significant effects
that were seen for CRP were consistently protective (see
TABLE 9).
C1q is also activated by enzymatically modified LDL containing free fatty acids together with phospholipid (151).
C1q enhanced the uptake of oxLDL into macrophages with
subsequent activation of the cells including increased
CD80, PECAM1, and MCP-1 release but also induction of
ABCA1 and ABCG1 with enhanced cholesterol efflux to
HDL (543). Remarkably, adiponectin has an analogous
structure to C1q, also opsonizes apoptotic cells and promotes their efferocytosis, binding to the same calreticulin/
CD91 receptor complex as C1q (1746). It should be noted,
however, that the main action of adiponectin appears to be
activation of ceramidase through binding its receptors AdipoR1 and AdipoR2, thereby causing the deacetylation of
ceramide with downstream formation of the anti-apoptotic
S1P (778). Overexpression of adiponectin decreased atherosclerosis (1339).
The alternate complement pathway is triggered when C3b,
continually produced at a low level, is activated when it
covalently binds apoptotic cells, enzymatically modified
LDL, cholesterol crystals or other atherosclerotic debris,
and other foreign surfaces (1663). Active C3b then binds
CFB (complement factor B) which is cleaved by factor D to
produce the alternative pathway C3 convertase consisting
of Bb and C3b. When BbC3b is joined by another C3b, it
becomes a C5 convertase. The alternative pathway thus
serves to amplify the classical pathway. Interestingly, no
effect of CFB KO was seen when mice were fed a low-fat
diet. During a high-fat diet, LDL-C was much lower in CFB
KO mice which likely explained the modestly decreased
atherosclerosis. However, CFB KO mice were almost entirely protected from an accelerated atherosclerosis that
was induced by chronic intraperitoneal LPS administration
(1124). These results demonstrate that enhancement of atherosclerosis by LPS or chronic infection need not be mediated solely by Toll-like receptors but that alternative complement pathway activation can also contribute.
Mannose- or mannan-binding lectin (MBL) is an initiating
factor in the lectin pathway of complement activation with
a structure highly analogous to C1q. It can coat and pro-
many copies of its target zymogen, resulting in an amplifying cascade that ends with the production of many copies of
active C9. Active C9 multimers are inserted through the cell
membrane to form numerous large pores causing osmotic
lysis of the targeted cell. Several of the lytic pieces generated
by zymogen activation are inflammatory or attach to cells
and target them for phagocytosis, a process called opsonization. The classical, alternative, and lectin pathways lead to
activation of the key C3 and C5 convertases. Host cells
carry inhibitors and regulators of the pathway to minimize
peripheral damage. Briefly, for classical activation, C1qa
(complement component 1, q subcomponent, A chain) recognizes and binds the Fc portion of antibodies bound to
antigen, commonly on the surface of an invading bacterium. C1q exists as a hexamer that looks like an inverted
umbrella, with a globular head at the tip of each of its
radiating spokes that binds targets. C1q is joined by two
copies each of C1r and C1s to form the C1 complex. C1q
bound to at least two separate Fc regions activates the protease C1r which then cleaves and activates C1s. C1s cleaves
C4 and C2 with resulting C4b and C2a forming a protease
to cleave C3. Cleavage of C3 and C4 exposes an internal,
reactive thioester bond which covalently binds nearby
membrane polysaccharides or proteins, with hydroxyls or
amines, respectively, displacing the sulfur in the bond. In an
acqueous environment such as plasma, the thioester bonds
are merely hydrolyzed, and the components are inactivated.
C3b, now covalently bound to the membrane joins with
C4bC2a to form a C5 convertase with C5b as its product.
C5b then assembles C6, C7, and C8 with insertion of C7
and C8 into the cell membrane followed by assembly of C9
multimers and pore formation. Factors C5b through C9 are
together referred to as the membrane attack complex
(MAC).
1477
PAUL N. HOPKINS
comial respiratory infection was reported to be increased
10-fold in patients with very low HDL (20 mg/dl) (406).
1. Emphasis on the role of HDL in innate immunity
may clarify observations regarding reverse
cholesterol transport
Two studies have recently examined cholesterol efflux capacity of plasma as a predictor of CAD in humans (902,
1038). Both studies utilized plasma from the test subjects
that had been treated to remove APOB-containing lipoproteins. This treated plasma was then diluted (2 4%) and
incubated with macrophages that had been previously
loaded with radiolabeled FC. The FC loading was done in
conditions such that essentially no radiolabeled CE was
formed. In both studies, greater efflux capacity of subject
plasma (the amount of radiolabeled FC transferred from the
macrophages to the diluted plasma) was strongly associated
with reduced risk of CAD in case-control (cross-sectional)
analyses. This efflux capacity of plasma was more closely
associated with CAD than with HDL-C or even APOA1
itself. In addition to HDL-C, the efflux capacity in this assay
was strongly dependent on the presence of precursor HDL
particles, pre--1 HDL, which are the primary cholesterol/
phospholipid acceptors via ABCA1 (395). In the second
study (1038), investigators examined how the radioactive
FC label distributed to different plasma components. Only
30% of the labeled FC was found in HDL particles.
About 45% was associated with APOA1 as either free or
possibly pre- HDL (but not in the HDL density range).
Much of the remainder was bound to albumin, demonstrating a significant nonspecific transfer of cholesterol from the
macrophages. Also in the second study, data were available
for 3-year incidence of new clinical coronary events within
a large angiographic cohort which was followed prospectively. The incidence of new coronary events was paradoxically higher among those with greater plasma efflux capacity (even though cross-sectional analyses in the same group
at baseline and in a separate community case-control group
1478
1479
In addition to its role as a cholesterol/phospholipid transporter, ABCA1 supports a remarkable array of anti-inflammatory intracellular signaling when bound by APOA1
(1349). In BMT experiments, lack of macrophage ABCA1
increased atherosclerosis in LDLR-deficient mice (1849).
Overexpression of ABCA1 on endothelium led to both a
reduction in atherosclerosis and an increase in HDL in
C57Bl/6 mice fed an atherogenic Paigen diet (1838). A brief
summary of the control of ABCA1 expression and antiinflammatory signaling mediated by ABCA1-APOA1 interaction follows.
PAUL N. HOPKINS
membrane away from cholesterol-enriched lipid rafts. The
exovesiculated domain is pinched off and surrounded by
APOA1 with the formation of nascent, PC-rich, cholesterol-poor HDL disks with two to four APOA1 molecules/disk
(814, 1869). These PC-rich discs are particularly good acceptors of free cholesterol (FC), which is transferred to the
nascent disks by diffusion from nearby lipid rafts. In other
models, transfer of PC and FC to HDL occurs together and
involves initial binding of APOA1 to a low-capacity binding
site on or near ABCA1, followed by activation of JAK2, and
transfer of the APOA1 to a high-capacity binding site followed by PC and FC loading (1065). Active ABCA1 is able
to transfer cholesterol to other apoproteins with amphipathic alpha helices, including most apoproteins carried by
HDL as well as ApoE (1065). Paradoxically, early cholesterol enrichment of the ER seems to promote ABCA1 ubiquitination and degradation (512), and cell membraine cholesterol enrichment impairs APOA1 binding to the highcapacity binding site on ABCA1 (814), phenomena that
could both promote foam cell formation.
1480
From the above paragraph, SMS2 might reasonably be expected to be protective. However, net results from alterating SMS2 activity appear to depend on cell status. It should
be noted that the activity ascribed to SMS2 has been described in the literature as belonging to a putative PC-PLC,
which is inhibited by the compound D609 (13). In some
models, inhibition of SMS2 with D609 increased ceramide
and resulted in impaired proliferation and increased apoptosis (13). In contrast, overexpressing SMS1 and/or SMS2
together with TNF- treatment in Chinese hamster ovary
cells or LPS treatment in macrophages led to increased apoptosis in cells while siRNA knockdown of SMS1/2 protected against apoptosis (427). In these models, SMS1/2
seemed to increase membrane SM with increased lipid raft
formation and greater TNFR1 and TLR4 activity. Overexpression of SMS1/2 also suppressed HDL-mediated cholesterol efflux in this study. More recently, administration of
D609 to ApoE-null mice was reported to arrest progression
of atherosclerosis and resulted in a more stable plaque phenotype (2077). The investigators further found that SMS2
activity appeared to be required for expression of the proinflammatory LOX-1 receptor in endothelial cells and that
D609 treatment decreased endothelial expression of
VCAM-1, ICAM-1, and MCP-1. In another study, adenoviral delivery of SMS2 to ApoE KO mice increased atherosclerosis substantially but also increased non-HDL lipid levels and decreased HDL, confounding the interpretation of
direct SMS2 effects (1914). Effects of these manipulations
on SM in lipoproteins should also be examined since APOBcontaining lipoproteins that have more SM are more subject
to aggregation upon treatment with sphingomyelinase.
As noted above, the calcium influx triggered by APOA1
binding to ABCA1 promotes calmodulin binding to ABCA1
and helps prevent calpain-mediated degradation. In addition, calmodulin activation also promotes JAK2 activation
in some manner that requires calcineurin (885). JAK2 apparently binds ABCA1 directly and its activation is required
for normal lipid transfer activity to APOA1 and HDL
(1065). Activation of JAK2 by APOA1 binding to ABCA1
also results in STAT3 binding to ABCA1 and STAT3 activation. JAK2/STAT3 signaling is strongly anti-inflammatory in endothelial cells and macrophages. Recently, STAT3
was shown to induce production of tristetraprolin (TTP), a
specific inhibitor of several inflammatory cytokines including TNF- and IL-1. TTP acts by binding the 3= end of the
Sphingomyelin (SM) is also transferred to HDL in its interaction with various cells. In artificial membranes, SM is said
to promote lipidation of reconstituted HDL or APOA1
(412, 564). One might postulate that this is due to SMmediated sequestration of membrane cholesterol into the
discrete, cholesterol-enriched lipid rafts, domains which remain separate from the PC-enriched areas where ABCA1
seems to partition itself. SM seems uniquely suited to this
task due to its saturated fatty acid tails which effectively
interact with cholesterol while the large, highly charged
head allows effective interaction with the aqueous environment.
with the membrane might lead to net transfer of phosphocholine from PC to ceramide, thereby regenerating SM and
decreasing ceramide but with a net result of increased DAG.
Such a reaction, together with increased calcium entry into
the cytoplasm, occurs after APOA1 binding to ABCA1, the
resulting DAG leading to activation of PKC and stabilization of ABCA1 (as noted above). An inhibitor of SMS2 (and
SMS1) decreased PKC activation and also impaired cholesterol transfer to APOA1 (831, 2010).
Whole-body SR-B1 KO in mice increased HDL-C and atherosclerosis substantially while liver-specific KO resulted in
similar effects on HDL-C but lesser effects on atherosclerosis (1523). However, bone marrow-derived cells (353,
1848) and endothelium deficient in SR-B1 each contribute
to excess atherosclerosis without affecting serum lipid levels
(1523). Macrophage-specific knockout of SR-B1 had a
lesser effect on atherosclerosis, suggesting that HDL interactions with SR-B1 on other cells may be more important
(1913, 2081). An altered ratio of plasma unesterified to
total cholesterol in SR-B1 deficiency leads to platelet activation and may contribute to the atherosclerosis (772).
These studies suggest reverse cholesterol transport is not the
only means whereby SR-B1 affects atherogenesis.
1481
PAUL N. HOPKINS
ularly S1PR1) (1338, 1888, 1953). HDL carries over 50%
of the S1P in plasma (77, 78).
Of potentially great interest is the relatively neglected apolipoprotein, apoM, found on only 5% of HDL particles
(as well as some non-HDL lipoproteins). ApoM apparently
mediates virtually all S1P removal from cells and loading
onto HDL, as well as subsequent receptor interaction (322,
323). ApoM also seems to be required for normal regeneration of pre- HDL, possibly by interacting with PLTP
or hepatic lipase (HL) (321). Overexpression of apoM
was reported to decrease atherosclerosis progression in
hyperlipidemic mice (321, 1970), but effects of knockout
on atherosclerosis have apparently not been examined
(see TABLE 8).
1482
Production of S1P occurs in response to cytokines that utilize TRAF2 and TRAF6 in their downstream signaling.
TRAF2 binds and activates sphingosine kinase 1 (SPHK1).
The S1P produced was shown to be a cofactor required for
TRAF2 (and probably TRAF6) to function as a E3 ubiquitin ligase (1665). The S1P produced by these reactions then
seems to be transported, in part, to HDL by apo M. Thus
S1P has the unusual distinction of supporting proinflammatory signaling directly as an intracellular cofactor (and
when bound to certain of its GPCR) while directing antiinflammatory signaling when presented to S1PR1 by HDL.
This duality may help explain the reported strong positive
association between CAD and the ratio of non-HDL-bound
S1P to HDL-bound S1P (1543).
The molecular biology and related signaling of the pathways presented in this section have been the topic of numerous recent reviews. This is particularly true of thrombosis
and angiogenesis pathways. For this reason, these topics
will be presented in a relatively abbreviated format with a
few orienting comments as they relate specifically to atherogenesis.
1483
PAUL N. HOPKINS
(1135). These findings provide new impetus for controlling
weight gain and other factors that may increase deposition
of coronary perivascular fat, recently shown to be strongly
associated with atherosclerotic plaques in humans (1114,
1161, 1750).
C. Vaso-vasorum
XII. PERSPECTIVES
Gene manipulation of factors which control the neovascularization of atherosclerotic plaques demonstrate that proliferation of vaso vasorum substantially aggravates the
growth of advanced plaques. Some investigators had hypothesized that treatment of mice to promote angiogenesis
(as with G-CSF and GM-CSF) would limit atherosclerosis
but were surprised to find the opposite (691). Details of a
number of such interventions are presented in TABLE 10.
Finally, perivascular fat has been suspected to promote increased vascular inflammation from the outside in (1455).
In a remarkable series of investigations, transplanting
perivascular fat around a carotid vessel was shown to markedly increase intimal proliferation after wire injury, enhance adventitial inflammation, stimulate angiogenesis of
new vaso-vasorum, and markedly promote localized atherosclerosis in LDLR KO mice. Furthermore, perivascular
fat was much more proinflammatory than fat from other
sources and showed greatly increased release of TNF-,
MCP-1 (CCL2), and MIP-1 (CCL3) upon high-fat feeding
1484
In choosing targets for intervention, the importance of balance must always be kept in mind, particularly for longterm use in asymptomatic populations. Reduction of inflammation, such as by increasing Treg (perhaps with vitamin D supplementation), or by targeted inhibition of
TNF-, PKC, NOX2, or other interventions must be balanced against risk of immunodeficiency (even if subtle), a
judgement that will ultimately require large clinical trials.
Direct TNF- inhibition appears to benefit patients with
autoimmune disease, but further understanding of why
there is a gradual increase in TNF signaling with age accompanied by a general deficit of cellular antioxidant defenses
would perhaps lead to even greater benefits. Pharmacological inhibition of the apoptosis-promoting actions of various signaling molecules (such as p66Shc or JNK) needs to
be weighed against a potential increased risk of cancer.
Increasing the strongly cytoprotective signaling of PKB paradoxically seems to decrease longevity. Bolstering natural
cellular antioxidant defenses seems worth pursuing, recog-
ACKNOWLEDGMENTS
Address for reprint requests and other correspondence:
P. N. Hopkins, Cardiovascular Genetics, Dept. of Internal
Medicine, Univ. of Utah, 420 Chipeta Way, Rm. 1160, Salt
Lake City, UT (e-mail: paul.hopkins@utah.edu).
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared
by the author.
REFERENCES
1. Abbas AK, Lichtman AH, Pillai S. Cellular and Molecular Immunology. Philadelphia, PA:
Saunders/Elsevier, 2012, p. viii.
2. Abbasi F, McLaughlin T, Lamendola C, Reaven GM. The relationship between glucose disposal in response to physiological hyperinsulinemia and basal glucose and
free fatty acid concentrations in healthy volunteers. J Clin Endocrinol Metab 85:
12511254, 2000.
3. Abela GS, Aziz K. Cholesterol crystals rupture biological membranes and human
plaques during acute cardiovascular eventsa novel insight into plaque rupture by
scanning electron microscopy. Scanning 28: 110, 2006.
4. Abela GS, Aziz K, Vedre A, Pathak DR, Talbott JD, Dejong J. Effect of cholesterol
crystals on plaques and intima in arteries of patients with acute coronary and cerebrovascular syndromes. Am J Cardiol 103: 959 968, 2009.
5. AbouAlaiwi WA, Takahashi M, Mell BR, Jones TJ, Ratnam S, Kolb RJ, Nauli SM. Ciliary
polycystin-2 is a mechanosensitive calcium channel involved in nitric oxide signaling
cascades. Circ Res 104: 860 869, 2009.
6. Abram SR, Hodnett BL, Summers RL, Coleman TG, Hester RL. Quantitative circulatory physiology: an integrative mathematical model of human physiology for medical education. Adv Physiol Educ 31: 202210, 2007.
7. Acton S, Rigotti A, Landschulz KT, Xu S, Hobbs HH, Krieger M. Identification of
scavenger receptor SR-BI as a high density lipoprotein receptor. Science 271: 518
520, 1996.
8. Adachi H, Fujiwara Y, Kondo T, Nishikawa T, Ogawa R, Matsumura T, Ishii N, Nagai
R, Miyata K, Tabata M, Motoshima H, Furukawa N, Tsuruzoe K, Kawashima J,
Takeya M, Yamashita S, Koh GY, Nagy A, Suda T, Oike Y, Araki E. Angptl 4 deficiency
improves lipid metabolism, suppresses foam cell formation and protects against
atherosclerosis. Biochem Biophys Res Commun 379: 806 811, 2009.
1485
The field seems ripe for gleaning new understanding in humans of how dietary fats such as saturated fat (particularly
palmitic acid), PUFA, and n-3 fatty acids might affect ceramide signaling, resolution-promoting eicosanoid production, ER stress, and other signaling pathways. Other areas
of great interest that may find application in atherosclerosis
prevention include potential dietary and pharmacological
effects on formation of electrophilic LDL, IKK1/2 inhibition (such as by salicylates or curcumin), manipulation of
protective S1P signaling stimulated by HDL, utility of selected electrophilic compounds, and generation of appropriate levels of H2S. The potential benefit of stimulating
immune tolerance with increased Treg activity, perhaps using human APOB-100 peptides as in a recent mouse study
(751) and/or HSP60, is an important avenue to explore in
human trials. The apparent utility of direct thrombin inhibition in advanced atherosclerosis also seems promising. All
these potential means of intervention appear to have relatively straightforward application and deserve further
study, hopefully with prompt translation into human studies. A human interventional trial to test whether antibodybased inhibition of IL-1 (using canakinamab) can prevent
recurrent coronary events is already underway (1485).
Other highly directed anti-inflammatory therapies may also
move to the forefront in the near future (1054).
PAUL N. HOPKINS
9. Adachi T, Pimentel DR, Heibeck T, Hou X, Lee YJ, Jiang B, Ido Y, Cohen RA.
S-glutathiolation of Ras mediates redox-sensitive signaling by angiotensin II in vascular smooth muscle cells. J Biol Chem 279: 2985729862, 2004.
10. Adam AP, Sharenko AL, Pumiglia K, Vincent PA. Src-induced tyrosine phosphorylation of VE-cadherin is not sufficient to decrease barrier function of endothelial monolayers. J Biol Chem 285: 70457055, 2010.
11. Adams MR, Register TC, Golden DL, Wagner JD, Williams JK. Medroxyprogesterone acetate antagonizes inhibitory effects of conjugated equine estrogens on coronary artery atherosclerosis. Arterioscler Thromb Vasc Biol 17: 217221, 1997.
12. Adamson S, Leitinger N. Phenotypic modulation of macrophages in response to
plaque lipids. Curr Opin Lipidol 22: 335342, 2011.
13. Adibhatla RM, Hatcher JF, Gusain A. Tricyclodecan-9-yl-xanthogenate (D609)
mechanism of actions: a mini-review of literature. Neurochem Res 2011.
14. Adler AS, Sinha S, Kawahara TL, Zhang JY, Segal E, Chang HY. Motif module map
reveals enforcement of aging by continual NF-kappaB activity. Genes Dev 21: 3244
3257, 2007.
15. Agrawal S, Febbraio M, Podrez E, Cathcart MK, Stark GR, Chisolm GM. Signal
transducer and activator of transcription 1 is required for optimal foam cell formation and atherosclerotic lesion development. Circulation 115: 2939 2947, 2007.
16. Aherne CM, Kewley EM, Eltzschig HK. The resurgence of A2B adenosine receptor
signaling. Biochim Biophys Acta 2010.
18. Ahmad Y, Shelmerdine J, Bodill H, Lunt M, Pattrick MG, Teh LS, Bernstein RM,
Walker MG, Bruce IN. Subclinical atherosclerosis in systemic lupus erythematosus
(SLE): the relative contribution of classic risk factors and the lupus phenotype. Rheumatology 46: 983988, 2007.
19. Ahmed A, Fujisawa T. Multiple roles of angiopoietins in atherogenesis. Curr Opin
Lipidol 22: 380 385, 2011.
20. Ahmed A, Fujisawa T, Niu XL, Ahmad S, Al-Ani B, Chudasama K, Abbas A, Potluri R,
Bhandari V, Findley CM, Lam GK, Huang J, Hewett PW, Cudmore M, Kontos CD.
Angiopoietin-2 confers atheroprotection in apoE/ mice by inhibiting LDL oxidation via nitric oxide. Circ Res 104: 13331336, 2009.
21. Aiello RJ, Bourassa PA, Lindsey S, Weng W, Freeman A, Showell HJ. Leukotriene B4
receptor antagonism reduces monocytic foam cells in mice. Arterioscler Thromb Vasc
Biol 22: 443 449, 2002.
22. Aiello RJ, Brees D, Bourassa PA, Royer L, Lindsey S, Coskran T, Haghpassand M,
Francone OL. Increased atherosclerosis in hyperlipidemic mice with inactivation of
ABCA1 in macrophages. Arterioscler Thromb Vasc Biol 22: 630 637, 2002.
40. Alber DG, Powell KL, Vallance P, Goodwin DA, Grahame-Clarke C. Herpesvirus
infection accelerates atherosclerosis in the apolipoprotein E-deficient mouse. Circulation 102: 779 785, 2000.
41. Alberici LC, Oliveira HCF, Paim BA, Mantello CC, Augusto AC, Zecchin KG, Gurgueira SA, Kowaltowski AJ, Vercesi AE. Mitochondrial ATP-sensitive K channels as
redox signals to liver mitochondria in response to hypertriglyceridemia. Free Radical
Biol Med 47: 14321439, 2009.
24. Aikawa M, Rabkin E, Okada Y, Voglic SJ, Clinton SK, Brinckerhoff CE, Sukhova GK,
Libby P. Lipid lowering by diet reduces matrix metalloproteinase activity and increases collagen content of rabbit atheroma: a potential mechanism of lesion stabilization. Circulation 97: 24332444, 1998.
42. Albers HM, Ovaa H. Chemical evolution of autotaxin inhibitors. Chem Rev 112:
25932603, 2012.
26. Aikawa M, Sugiyama S, Hill CC, Voglic SJ, Rabkin E, Fukumoto Y, Schoen FJ, Witztum
JL, Libby P. Lipid lowering reduces oxidative stress and endothelial cell activation in
rabbit atheroma. Circulation 106: 1390 1396, 2002.
27. Aikawa M, Voglic SJ, Sugiyama S, Rabkin E, Taubman MB, Fallon JT, Libby P. Dietary
lipid lowering reduces tissue factor expression in rabbit atheroma. Circulation 100:
12151222, 1999.
1486
45. Alexander A, Walker CL. The role of LKB1 and AMPK in cellular responses to stress
and damage. FEBS Lett 585: 952957, 2011.
28. Ait-Oufella H, Herbin O, Bouaziz JD, Binder CJ, Uyttenhove C, Laurans L, Taleb S,
Van Vre E, Esposito B, Vilar J, Sirvent J, Van Snick J, Tedgui A, Tedder TF, Mallat Z.
B cell depletion reduces the development of atherosclerosis in mice. J Exp Med 207:
1579 1587, 2010.
54. Allingham MJ, van Buul JD, Burridge K. ICAM-1-mediated, Src- and Pyk2-dependent
vascular endothelial cadherin tyrosine phosphorylation is required for leukocyte
transendothelial migration. J Immunol 179: 4053 4064, 2007.
55. Almontashiri NA, Fan M, Chen HH, Teng AC, Chen BL, McPherson R, Roberts R,
Stewart AF. Abstract 15730: serum interferon alpha 21 is a biomarker of the 9p21.3
risk locus for coronary artery disease (Abstract). Circulation 124: A15730.
56. Alp NJ, McAteer MA, Khoo J, Choudhury RP, Channon KM. Increased endothelial
tetrahydrobiopterin synthesis by targeted transgenic GTP-cyclohydrolase I overexpression reduces endothelial dysfunction and atherosclerosis in ApoE-knockout
mice. Arterioscler Thromb Vasc Biol 24: 445 450, 2004.
67. Anthis NJ, Haling JR, Oxley CL, Memo M, Wegener KL, Lim CJ, Ginsberg MH,
Campbell ID. Beta integrin tyrosine phosphorylation is a conserved mechanism for
regulating talin-induced integrin activation. J Biol Chem 284: 36700 36710, 2009.
68. Aono J, Suzuki J, Ogimoto A, Okayama H, Iwai M, Horiuchi M, Higaki J. Abstract
12460: deletion of the angiotensin II type 1a receptor prevents atherosclerotic
plaque rupture. Circulation 122: A12460, 2010.
69. Aoyama T, Takeshita K, Kikuchi R, Yamamoto K, Cheng XW, Liao JK, Murohara T.
gamma-Secretase inhibitor reduces diet-induced atherosclerosis in apolipoprotein
E-deficient mice. Biochem Biophys Res Commun 383: 216 221, 2009.
70. Apostolov EO, Basnakian AG, Yin X, Ok E, Shah SV. Modified LDLs induce proliferation-mediated death of human vascular endothelial cells through MAPK pathway.
Am J Physiol Heart Circ Physiol 292: H1836 H1846, 2007.
71. Apostolov EO, Shah SV, Ok E, Basnakian AG. Carbamylated low-density lipoprotein
induces monocyte adhesion to endothelial cells through intercellular adhesion molecule-1 and vascular cell adhesion molecule-1. Arterioscler Thromb Vasc Biol 27:
826 832, 2007.
72. Apostolov EO, Shah SV, Ray D, Basnakian AG. Scavenger receptors of endothelial
cells mediate the uptake and cellular proatherogenic effects of carbamylated LDL.
Arterioscler Thromb Vasc Biol 29: 16221630, 2009.
73. Aprahamian T, Rifkin I, Bonegio R, Hugel B, Freyssinet JM, Sato K, Castellot JJ Jr,
Walsh K. Impaired clearance of apoptotic cells promotes synergy between atherogenesis and autoimmune disease. J Exp Med 199: 11211131, 2004.
74. Arai S, Shelton JM, Chen M, Bradley MN, Castrillo A, Bookout AL, Mak PA, Edwards
PA, Mangelsdorf DJ, Tontonoz P, Miyazaki T. A role for the apoptosis inhibitory
factor AIM/Spalpha/Api6 in atherosclerosis development. Cell Metab 1: 201213,
2005.
75. Arakawa K, Urata H. Hypothesis regarding the pathophysiological role of alternative
pathways of angiotensin II formation in atherosclerosis. Hypertension 36: 638 641,
2000.
76. Araki R, Fujimori A, Hamatani K, Mita K, Saito T, Mori M, Fukumura R, Morimyo M,
Muto M, Itoh M, Tatsumi K, Abe M. Nonsense mutation at Tyr-4046 in the DNAdependent protein kinase catalytic subunit of severe combined immune deficiency
mice. Proc Natl Acad Sci USA 94: 2438 2443, 1997.
77. Argraves KM, Argraves WS. HDL serves as a S1P signaling platform mediating a
multitude of cardiovascular effects. J Lipid Res 48: 23252333, 2007.
58. Ambrose JA, Barua RS. The pathophysiology of cigarette smoking and cardiovascular
disease: an update. J Am Coll Cardiol 43: 17311737, 2004.
78. Argraves KM, Gazzolo PJ, Groh EM, Wilkerson BA, Matsuura BS, Twal WO, Hammad SM, Argraves WS. High density lipoprotein-associated sphingosine 1-phosphate
promotes endothelial barrier function. J Biol Chem 283: 25074 25081, 2008.
59. Amir S, Binder CJ. Experimental immunotherapeutic approaches for atherosclerosis. Clin Immunol 134: 66 79, 2010.
60. An G, Miwa T, Song WL, Lawson JA, Rader DJ, Zhang Y, Song WC. CD59 but not
DAF deficiency accelerates atherosclerosis in female ApoE knockout mice. Mol
Immunol 46: 17021709, 2009.
61. Andersson J, Libby P, Hansson GK. Adaptive immunity and atherosclerosis. Clin
Immunol 134: 33 46, 2010.
62. Ando Y, Shimizugawa T, Takeshita S, Ono M, Shimamura M, Koishi R, Furukawa H.
A decreased expression of angiopoietin-like 3 is protective against atherosclerosis in
apoE-deficient mice. J Lipid Res 44: 1216 1223, 2003.
79. Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, Long JM, Wynshaw-Boris A,
Poli G, Olefsky J, Karin M. IKK-beta links inflammation to obesity-induced insulin
resistance. Nat Med 11: 191198, 2005.
80. Arunachalam G, Yao H, Sundar IK, Caito S, Rahman I. SIRT1 regulates oxidant- and
cigarette smoke-induced eNOS acetylation in endothelial cells: role of resveratrol.
Biochem Biophys Res Commun 393: 66 72, 2010.
81. Asai K, Funaki C, Hayashi T, Yamada K, Naito M, Kuzuya M, Yoshida F, Yoshimine N,
Kuzuya F. Dexamethasone-induced suppression of aortic atherosclerosis in cholesterol-fed rabbits. Possible mechanisms. Arterioscler Thromb 13: 892 899, 1993.
82. Aslanian AM, Chapman HA, Charo IF. Transient role for CD1d-restricted natural
killer T cells in the formation of atherosclerotic lesions. Arterioscler Thromb Vasc Biol
25: 628 632, 2005.
64. Andres V, Gascon-Irun M, Pandolfi PP, Gonzalez-Navarro H. Atheroma development in apolipoprotein E-null mice is not affected by partial inactivation of PTEN.
Front Biosci 11: 2739 2745, 2006.
83. Aslanian AM, Charo IF. Targeted disruption of the scavenger receptor and chemokine CXCL16 accelerates atherosclerosis. Circulation 114: 583590, 2006.
84. Assimes TL, Knowles JW, Priest JR, Basu A, Borchert A, Volcik KA, Grove ML, Tabor
HK, Southwick A, Tabibiazar R, Sidney S, Boerwinkle E, Go AS, Iribarren C, Hlatky
MA, Fortmann SP, Myers RM, Kuhn H, Risch N, Quertermous T. A near null variant
of 12/15-LOX encoded by a novel SNP in ALOX15 and the risk of coronary artery
disease. Atherosclerosis 198: 136 144, 2008.
1487
53. Allen S, Khan S, Mohanna FA, PB, Yacoub M. Native low density lipoprotein-induced
calcium transients trigger VCAM-1 and E-selectin expression in cultured human
vascular endothelial cells. J Clin Invest 101: 1064 1075, 1998.
66. Anthis NJ, Campbell ID. The tail of integrin activation. Trends Biochem Sci 36: 191
198, 2011.
PAUL N. HOPKINS
85. Atkins GB, Wang Y, Mahabeleshwar GH, Shi H, Gao H, Kawanami D, Natesan V, Lin
Z, Simon DI, Jain MK. Hemizygous deficiency of Kruppel-like factor 2 augments
experimental atherosclerosis. Circ Res 103: 690 693, 2008.
104. Bansode RR, Huang W, Roy SK, Mehta M, Mehta KD. Protein kinase C deficiency
increases fatty acid oxidation and reduces fat storage. J Biol Chem 283: 231236,
2008.
86. Augustin HG, Koh GY, Thurston G, Alitalo K. Control of vascular morphogenesis and
homeostasis through the angiopoietin-Tie system. Nat Rev Mol Cell Biol 10: 165177,
2009.
87. Aveleira CA, Lin CM, Abcouwer SF, Ambrsio AF, Antonetti DA. TNF- signals
through PKC/NF-B to alter the tight junction complex and increase retinal endothelial cell permeability. Diabetes 59: 28722882, 2010.
106. Barajas B, Che N, Yin F, Rowshanrad A, Orozco LD, Gong KW, Wang X, Castellani
LW, Reue K, Lusis AJ, Araujo JA. NF-E2-related factor 2 promotes atherosclerosis by
effects on plasma lipoproteins and cholesterol transport that overshadow antioxidant protection. Arterioscler Thromb Vasc Biol 31: 58 66, 2011.
88. Averill MM, Barnhart S, Becker L, Li X, Heinecke JW, Leboeuf RC, Hamerman JA,
Sorg C, Kerkhoff C, Bornfeldt KE. S100A9 differentially modifies phenotypic states
of neutrophils, macrophages, and dendritic cells: implications for atherosclerosis and
adipose tissue inflammation. Circulation 123: 1216 1226, 2011.
89. Avraham R, Yarden Y. Feedback regulation of EGFR signalling: decision making by
early and delayed loops. Nat Rev Mol Cell Biol 12: 104 117, 2011.
90. Baba SP, Barski OA, Ahmed Y, OToole TE, Conklin DJ, Bhatnagar A, Srivastava S.
Reductive metabolism of AGE precursors: a metabolic route for preventing AGE
accumulation in cardiovascular tissue. Diabetes 58: 2486 2497, 2009.
91. Babaev VR, Chew JD, Ding L, Davis S, Breyer MD, Breyer RM, Oates JA, Fazio S,
Linton MF. Macrophage EP4 deficiency increases apoptosis and suppresses early
atherosclerosis. Cell Metab 8: 492501, 2008.
93. Babaev VR, Li L, Shah S, Fazio S, Linton MF, May JM. Combined vitamin C and vitamin
E deficiency worsens early atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 30: 17511757, 2010.
94. Babaev VR, Patel MB, Semenkovich CF, Fazio S, Linton MF. Macrophage lipoprotein
lipase promotes foam cell formation and atherosclerosis in low density lipoprotein
receptor-deficient mice. J Biol Chem 275: 2629326299, 2000.
95. Babaev VR, Runner RP, Fan D, Ding L, Zhang Y, Tao H, Erbay E, Gorgun CZ, Fazio
S, Hotamisligil GS, Linton MF. Macrophage Mal1 deficiency suppresses atherosclerosis in low-density lipoprotein receptor-null mice by activating peroxisome proliferator-activated receptor--regulated genes. Arterioscler Thromb Vasc Biol 2011.
96. Babaev VR, Whitesell RR, Li L, Linton MF, Fazio S, May JM. Selective macrophage
ascorbate deficiency suppresses early atherosclerosis. Free Radic Biol Med 50: 2736,
2011.
97. Babaev VR, Yancey PG, Ryzhov SV, Kon V, Breyer MD, Magnuson MA, Fazio S,
Linton MF. Conditional knockout of macrophage PPARgamma increases atherosclerosis in C57BL/6 and low-density lipoprotein receptor-deficient mice. Arterioscler
Thromb Vasc Biol 25: 16471653, 2005.
98. Back M, Sultan A, Ovchinnikova O, Hansson GK. 5-Lipoxygenase-activating protein:
a potential link between innate and adaptive immunity in atherosclerosis and adipose
tissue inflammation. Circ Res 100: 946 949, 2007.
99. Baitsch D, Bock HH, Engel T, Telgmann R, Muller-Tidow C, Varga G, Bot M, Herz
J, Robenek H, von Eckardstein A, Nofer JR. Apolipoprotein E induces antiinflammatory phenotype in macrophages. Arterioscler Thromb Vasc Biol 31: 1160 1168, 2011.
100. Bakker W, Sipkema P, Stehouwer CD, Serne EH, Smulders YM, van Hinsbergh VW,
Eringa EC. Protein kinase C theta activation induces insulin-mediated constriction of
muscle resistance arteries. Diabetes 57: 706 713, 2008.
101. Balligand JL, Feron O, Dessy C. eNOS activation by physical forces: from short-term
regulation of contraction to chronic remodeling of cardiovascular tissues. Physiol Rev
89: 481534, 2009.
102. Ballinger SW, Patterson C, Knight-Lozano CA, Burow DL, Conklin CA, Hu Z, Reuf
J, Horaist C, Lebovitz R, Hunter GC, McIntyre K, Runge MS. Mitochondrial integrity
and function in atherogenesis. Circulation 106: 544 549, 2002.
103. Bancells C, Benitez S, Villegas S, Jorba O, Ordonez-Llanos J, Sanchez-Quesada JL.
Novel phospholipolytic activities associated with electronegative low-density lipoprotein are involved in increased self-aggregation. Biochemistry 47: 8186 8194,
2008.
1488
108. Barish GD, Atkins AR, Downes M, Olson P, Chong LW, Nelson M, Zou Y, Hwang H,
Kang H, Curtiss L, Evans RM, Lee CH. PPARdelta regulates multiple proinflammatory pathways to suppress atherosclerosis. Proc Natl Acad Sci USA 105: 4271 4276,
2008.
109. Barish GD, Yu RT, Karunasiri M, Ocampo CB, Dixon J, Benner C, Dent AL, Tangirala
RK, Evans RM. Bcl-6 and NF-kappaB cistromes mediate opposing regulation of the
innate immune response. Genes Dev 24: 2760 2765, 2010.
110. Barish GD, Yu RT, Karunasiri MS, Becerra D, Kim J, Tseng TW, Tai LJ, Leblanc M,
Diehl C, Cerchietti L, Miller YI, Witztum JL, Melnick AM, Dent AL, Tangirala RK,
Evans RM. The Bcl6-SMRT/NCoR cistrome represses inflammation to attenuate
atherosclerosis. Cell Metab 15: 554 562, 2012.
111. Barlic J, Murphy PM. Chemokine regulation of atherosclerosis. J Leukoc Biol 82:
226 236, 2007.
112. Barnholt KE, Kota RS, Aung HH, Rutledge JC. Adenosine blocks IFN-gamma-induced phosphorylation of STAT1 on serine 727 to reduce macrophage activation. J
Immunol 183: 6767 6777, 2009.
113. Barry-Lane PA, Patterson C, van der Merwe M, Hu Z, Holland SM, Yeh ET, Runge
MS. p47phox is required for atherosclerotic lesion progression in ApoE(/) mice.
J Clin Invest 108: 15131522, 2001.
114. Bason C, Corrocher R, Lunardi C, Puccetti P, Olivieri O, Girelli D, Navone R, Beri R,
Millo E, Margonato A, Martinelli N, Puccetti A. Interaction of antibodies against
cytomegalovirus with heat-shock protein 60 in pathogenesis of atherosclerosis. Lancet 362: 19711977, 2003.
115. Batt KV, Avella M, Moore EH, Jackson B, Suckling KE, Botham KM. Differential
effects of low-density lipoprotein and chylomicron remnants on lipid accumulation in
human macrophages. Exp Biol Med 229: 528 537, 2004.
116. Baumgartl J, Baudler S, Scherner M, Babaev V, Makowski L, Suttles J, McDuffie M,
Tobe K, Kadowaki T, Fazio S, Kahn CR, Hotamisligil GS, Krone W, Linton M, Bruning
JC. Myeloid lineage cell-restricted insulin resistance protects apolipoproteinE-deficient mice against atherosclerosis. Cell Metab 3: 247256, 2006.
117. Bea F, Kreuzer J, Preusch M, Schaab S, Isermann B, Rosenfeld ME, Katus H, Blessing
E. Melagatran reduces advanced atherosclerotic lesion size and may promote plaque
stability in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 26: 2787
2792, 2006.
118. Beard RS Jr, Bearden SE. Vascular complications of cystathionine beta-synthase
deficiency: future directions for homocysteine-to-hydrogen sulfide research. Am J
Physiol Heart Circ Physiol 300: H13H26, 2011.
119. Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 87: 245313, 2007.
120. Bedford L, Lowe J, Dick LR, Mayer RJ, Brownell JE. Ubiquitin-like protein conjugation and the ubiquitin-proteasome system as drug targets. Nat Rev Drug Discov 2010.
121. Behm DJ, Ogbonna A, Wu C, Burns-Kurtis CL, Douglas SA. Epoxyeicosatrienoic
acids function as selective, endogenous antagonists of native thromboxane receptors: identification of a novel mechanism of vasodilation. J Pharmacol Exp Ther 328:
231239, 2009.
122. Beier KC, Kallinich T, Hamelmann E. Master switches of T-cell activation and differentiation. Eur Respir J 29: 804 812, 2007.
92. Babaev VR, Ishiguro H, Ding L, Yancey PG, Dove DE, Kovacs WJ, Semenkovich CF,
Fazio S, Linton MF. Macrophage expression of peroxisome proliferator-activated
receptor-alpha reduces atherosclerosis in low-density lipoprotein receptor-deficient mice. Circulation 116: 1404 1412, 2007.
131. Berliner JA, Leitinger N, Tsimikas S. The role of oxidized phospholipids in atherosclerosis. J Lipid Res 50: S207212, 2009.
132. Bernal-Mizrachi C, Gates AC, Weng S, Imamura T, Knutsen RH, DeSantis P, Coleman T, Townsend RR, Muglia LJ, Semenkovich CF. Vascular respiratory uncoupling
increases blood pressure and atherosclerosis. Nature 435: 502506, 2005.
133. Bernatchez PN, Allen BG, Gelinas DS, Guillemette G, Sirois MG. Regulation of
VEGF-induced endothelial cell PAF synthesis: role of p42/44 MAPK, p38 MAPK and
PI3K pathways. Br J Pharmacol 134: 12531262, 2001.
134. Bernatchez PN, Winstead MV, Dennis EA, Sirois MG. VEGF stimulation of endothelial cell PAF synthesis is mediated by group V 14 kDa secretory phospholipase A2. Br
J Pharmacol 134: 197205, 2001.
135. Bernhagen J, Krohn R, Lue H, Gregory JL, Zernecke A, Koenen RR, Dewor M,
Georgiev I, Schober A, Leng L, Kooistra T, Fingerle-Rowson G, Ghezzi P, Kleemann
R, McColl SR, Bucala R, Hickey MJ, Weber C. MIF is a noncognate ligand of CXC
chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med 13:
587596, 2007.
136. Berridge MJ. Cell Signalling Biology. Portland, OR: Portland Press Limited, 2012.
137. Berti JA, de Faria EC, Oliveira HC. Atherosclerosis in aged mice over-expressing the
reverse cholesterol transport genes. Braz J Med Biol Res 38: 391398, 2005.
138. Besmer P, Murphy JE, George PC, Qiu FH, Bergold PJ, Lederman L, Snyder HW Jr,
Brodeur D, Zuckerman EE, Hardy WD. A new acute transforming feline retrovirus
and relationship of its oncogene v-kit with the protein kinase gene family. Nature
320: 415 421, 1986.
139. Bhakat KK, Mantha AK, Mitra S. Transcriptional regulatory functions of mammalian
AP-endonuclease (APE1/Ref-1), an essential multifunctional protein. Antioxid Redox
Signal 11: 621 638, 2009.
140. Bhatia VK, Yun S, Leung V, Grimsditch DC, Benson GM, Botto MB, Boyle JJ, Haskard
DO. Complement C1q reduces early atherosclerosis in low-density lipoprotein
receptor-deficient mice. Am J Pathol 170: 416 426, 2007.
141. Bhattacharyya S, Borthakur A, Dudeja PK, Tobacman JK. Lipopolysaccharide-induced activation of NF-B non-canonical pathway requires BCL10 serine 138 and
NIK phosphorylations. Exp Cell Res 316: 33173327, 2010.
142. Bhattacharyya S, Borthakur A, Tyagi S, Gill R, Chen ML, Dudeja PK, Tobacman JK.
B-cell CLL/lymphoma 10 (BCL10) is required for NF-kappaB production by both
canonical and noncanonical pathways and for NF-kappaB-inducing kinase (NIK)
phosphorylation. J Biol Chem 285: 522530, 2010.
144. Bie J, Zhao B, Song J, Ghosh S. Improved insulin sensitivity in high fat- and high
cholesterol-fed Ldlr/ mice with macrophage-specific transgenic expression of
cholesteryl ester hydrolase: role of macrophage inflammation and infiltration into
adipose tissue. J Biol Chem 285: 13630 13637, 2010.
145. Bijli KM, Fazal F, Minhajuddin M, Rahman A. Activation of Syk by protein kinase
C-delta regulates thrombin-induced intercellular adhesion molecule-1 expression in
endothelial cells via tyrosine phosphorylation of RelA/p65. J Biol Chem 283: 14674
14684, 2008.
146. Billon-Gales A, Fontaine C, Douin-Echinard V, Delpy L, Berges H, Calippe B, Lenfant
F, Laurell H, Guery JC, Gourdy P, Arnal JF. Endothelial estrogen receptor-alpha plays
a crucial role in the atheroprotective action of 17beta-estradiol in low-density lipoprotein receptor-deficient mice. Circulation 120: 25672576, 2009.
147. Binder CJ, Hartvigsen K, Chang MK, Miller M, Broide D, Palinski W, Curtiss LK, Corr
M, Witztum JL. IL-5 links adaptive and natural immunity specific for epitopes of
oxidized LDL and protects from atherosclerosis. J Clin Invest 114: 427 437, 2004.
148. Binder CJ, Horkko S, Dewan A, Chang MK, Kieu EP, Goodyear CS, Shaw PX,
Palinski W, Witztum JL, Silverman GJ. Pneumococcal vaccination decreases atherosclerotic lesion formation: molecular mimicry between Streptococcus pneumoniae
and oxidized LDL. Nat Med 9: 736 743, 2003.
149. Bingham TC, Fisher EA, Parathath S, Reiss AB, Chan ES, Cronstein BN. A2A adenosine receptor stimulation decreases foam cell formation by enhancing ABCA1dependent cholesterol efflux. J Leukoc Biol 87: 683 690, 2010.
150. Birnbaumer L. Signal transduction by G proteins: basic principles, molecular diversity, and structural basis of their actions. In: Transduction Mechanisms in Cellular
Signaling, edited by Dennis EA, and Bradshaw RA. New York: Academic, 2011, p.
449 466.
151. Biro A, Ling WL, Arlaud GJ. Complement protein C1q recognizes enzymatically
modified low-density lipoprotein through unesterified fatty acids generated by cholesterol esterase. Biochemistry 49: 21672176, 2010.
152. Bischoff ED, Daige CL, Petrowski M, Dedman H, Pattison J, Juliano J, Li AC, Schulman IG. Non-redundant roles for LXRalpha and LXRbeta in atherosclerosis susceptibility in low density lipoprotein receptor knockout mice. J Lipid Res 51: 900 906,
2010.
153. Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte
subsets: cancer as a paradigm. Nat Immunol 11: 889 896, 2010.
154. Biyashev D, Veliceasa D, Kwiatek A, Sutanto MM, Cohen RN, Volpert OV. Natural
angiogenesis inhibitor signals through Erk5 activation of peroxisome proliferatoractivated receptor gamma (PPARgamma). J Biol Chem 285: 1351713524, 2010.
155. Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, Gluud C. Antioxidant supplements for prevention of mortality in healthy participants and patients with various
diseases. Cochrane Database Syst Rev CD007176, 2008.
156. Bjorkbacka H, Kunjathoor VV, Moore KJ, Koehn S, Ordija CM, Lee MA, Means T,
Halmen K, Luster AD, Golenbock DT, Freeman MW. Reduced atherosclerosis in
MyD88-null mice links elevated serum cholesterol levels to activation of innate
immunity signaling pathways. Nat Med 10: 416 421, 2004.
157. Blanc J, Alves-Guerra MC, Esposito B, Rousset S, Gourdy P, Ricquier D, Tedgui A,
Miroux B, Mallat Z. Protective role of uncoupling protein 2 in atherosclerosis. Circulation 107: 388 390, 2003.
158. Blaukat A, Ivankovic-Dikic I, Gronroos E, Dolfi F, Tokiwa G, Vuori K, Dikic I. Adaptor
proteins Grb2 and Crk couple Pyk2 with activation of specific mitogen-activated
protein kinase cascades. J Biol Chem 274: 1489314901, 1999.
159. Blum J, Fridovich I. Inactivation of glutathione peroxidase by superoxide radical. Arch
Biochem Biophys 240: 500 508, 1985.
160. Blumenthal A, Kobayashi T, Pierini LM, Banaei N, Ernst JD, Miyake K, Ehrt S. RP105
facilitates macrophage activation by Mycobacterium tuberculosis lipoproteins. Cell
Host Microbe 5: 35 46, 2009.
161. Bobryshev YV. Dendritic cells in atherosclerosis: current status of the problem and
clinical relevance. Eur Heart J 26: 1700 1704, 2005.
1489
130. Berk BC. Atheroprotective signaling mechanisms activated by steady laminar flow in
endothelial cells. Circulation 117: 10821089, 2008.
143. Bianchi K, Meier P. A tangled web of ubiquitin chains: breaking news in TNF-R1
signaling. Mol Cell 36: 736 742, 2009.
PAUL N. HOPKINS
162. Bodary PF, Shen Y, Vargas FB, Bi X, Ostenso KA, Gu S, Shayman JA, Eitzman DT.
Alpha-galactosidase A deficiency accelerates atherosclerosis in mice with apolipoprotein E deficiency. Circulation 111: 629 632, 2005.
179. Boring L, Gosling J, Cleary M, Charo IF. Decreased lesion formation in CCR2/
mice reveals a role for chemokines in the initiation of atherosclerosis. Nature 394:
894 897, 1998.
163. Boesten LS, Zadelaar AS, van Nieuwkoop A, Hu L, Jonkers J, van de Water B, Gijbels
MJ, van der Made I, de Winther MP, Havekes LM, van Vlijmen BJ. Macrophage
retinoblastoma deficiency leads to enhanced atherosclerosis development in ApoEdeficient mice. FASEB J 20: 953955, 2006.
180. Borisov N, Aksamitiene E, Kiyatkin A, Legewie S, Berkhout J, Maiwald T, Kaimachnikov NP, Timmer J, Hoek JB, Kholodenko BN. Systems-level interactions between
insulin-EGF networks amplify mitogenic signaling. Mol Syst Biol 5: 256, 2009.
164. Boesten LS, Zadelaar AS, van Nieuwkoop A, Hu L, Teunisse AF, Jochemsen AG,
Evers B, van de Water B, Gijbels MJ, van Vlijmen BJ, Havekes LM, de Winther MP.
Macrophage p53 controls macrophage death in atherosclerotic lesions of apolipoprotein E deficient mice. Atherosclerosis 207: 399 404, 2009.
165. Bogachev O, Majdalawieh A, Pan X, Zhang L, Ro HS. Adipocyte enhancer-binding
protein 1 (AEBP1) (a novel macrophage proinflammatory mediator) overexpression
promotes and ablation attenuates atherosclerosis in ApoE (/) and LDLR (/)
mice. Mol Med 17: 1056 1064, 2011.
166. Boisvert WA, Rose DM, Boullier A, Quehenberger O, Sydlaske A, Johnson KA,
Curtiss LK, Terkeltaub R. Leukocyte transglutaminase 2 expression limits atherosclerotic lesion size. Arterioscler Thromb Vasc Biol 26: 563569, 2006.
167. Boisvert WA, Rose DM, Johnson KA, Fuentes ME, Lira SA, Curtiss LK, Terkeltaub
RA. Up-regulated expression of the CXCR2 ligand KC/GRO-alpha in atherosclerotic
lesions plays a central role in macrophage accumulation and lesion progression. Am
J Pathol 168: 13851395, 2006.
182. Borissoff JI, Loubele ST, Leenders P, van Oerle R, Hamulyak K, Heeneman S, Daemen MJ, Degen JL, Spronk HM, ten Cate H. Abstract 19565: genetically imposed
hypocoagulability inhibits atherogenesis and promotes plaque stability. Circulation
122: A19565, 2010.
183. Borissoff JI, Spronk HM, ten Cate H. The hemostatic system as a modulator of
atherosclerosis. N Engl J Med 364: 1746 1760, 2011.
184. Borissoff JI, Spronk HMH, Heeneman S, ten Cate H. Is thrombin a key player in the
coagulation-atherogenesis maze? Cardiovasc Res 82: 392 403, 2009.
185. Bostrom KI, Jumabay M, Matveyenko A, Nicholas SB, Yao Y. Activation of vascular
bone morphogenetic protein signaling in diabetes mellitus. Circ Res 108: 446 457,
2011.
186. Bostrom MA, Boyanovsky BB, Jordan CT, Wadsworth MP, Taatjes DJ, de Beer FC,
Webb NR. Group V secretory phospholipase A2 promotes atherosclerosis: evidence
from genetically altered mice. Arterioscler Thromb Vasc Biol 27: 600 606, 2007.
169. Boisvert WA, Spangenberg J, Curtiss LK. Role of leukocyte-specific LDL receptors
on plasma lipoprotein cholesterol and atherosclerosis in mice. Arterioscler Thromb
Vasc Biol 17: 340 347, 1997.
187. Bot I, Bot M, van Heiningen SH, van Santbrink PJ, Lankhuizen IM, Hartman P,
Gruener S, Hilpert H, van Berkel TJ, Fingerle J, Biessen EA. Mast cell chymase
inhibition reduces atherosclerotic plaque progression and improves plaque stability
in ApoE/ mice. Cardiovasc Res 89: 244 252, 2011.
170. Bolick DT, Orr AW, Whetzel A, Srinivasan S, Hatley ME, Schwartz MA, Hedrick CC.
12/15-Lipoxygenase regulates intercellular adhesion molecule-1 expression and
monocyte adhesion to endothelium through activation of RhoA and nuclear factorkappaB. Arterioscler Thromb Vasc Biol 25: 23012307, 2005.
188. Bot M, Bot I, Lopez-Vales R, van de Lest CH, Saulnier-Blache JS, Helms JB, David S,
van Berkel TJ, Biessen EA. Atherosclerotic lesion progression changes lysophosphatidic acid homeostasis to favor its accumulation. Am J Pathol 176: 30733084, 2010.
171. Bolick DT, Skaflen MD, Johnson LE, Kwon SC, Howatt D, Daugherty A, Ravichandran KS, Hedrick CC. G2A deficiency in mice promotes macrophage activation and
atherosclerosis. Circ Res 104: 318 327, 2009.
172. Bolick DT, Srinivasan S, Kim KW, Hatley ME, Clemens JJ, Whetzel A, Ferger N,
Macdonald TL, Davis MD, Tsao PS, Lynch KR, Hedrick CC. Sphingosine-1-phosphate prevents tumor necrosis factor--mediated monocyte adhesion to aortic endothelium in mice. Arterioscler Thromb Vasc Biol 25: 976 981, 2005.
173. Bolick DT, Srinivasan S, Whetzel A, Fuller LC, Hedrick CC. 12/15 Lipoxygenase
mediates monocyte adhesion to aortic endothelium in apolipoprotein E-deficient
mice through activation of RhoA and NF-kappaB. Arterioscler Thromb Vasc Biol 26:
1260 1266, 2006.
174. Bolick DT, Whetzel AM, Skaflen M, Deem TL, Lee J, Hedrick CC. Absence of the G
protein-coupled receptor G2A in mice promotes monocyte/endothelial interactions
in aorta. Circ Res 100: 572580, 2007.
175. Bonta PI, van Tiel CM, Vos M, Pols TW, van Thienen JV, Ferreira V, Arkenbout EK,
Seppen J, Spek CA, van der Poll T, Pannekoek H, de Vries CJ. Nuclear receptors
Nur77, Nurr1, and NOR-1 expressed in atherosclerotic lesion macrophages reduce
lipid loading and inflammatory responses. Arterioscler Thromb Vasc Biol 26: 2288
2294, 2006.
176. Boomer JS, Green JM. An enigmatic tail of CD28 signaling. Cold Spring Harb Perspect
Biol 2: a002436, 2010.
177. Boon RA, Fledderus JO, Volger OL, van Wanrooij EJ, Pardali E, Weesie F, Kuiper J,
Pannekoek H, ten Dijke P, Horrevoets AJ. KLF2 suppresses TGF-beta signaling in
endothelium through induction of Smad7 and inhibition of AP-1. Arterioscler Thromb
Vasc Biol 27: 532539, 2007.
178. Boord JB, Maeda K, Makowski L, Babaev VR, Fazio S, Linton MF, Hotamisligil GS.
Combined adipocyte-macrophage fatty acid-binding protein deficiency improves
metabolism, atherosclerosis, and survival in apolipoprotein E-deficient mice. Circulation 110: 14921498, 2004.
1490
189. Bouallegue A, Pandey NR, Srivastava AK. CaMKII knockdown attenuates H2O2induced phosphorylation of ERK1/2, PKB/Akt, and IGF-1R in vascular smooth muscle
cells. Free Radic Biol Med 47: 858 866, 2009.
190. Bouaziz JD, Yanaba K, Venturi GM, Wang Y, Tisch RM, Poe JC, Tedder TF. Therapeutic B cell depletion impairs adaptive and autoreactive CD4 T cell activation in
mice. Proc Natl Acad Sci USA 104: 20878 20883, 2007.
191. Boucher P, Gotthardt M, Li WP, Anderson RGW, Herz J. LRP: role in vascular wall
integrity and protection from atherosclerosis. Science 300: 329 332, 2003.
192. Boucher P, Herz J. Signaling through LRP1: protection from atherosclerosis and
beyond. Biochem Pharmacol 81: 15, 2011.
193. Boucher P, Li WP, Matz RL, Takayama Y, Auwerx J, Anderson RG, Herz J. LRP1
functions as an atheroprotective integrator of TGFbeta and PDFG signals in the
vascular wall: implications for Marfan syndrome. PLoS ONE 2: e448, 2007.
194. Bouhlel MA, Derudas B, Rigamonti E, Dievart R, Brozek J, Haulon S, Zawadzki C,
Jude B, Torpier G, Marx N, Staels B, Chinetti-Gbaguidi G. PPARgamma activation
primes human monocytes into alternative M2 macrophages with anti-inflammatory
properties. Cell Metab 6: 137143, 2007.
195. Bouis D, McCubbrey AL, Hyman MC, Thompson LF, Pinsky DJ. Abstract P452:
bimodal atherogenic effects of CD73 in apolipoprotein E deficient mice. Arterioscler
Thromb Vasc Biol 28: e115, 2008.
196. Boullier A, Li Y, Quehenberger O, Palinski W, Tabas I, Witztum JL, Miller YI. Minimally oxidized LDL offsets the apoptotic effects of extensively oxidized LDL and free
cholesterol in macrophages. Arterioscler Thromb Vasc Biol 26: 1169 1176, 2006.
197. Bouras T, Fu M, Sauve AA, Wang F, Quong AA, Perkins ND, Hay RT, Gu W, Pestell
RG. SIRT1 deacetylation and repression of p300 involves lysine residues 1020/1024
within the cell cycle regulatory domain 1. J Biol Chem 280: 10264 10276, 2005.
198. Boyanovsky B, Zack M, Forrest K, Webb NR. The capacity of group V sPLA2 to
increase atherogenicity of ApoE/ and LDLR/ mouse LDL in vitro predicts its
atherogenic role in vivo. Arterioscler Thromb Vasc Biol 29: 532538, 2009.
168. Boisvert WA, Santiago R, Curtiss LK, Terkeltaub RA. A leukocyte homologue of the
IL-8 receptor CXCR-2 mediates the accumulation of macrophages in atherosclerotic lesions of LDL receptor-deficient mice. J Clin Invest 101: 353363, 1998.
181. Borissoff JI, Loubele ST, Leenders P, Soehnlein O, Koenen RR, van Oerle R, Hamulyak K, Heeneman S, Daemen MJ, Weiler H, van Ryn J, Spronk HM, ten Cate H.
Abstract 19384: direct thrombin inhibition by dabigatran protects against severe
atherosclerosis progression in prothrombotic mice. Circulation 122: A19384, 2010.
218. Brunelli L, Cieslik KA, Alcorn JL, Vatta M, Baldini A. Peroxisome proliferator-activated receptor-delta upregulates 14-3-3 epsilon in human endothelial cells via
CCAAT/enhancer binding protein-beta. Circ Res 100: e59 71, 2007.
200. Boyle JJ, Harrington HA, Piper E, Elderfield K, Stark J, Landis RC, Haskard DO.
Coronary intraplaque hemorrhage evokes a novel atheroprotective macrophage
phenotype. Am J Pathol 174: 10971108, 2009.
219. Brunham LR, Kruit JK, Pape TD, Parks JS, Kuipers F, Hayden MR. Tissue-specific
induction of intestinal ABCA1 expression with a liver X receptor agonist raises
plasma HDL cholesterol levels. Circ Res 99: 672 674, 2006.
201. Boyle JJ, Johns M, Kampfer T, Nguyen AT, Game L, Schaer DJ, Mason JC, Haskard
DO. Activating transcription factor 1 directs Mhem atheroprotective macrophages
through coordinated iron handling and foam cell protection. Circ Res 110: 20 33,
2012.
220. Brunham LR, Singaraja RR, Duong M, Timmins JM, Fievet C, Bissada N, Kang MH,
Samra A, Fruchart JC, McManus B, Staels B, Parks JS, Hayden MR. Tissue-specific
roles of ABCA1 influence susceptibility to atherosclerosis. Arterioscler Thromb Vasc
Biol 29: 548 554, 2009.
203. Brasacchio D, Okabe J, Tikellis C, Balcerczyk A, George P, Baker EK, Calkin AC,
Brownlee M, Cooper ME, El-Osta A. Hyperglycemia induces a dynamic cooperativity of histone methylase and demethylase enzymes associated with gene-activating
epigenetic marks that coexist on the lysine tail. Diabetes 58: 1229 1236, 2009.
222. Budoff M. Aged garlic extract retards progression of coronary artery calcification. J
Nutr 136: 741S744, 2006.
204. Braun A, Trigatti BL, Post MJ, Sato K, Simons M, Edelberg JM, Rosenberg RD,
Schrenzel M, Krieger M. Loss of SR-BI expression leads to the early onset of occlusive atherosclerotic coronary artery disease, spontaneous myocardial infarctions,
severe cardiac dysfunction, and premature death in apolipoprotein E-deficient mice.
Circ Res 90: 270 276, 2002.
206. Brennan JP, Bardswell SC, Burgoyne JR, Fuller W, Schroder E, Wait R, Begum S,
Kentish JC, Eaton P. Oxidant-induced activation of type I protein kinase A is mediated by RI subunit interprotein disulfide bond formation. J Biol Chem 281: 21827
21836, 2006.
207. Brennan ML, Anderson MM, Shih DM, Qu XD, Wang X, Mehta AC, Lim LL, Shi W,
Hazen SL, Jacob JS, Crowley JR, Heinecke JW, Lusis AJ. Increased atherosclerosis in
myeloperoxidase-deficient mice. J Clin Invest 107: 419 430, 2001.
208. Brigelius-Flohe R, Flohe L. Basic principles and emerging concepts in the redox
control of transcription factors. Antioxid Redox Signal 15: 23352381, 2011.
209. Brinkmann V, Zychlinsky A. Beneficial suicide: why neutrophils die to make NETs.
Nat Rev Microbiol 5: 577582, 2007.
210. Brodsky SV, Gealekman O, Chen J, Zhang F, Togashi N, Crabtree M, Gross SS,
Nasjletti A, Goligorsky MS. Prevention and reversal of premature endothelial cell
senescence and vasculopathy in obesity-induced diabetes by ebselen. Circ Res 94:
377384, 2004.
211. Brookes PS, Yoon Y, Robotham JL, Anders MW, Sheu SS. Calcium, ATP, and ROS: a
mitochondrial love-hate triangle. Am J Physiol Cell Physiol 287: C817C833, 2004.
212. Brooks AR, Lelkes PI, Rubanyi GM. Gene expression profiling of human aortic endothelial cells exposed to disturbed flow and steady laminar flow. Physiol Genomics 9:
27 41, 2002.
224. Budoff MJ, McClelland RL, Nasir K, Greenland P, Kronmal RA, Kondos GT, Shea S,
Lima JA, Blumenthal RS. Cardiovascular events with absent or minimal coronary
calcification: the Multi-Ethnic Study of Atherosclerosis (MESA). Am Heart J 158:
554 561, 2009.
225. Budoff MJ, Shaw LJ, Liu ST, Weinstein SR, Mosler TP, Tseng PH, Flores FR, Callister
TQ, Raggi P, Berman DS. Long-term prognosis associated with coronary calcification: observations from a registry of 25,253 patients. J Am Coll Cardiol 49: 1860
1870, 2007.
226. Buja LM, Kovanen PT, Bilheimer DW. Cellular pathology of homozygous familial
hypercholesterolemia. Am J Pathol 97: 327357, 1979.
227. Bultmann A, Li Z, Wagner S, Peluso M, Schonberger T, Weis C, Konrad I, Stellos K,
Massberg S, Nieswandt B, Gawaz M, Ungerer M, Munch G. Impact of glycoprotein
VI and platelet adhesion on atherosclerosisa possible role of fibronectin. J Mol Cell
Cardiol 49: 532542, 2010.
228. Buono C, Binder CJ, Stavrakis G, Witztum JL, Glimcher LH, Lichtman AH. T-bet
deficiency reduces atherosclerosis and alters plaque antigen-specific immune responses. Proc Natl Acad Sci USA 102: 1596 1601, 2005.
229. Buono C, Come CE, Witztum JL, Maguire GF, Connelly PW, Carroll M, Lichtman
AH. Influence of C3 deficiency on atherosclerosis. Circulation 105: 30253031, 2002.
230. Buono C, Pang H, Uchida Y, Libby P, Sharpe AH, Lichtman AH. B71/B72 costimulation regulates plaque antigen-specific T-cell responses and atherogenesis in lowdensity lipoprotein receptor-deficient mice. Circulation 109: 2009 2015, 2004.
231. Burbach BJ, Medeiros RB, Mueller KL, Shimizu Y. T-cell receptor signaling to integrins. Immunol Rev 218: 65 81, 2007.
232. Burger PC, Wagner DD. Platelet P-selectin facilitates atherosclerotic lesion development. Blood 101: 26612666, 2003.
213. Brown J, Wallet MA, Krastins B, Sarracino D, Goodenow MM. Proteome bioprofiles
distinguish between M1 priming and activation states in human macrophages. J
Leukoc Biol 87: 655 662, 2010.
233. Burke AP, Kolodgie FD, Farb A, Weber DK, Malcom GT, Smialek J, Virmani R.
Healed plaque ruptures and sudden coronary death: evidence that subclinical rupture has a role in plaque progression. Circulation 103: 934 940, 2001.
214. Brown MS, Goldstein JL. Lipoprotein metabolism in the macrophage: implications
for cholesterol deposition in atherosclerosis. Annu Rev Biochem 52: 223261, 1983.
234. Burke JE, Dennis EA. Phospholipase A2 biochemistry. Cardiovasc Drugs Ther 23:
49 59, 2009.
236. Bustos M, Coffman TM, Saadi S, Platt JL. Modulation of eicosanoid metabolism in
endothelial cells in a xenograft model. Role of cyclooxygenase-2. J Clin Invest 100:
1150 1158, 1997.
237. Caesar R, Fak F, Backhed F. Effects of gut microbiota on obesity and atherosclerosis
via modulation of inflammation and lipid metabolism. J Intern Med 268: 320 328,
2010.
1491
223. Budoff MJ, Ahmadi N, Gul KM, Liu ST, Flores FR, Tiano J, Takasu J, Miller E, Tsimikas
S. Aged garlic extract supplemented with B vitamins, folic acid and L-arginine retards
the progression of subclinical atherosclerosis: a randomized clinical trial. Prev Med
49: 101107, 2009.
PAUL N. HOPKINS
238. Cai W, He JC, Zhu L, Lu C, Vlassara H. Advanced glycation end product (AGE)
receptor 1 suppresses cell oxidant stress and activation signaling via EGF receptor.
Proc Natl Acad Sci USA 103: 1380113806, 2006.
257. Capettini LS, Cortes SF, Silva JF, Alvarez-Leite JI, Lemos VS. Decreased production
of neuronal NOS-derived hydrogen peroxide contributes to endothelial dysfunction
in atherosclerosis. Br J Pharmacol 164: 1738 1748, 2011.
239. Cai W, Torreggiani M, Zhu L, Chen X, He JC, Striker GE, Vlassara H. AGER1
regulates endothelial cell NADPH oxidase-dependent oxidant stress via PKC-:
implications for vascular disease. Am J Physiol Cell Physiol 298: C624 C634, 2010.
260. Carmena R, Roy M, Roederer G, Minnich A, Davignon J. Coexisting dysbetalipoproteinemia and familial hypercholesterolemia. Clin Lab Observations Atherosclerosis 148:
113124, 2000.
242. Caligiuri G, Levy B, Pernow J, Thorn P, Hansson GK. Myocardial infarction mediated by endothelin receptor signaling in hypercholesterolemic mice. Proc Natl Acad
Sci USA 96: 6920 6924, 1999.
261. Carmona G, Gottig S, Orlandi A, Scheele J, Bauerle T, Jugold M, Kiessling F, Henschler R, Zeiher AM, Dimmeler S, Chavakis E. Role of the small GTPase Rap1 for
integrin activity regulation in endothelial cells and angiogenesis. Blood 113: 488 497,
2009.
243. Calkin AC, Tontonoz P. Liver X receptor signaling pathways and atherosclerosis.
Arterioscler Thromb Vasc Biol 30: 15131518, 2010.
244. Callegari A, Liu Y, White CC, Chait A, Gough P, Raines EW, Cox D, Kavanagh TJ,
Rosenfeld ME. Gain and loss of function for glutathione synthesis: impact on advanced atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc
Biol 31: 24732482, 2011.
263. Cassis LA, Rateri DL, Lu H, Daugherty A. Bone marrow transplantation reveals that
recipient AT1a receptors are required to initiate angiotensin II-induced atherosclerosis and aneurysms. Arterioscler Thromb Vasc Biol 27: 380 386, 2007.
264. Castellani LW, Navab M, Van Lenten BJ, Hedrick CC, Hama SY, Goto AM, Fogelman
AM, Lusis AJ. Overexpression of apolipoprotein AII in transgenic mice converts high
density lipoproteins to proinflammatory particles. J Clin Invest 100: 464 474, 1997.
265. Castellino FJ, Ganopolsky JG, Noria F, Sandoval-Cooper MJ, Ploplis VA. Focal arterial
inflammation is augmented in mice with a deficiency of the protein C gene. Thromb
Haemost 96: 794 801, 2006.
247. Campbell JH, Efendy JL, Smith NJ, Campbell GR. Molecular basis by which garlic
suppresses atherosclerosis. J Nutr 131: 1006S1009S, 2001.
266. Celletti FL, Waugh JM, Amabile PG, Brendolan A, Hilfiker PR, Dake MD. Vascular
endothelial growth factor enhances atherosclerotic plaque progression. Nat Med 7:
425 429, 2001.
248. Cancel LM, Fitting A, Tarbell JM. In vitro study of LDL transport under pressurized
(convective) conditions. Am J Physiol Heart Circ Physiol 293: H126 H132, 2007.
249. Cannon CP, Braunwald E, McCabe CH, Grayston JT, Muhlestein B, Giugliano RP,
Cairns R, Skene AM. Antibiotic treatment of Chlamydia pneumoniae after acute
coronary syndrome. N Engl J Med 352: 1646 1654, 2005.
250. Cannon CP, Shah S, Dansky HM, Davidson M, Brinton EA, Gotto AM, Stepanavage
M, Liu SX, Gibbons P, Ashraf TB, Zafarino J, Mitchel Y, Barter P. Safety of anacetrapib in patients with or at high risk for coronary heart disease. N Engl J Med 363:
2406 2415, 2010.
251. Canturk NZ, Canturk Z, Okay E, Yirmibesoglu O, Eraldemir B. Risk of nosocomial
infections and effects of total cholesterol, HDL cholesterol in surgical patients. Clin
Nutr 21: 431 436, 2002.
252. Cao M, Khan JA, Kang BY, Mehta JL, Hermonat PL. Dual AAV/IL-10 plus STAT3
anti-inflammatory gene delivery lowers atherosclerosis in LDLR KO mice, but without increased benefit. Int J Vasc Med 2012: 524235, 2012.
253. Cao RY, St Amand T, Grabner R, Habenicht AJ, Funk CD. Genetic and pharmacological inhibition of the 5-lipoxygenase/leukotriene pathway in atherosclerotic lesion
development in ApoE deficient mice. Atherosclerosis 203: 395 400, 2009.
254. Capell BC, Collins FS, Nabel EG. Mechanisms of cardiovascular disease in accelerated aging syndromes. Circ Res 101: 1326, 2007.
255. Capettini LS, Cortes SF, Gomes MA, Silva GA, Pesquero JL, Lopes MJ, Teixeira MM,
Lemos VS. Neuronal nitric oxide synthase-derived hydrogen peroxide is a major
endothelium-dependent relaxing factor. Am J Physiol Heart Circ Physiol 295: H2503
H2511, 2008.
256. Capettini LS, Cortes SF, Lemos VS. Relative contribution of eNOS and nNOS to
endothelium-dependent vasodilation in the mouse aorta. Eur J Pharmacol 643: 260
266, 2010.
1492
267. Chachisvilis M, Zhang YL, Frangos JA. G protein-coupled receptors sense fluid shear
stress in endothelial cells. Proc Natl Acad Sci USA 103: 1546315468, 2006.
268. Chadjichristos CE, Scheckenbach KE, van Veen TA, Richani Sarieddine MZ, de Wit
C, Yang Z, Roth I, Bacchetta M, Viswambharan H, Foglia B, Dudez T, van Kempen
MJ, Coenjaerts FE, Miquerol L, Deutsch U, Jongsma HJ, Chanson M, Kwak BR.
Endothelial-specific deletion of connexin40 promotes atherosclerosis by increasing
CD73-dependent leukocyte adhesion. Circulation 121: 123131, 2010.
269. Chae HZ, Oubrahim H, Park JW, Rhee SG, Chock PB. Protein glutathionylation in
the regulation of peroxiredoxins: a family of thiol-specific peroxidases that function
as antioxidants, molecular chaperones, and signal modulators. Antioxid Redox Signal
16: 506 523, 2012.
270. Chamberlain J, Francis S, Brookes Z, Shaw G, Graham D, Alp NJ, Dower S, Crossman DC. Interleukin-1 regulates multiple atherogenic mechanisms in response to fat
feeding. PLoS ONE 4: e5073, 2009.
271. Chambliss KL, Wu Q, Oltmann S, Konaniah ES, Umetani M, Korach KS, Thomas
GD, Mineo C, Yuhanna IS, Kim SH, Madak-Erdogan Z, Maggi A, Dineen SP,
Roland CL, Hui DY, Brekken RA, Katzenellenbogen JA, Katzenellenbogen BS,
Shaul PW. Non-nuclear estrogen receptor alpha signaling promotes cardiovascular protection but not uterine or breast cancer growth in mice. J Clin Invest
120: 2319 2330, 2010.
272. Chan MM, Moore AR. Resolution of inflammation in murine autoimmune arthritis is
disrupted by cyclooxygenase-2 inhibition and restored by prostaglandin E2-mediated lipoxin A4 production. J Immunol 184: 6418 6426, 2010.
273. Chandak PG, Obrowsky S, Radovic B, Doddapattar P, Aflaki E, Kratzer A, Doshi LS,
Povoden S, Ahammer H, Hoefler G, Levak-Frank S, Kratky D. Lack of acyl-CoA:
diacylglycerol acyltransferase 1 reduces intestinal cholesterol absorption and attenuates atherosclerosis in apolipoprotein E knockout mice. Biochim Biophys Acta 1811:
10111020, 2011.
291. Chen CY, Ching LC, Liao YJ, Yu YB, Tsou CY, Shyue SK, Chen YM, Lee TS. Deficiency of glycine N-methyltransferase aggravates atherosclerosis in apolipoprotein
E-null mice. Mol Med 2012.
275. Chandrasekharan UM, Mavrakis L, Bonfield TL, Smith JD, DiCorleto PE. Decreased
atherosclerosis in mice deficient in tumor necrosis factor-alpha receptor-II (p75).
Arterioscler Thromb Vasc Biol 27: e16 17, 2007.
292. Chen L, Zhang JJ, Huang XY. cAMP inhibits cell migration by interfering with Racinduced lamellipodium formation. J Biol Chem 283: 13799 13805, 2008.
1493
293. Chen S, Shimada K, Zhang W, Huang G, Crother TR, Arditi M. IL-17A is proatherogenic in high-fat diet-induced and Chlamydia pneumoniae infection-accelerated atherosclerosis in mice. J Immunol 185: 5619 5627, 2010.
PAUL N. HOPKINS
311. Chiu JJ, Usami S, Chien S. Vascular endothelial responses to altered shear stress:
pathologic implications for atherosclerosis. Ann Med 41: 19 28, 2009.
312. Chiu TT, Jensen TE, Sylow L, Richter EA, Klip A. Rac1 signalling towards GLUT4/
glucose uptake in skeletal muscle. Cell Signal 23: 1546 1554, 2011.
313. Chiu YJ, McBeath E, Fujiwara K. Mechanotransduction in an extracted cell model:
Fyn drives stretch- and flow-elicited PECAM-1 phosphorylation. J Cell Biol 182:
753763, 2008.
314. Choi JH, Cheong C, Dandamudi DB, Park CG, Rodriguez A, Mehandru S, Velinzon
K, Jung IH, Yoo JY, Oh GT, Steinman RM. Flt3 signaling-dependent dendritic cells
protect against atherosclerosis. Immunity 35: 819 831, 2011.
315. Choi JH, Park JG, Jeon HJ, Kim MS, Lee MR, Lee MN, Sonn S, Kim JH, Lee MH, Choi
MS, Park YB, Kwon OS, Jeong TS, Lee WS, Shim HB, Shin DH, Oh GT. 5-(4Hydroxy-2,3,5-trimethylbenzylidene) thiazolidine-2,4-dione attenuates atherosclerosis possibly by reducing monocyte recruitment to the lesion. Exp Mol Med 43:
471 478, 2011.
316. Choi JW, Herr DR, Noguchi K, Yung YC, Lee CW, Mutoh T, Lin ME, Teo ST, Park
KE, Mosley AN, Chun J. LPA receptors: subtypes and biological actions. Annu Rev
Pharmacol Toxicol 50: 157186, 2010.
330. Clark PR, Jensen TJ, Kluger MS, Morelock M, Hanidu A, Qi Z, Tatake RJ, Pober JS.
MEK5 is activated by shear stress, activates ERK5 and induces KLF4 to modulate
TNF responses in human dermal microvascular endothelial cells. Microcirculation 18:
102117, 2011.
331. Clarke MC, Littlewood TD, Figg N, Maguire JJ, Davenport AP, Goddard M, Bennett
MR. Chronic apoptosis of vascular smooth muscle cells accelerates atherosclerosis
and promotes calcification and medial degeneration. Circ Res 102: 1529 1538, 2008.
332. Clarke MC, Talib S, Figg NL, Bennett MR. Vascular smooth muscle cell apoptosis
induces interleukin-1-directed inflammation: effects of hyperlipidemia-mediated inhibition of phagocytosis. Circ Res 106: 363372, 2010.
333. Clarke R, Halsey J, Lewington S, Lonn E, Armitage J, Manson JE, Bonaa KH, Spence
JD, Nygard O, Jamison R, Gaziano JM, Guarino P, Bennett D, Mir F, Peto R, Collins
R. Effects of lowering homocysteine levels with B vitamins on cardiovascular disease,
cancer, and cause-specific mortality: meta-analysis of 8 randomized trials involving
37 485 individuals. Arch Intern Med 170: 16221631, 2010.
334. Clausell N, Rabinovitch M. Upregulation of fibronectin synthesis by interleukin-1
beta in coronary artery smooth muscle cells is associated with the development of
the post-cardiac transplant arteriopathy in piglets. J Clin Invest 92: 1850 1858, 1993.
335. Clifford PS. Local control of blood flow. Adv Physiol Educ 35: 515, 2011.
317. Choi NM, Majumder P, Boss JM. Regulation of major histocompatibility complex
class II genes. Curr Opin Immunol 23: 81 87, 2011.
319. Choi SH, Wiesner P, Almazan F, Kim J, Miller YI. Spleen tyrosine kinase regulates
AP-1 dependent transcriptional response to minimally oxidized LDL. PLoS ONE 7:
e32378, 2012.
337. Coffa S, Breitman M, Hanson SM, Callaway K, Kook S, Dalby KN, Gurevich VV. The
effect of arrestin conformation on the recruitment of c-Raf1, MEK1, and ERK1/2
activation. PLoS ONE 6: e28723, 2011.
320. Chow BJ, Small G, Yam Y, Chen L, Achenbach S, Al-Mallah M, Berman DS, Budoff
MJ, Cademartiri F, Callister TQ, Chang HJ, Cheng V, Chinnaiyan KM, Delago A,
Dunning A, Hadamitzky M, Hausleiter J, Kaufmann P, Lin F, Maffei E, Raff GL, Shaw
LJ, Villines TC, Min JK. Incremental prognostic value of cardiac computed tomography in coronary artery disease using CONFIRM: COroNary computed tomography
angiography evaluation for clinical outcomes: an InteRnational Multicenter registry.
Circ Cardiovasc Imaging 4: 463 472, 2011.
338. Cole BK, Kuhn NS, Green-Mitchell SM, Leone KA, Raab RM, Nadler JL, Chakrabarti
SK. 12/15-Lipoxygenase signaling in the endoplasmic reticulum stress response. Am
J Physiol Endocrinol Metab 302: E654 E665, 2012.
340. Coller BS. Leukocytosis and ischemic vascular disease morbidity and mortality: is it
time to intervene? Arterioscler Thromb Vasc Biol 25: 658 670, 2005.
339. Cole JE, Navin TJ, Cross AJ, Goddard ME, Alexopoulou L, Mitra AT, Davies AH,
Flavell RA, Feldmann M, Monaco C. Unexpected protective role for Toll-like receptor 3 in the arterial wall. Proc Natl Acad Sci USA 108: 23722377, 2011.
341. Collins AR, Lyon CJ, Xia X, Liu JZ, Tangirala RK, Yin F, Boyadjian R, Bikineyeva A,
Pratico D, Harrison DG, Hsueh WA. Age-accelerated atherosclerosis correlates
with failure to upregulate antioxidant genes. Circ Res 104: e4254, 2009.
342. Collins M, Bartelt RR, Houtman JC. T cell receptor activation leads to two distinct
phases of Pyk2 activation and actin cytoskeletal rearrangement in human T cells. Mol
Immunol 47: 16651674, 2010.
343. Collins RG, Velji R, Guevara NV, Hicks MJ, Chan L, Beaudet AL. P-Selectin or
intercellular adhesion molecule (ICAM)-1 deficiency substantially protects against
atherosclerosis in apolipoprotein E-deficient mice. J Exp Med 191: 189 194, 2000.
344. Combadiere C, Potteaux S, Gao JL, Esposito B, Casanova S, Lee EJ, Debre P, Tedgui
A, Murphy PM, Mallat Z. Decreased atherosclerotic lesion formation in CX3CR1/
apolipoprotein E double knockout mice. Circulation 107: 1009 1016, 2003.
345. Combadiere C, Potteaux S, Rodero M, Simon T, Pezard A, Esposito B, Merval R,
Proudfoot A, Tedgui A, Mallat Z. Combined inhibition of CCL2, CX3CR1, and CCR5
abrogates Ly6C(hi) and Ly6C(lo) monocytosis and almost abolishes atherosclerosis
in hypercholesterolemic mice. Circulation 117: 1649 1657, 2008.
346. Conde-Knape K, Bensadoun A, Sobel JH, Cohn JS, Shachter NS. Overexpression of
apoC-I in apoE-null mice: severe hypertriglyceridemia due to inhibition of hepatic
lipase. J Lipid Res 43: 2136 2145, 2002.
328. Citri A, Yarden Y. EGF-ERBB signalling: towards the systems level. Nat Rev Mol Cell
Biol 7: 505516, 2006.
329. Civelek M, Manduchi E, Riley RJ, Stoeckert CJ Jr, Davies PF. Chronic endoplasmic
reticulum stress activates unfolded protein response in arterial endothelium in regions of susceptibility to atherosclerosis. Circ Res 105: 453 461, 2009.
1494
318. Choi SH, Harkewicz R, Lee JH, Boullier A, Almazan F, Li AC, Witztum JL, Bae YS,
Miller YI. Lipoprotein accumulation in macrophages via toll-like receptor-4-dependent fluid phase uptake. Circ Res 104: 13551363, 2009.
monocyte chemotactic protein 1 in human endothelial cells and smooth muscle cells.
Proc Natl Acad Sci USA 87: 5134 5138, 1990.
366. Cybulsky MI, Iiyama K, Li H, Zhu S, Chen M, Iiyama M, Davis V, Gutierrez-Ramos JC,
Connelly PW, Milstone DS. A major role for VCAM-1, but not ICAM-1, in early
atherosclerosis. J Clin Invest 107: 12551262, 2001.
350. Conover CA, Mason MA, Bale LK, Harrington SC, Nyegaard M, Oxvig C, Overgaard
MT. Transgenic overexpression of pregnancy-associated plasma protein-A in murine
arterial smooth muscle accelerates atherosclerotic lesion development. Am J Physiol
Heart Circ Physiol 299: H284 H291, 2010.
351. Constantinides P. Cause of thrombosis in human atherosclerotic arteries. Am J Cardiol 66: 37G 40G, 1990.
352. Costanzo M, Baryshnikova A, Bellay J, Kim Y, Spear ED, Sevier CS, Ding H, Koh JL,
Toufighi K, Mostafavi S, Prinz J, St Onge RP, VanderSluis B, Makhnevych T, Vizeacoumar FJ, Alizadeh S, Bahr S, Brost RL, Chen Y, Cokol M, Deshpande R, Li Z, Lin
ZY, Liang W, Marback M, Paw J, San Luis BJ, Shuteriqi E, Tong AH, van Dyk N,
Wallace IM, Whitney JA, Weirauch MT, Zhong G, Zhu H, Houry WA, Brudno M,
Ragibizadeh S, Papp B, Pal C, Roth FP, Giaever G, Nislow C, Troyanskaya OG,
Bussey H, Bader GD, Gingras AC, Morris QD, Kim PM, Kaiser CA, Myers CL,
Andrews BJ, Boone C. The genetic landscape of a cell. Science 327: 425 431, 2010.
353. Covey SD, Krieger M, Wang W, Penman M, Trigatti BL. Scavenger receptor class B
type I-mediated protection against atherosclerosis in LDL receptor-negative mice
involves its expression in bone marrow-derived cells. Arterioscler Thromb Vasc Biol
23: 1589 1594, 2003.
355. Criollo A, Senovilla L, Authier H, Maiuri MC, Morselli E, Vitale I, Kepp O, Tasdemir
E, Galluzzi L, Shen S, Tailler M, Delahaye N, Tesniere A, De Stefano D, Younes AB,
Harper F, Pierron G, Lavandero S, Zitvogel L, Israel A, Baud V, Kroemer G. The IKK
complex contributes to the induction of autophagy. EMBO J 29: 619 631, 2010.
356. Croce K, Gao H, Wang Y, Mooroka T, Sakuma M, Shi C, Sukhova GK, Packard RR,
Hogg N, Libby P, Simon DI. Myeloid-related protein-8/14 is critical for the biological
response to vascular injury. Circulation 120: 427 436, 2009.
357. Cruz D, Watson AD, Miller CS, Montoya D, Ochoa MT, Sieling PA, Gutierrez MA,
Navab M, Reddy ST, Witztum JL, Fogelman AM, Rea TH, Eisenberg D, Berliner J,
Modlin RL. Host-derived oxidized phospholipids and HDL regulate innate immunity
in human leprosy. J Clin Invest 118: 29172928, 2008.
358. Csiszar A, Lehoux S, Ungvari Z. Hemodynamic forces, vascular oxidative stress, and
regulation of BMP-2/4 expression. Antioxid Redox Signal 11: 16831697, 2009.
359. Csiszar A, Smith KE, Koller A, Kaley G, Edwards JG, Ungvari Z. Regulation of bone
morphogenetic protein-2 expression in endothelial cells: role of nuclear factorkappaB activation by tumor necrosis factor-alpha, H2O2, and high intravascular pressure. Circulation 111: 2364 2372, 2005.
360. Cuff CA, Kothapalli D, Azonobi I, Chun S, Zhang Y, Belkin R, Yeh C, Secreto A,
Assoian RK, Rader DJ, Pure E. The adhesion receptor CD44 promotes atherosclerosis by mediating inflammatory cell recruitment and vascular cell activation. J Clin
Invest 108: 10311040, 2001.
361. Cuhlmann S, Van der Heiden K, Saliba D, Tremoleda JL, Khalil M, Zakkar M, Chaudhury H, Luong le A, Mason JC, Udalova I, Gsell W, Jones H, Haskard DO, Krams R,
Evans PC. Disturbed blood flow induces RelA expression via c-Jun N-terminal kinase
1: a novel mode of NF-kappaB regulation that promotes arterial inflammation. Circ
Res 108: 950 959, 2011.
362. Curmi PA, Juan L, Tedgui A. Effect of transmural pressure on low density lipoprotein
and albumin transport and distribution across the intact arterial wall. Circ Res 66:
16921702, 1990.
363. Curtiss LK, Black AS, Bonnet DJ, Tobias PS. Atherosclerosis induced by endogenous
and exogenous toll-like receptor (TLR)1 or TLR6 agonists. J Lipid Res 53: 2126
2132, 2012.
368. Cyrus T, Witztum JL, Rader DJ, Tangirala R, Fazio S, Linton MF, Funk CD. Disruption
of the 12/15-lipoxygenase gene diminishes atherosclerosis in ApoE-deficient mice. J
Clin Invest 103: 15971604, 1999.
369. Czymai T, Viemann D, Sticht C, Molema G, Goebeler M, Schmidt M. FOXO3
modulates endothelial gene expression and function by classical and alternative
mechanisms. J Biol Chem 285: 1016310178, 2010.
370. DAlessio A, Kluger MS, Li JH, Al-Lamki R, Bradley JR, Pober JS. Targeting of tumor
necrosis factor receptor 1 to low density plasma membrane domains in human
endothelial cells. J Biol Chem 285: 23868 23879, 2010.
371. DAlessio F, Hentze MW, Muckenthaler MU. The hemochromatosis proteins HFE,
TfR2, and HJV form a membrane-associated protein complex for hepcidin regulation. J Hepatol 57: 10521060, 2012.
372. da Silva CG, Specht A, Wegiel B, Ferran C, Kaczmarek E. Mechanism of purinergic
activation of endothelial nitric oxide synthase in endothelial cells. Circulation 119:
871 879, 2009.
373. Dahabreh IJ, Paulus JK. Association of episodic physical and sexual activity with
triggering of acute cardiac events: systematic review and meta-analysis. JAMA 305:
12251233, 2011.
374. Dahl M, Tybjaerg-Hansen A, Schnohr P, Nordestgaard BG. A population-based
study of morbidity and mortality in mannose-binding lectin deficiency. J Exp Med 199:
13911399, 2004.
375. Dai G, Kaazempur-Mofrad MR, Natarajan S, Zhang Y, Vaughn S, Blackman BR,
Kamm RD, Garcia-Cardena G, Gimbrone MA Jr. Distinct endothelial phenotypes
evoked by arterial waveforms derived from atherosclerosis-susceptible and -resistant regions of human vasculature. Proc Natl Acad Sci USA 101: 1487114876, 2004.
376. Daissormont IT, Christ A, Temmerman L, Sampedro Millares S, Seijkens T, Manca
M, Rousch M, Poggi M, Boon L, van der Loos C, Daemen M, Lutgens E, Halvorsen B,
Aukrust P, Janssen E, Biessen EA. Plasmacytoid dendritic cells protect against atherosclerosis by tuning T-cell proliferation and activity. Circ Res 109: 13871395,
2011.
377. Dancu MB, Tarbell JM. Coronary endothelium expresses a pathologic gene pattern
compared to aortic endothelium: correlation of asynchronous hemodynamics and
pathology in vivo. Atherosclerosis 192: 9 14, 2007.
378. Danesh J, Collins R, Appleby P, Peto R. Association of fibrinogen, C-reactive protein,
albumin, or leukocyte count with coronary heart disease: meta-analyses of prospective studies. JAMA 279: 14771482, 1998.
379. Dansky HM, Charlton SA, Harper MM, Smith JD. T and B lymphocytes play a minor
role in atherosclerotic plaque formation in the apolipoprotein E-deficient mouse.
Proc Natl Acad Sci USA 94: 4642 4646, 1997.
380. Danzaki K, Matsui Y, Ikesue M, Ohta D, Ito K, Kanayama M, Kurotaki D, Morimoto
J, Iwakura Y, Yagita H, Tsutsui H, Uede T. Interleukin-17A deficiency accelerates
unstable atherosclerotic plaque formation in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 32: 273280, 2012.
381. Dastani Z, Dangoisse C, Boucher B, Desbiens K, Krimbou L, Dufour R, Hegele RA,
Pajukanta P, Engert JC, Genest J, Marcil M. A novel nonsense apolipoprotein A-I
mutation (apoA-I(E136X)) causes low HDL cholesterol in French Canadians. Atherosclerosis 185: 127136, 2006.
364. Curtiss LK, Valenta DT, Hime NJ, Rye KA. What is so special about apolipoprotein AI
in reverse cholesterol transport? Arterioscler Thromb Vasc Biol 26: 1219, 2006.
365. Cushing SD, Berliner JA, Valente AJ, Territo MC, Navab M, Parhami F, Gerrity R,
Schwartz CJ, Fogelman AM. Minimally modified low density lipoprotein induces
1495
354. Cozen AE, Moriwaki H, Kremen M, DeYoung MB, Dichek HL, Slezicki KI, Young SG,
Veniant M, Dichek DA. Macrophage-targeted overexpression of urokinase causes
accelerated atherosclerosis, coronary artery occlusions, and premature death. Circulation 109: 2129 2135, 2004.
PAUL N. HOPKINS
384. Daugherty A, Rateri DL, Lu H, Inagami T, Cassis LA. Hypercholesterolemia stimulates angiotensin peptide synthesis and contributes to atherosclerosis through the
AT1A receptor. Circulation 110: 3849 3857, 2004.
385. Davies DF, Davies JR, Richards MA. Antibodies to reconstituted dried cows milk
protein in coronary heart disease. J Atheroscler Res 9: 103107, 1969.
386. Davies MJ, Thomas AC. Plaque fissuringthe cause of acute myocardial infarction,
sudden ischaemic death, and crescendo angina. Br Heart J 53: 363373, 1985.
387. Davies PF. Endothelial transcriptome profiles in vivo in complex arterial flow fields.
Ann Biomed Eng 36: 563570, 2008.
388. Davies PF. Hemodynamic shear stress and the endothelium in cardiovascular pathophysiology. Nat Clin Pract Cardiovasc Med 6: 16 26, 2009.
404. Dekker RJ, van Thienen JV, Rohlena J, de Jager SC, Elderkamp YW, Seppen J, de Vries
CJ, Biessen EA, van Berkel TJ, Pannekoek H, Horrevoets AJ. Endothelial KLF2 links
local arterial shear stress levels to the expression of vascular tone-regulating genes.
Am J Pathol 167: 609 618, 2005.
405. Delekta PC, Apel IJ, Gu S, Siu K, Hattori Y, McAllister-Lucas LM, Lucas PC.
Thrombin-dependent NF-B activation and monocyte/endothelial adhesion are
mediated by the CARMA3-Bcl10-MALT1 signalosome. J Biol Chem 285: 41432
41442, 2010.
406. Delgado-Rodriguez M, Medina-Cuadros M, Martinez-Gallego G, Sillero-Arenas M.
Total cholesterol, HDL-cholesterol, and risk of nosocomial infection: a prospective
study in surgical patients Infection control and hospital epidemiology. Official J Soc
Hosp Epidemiol Am 18: 9 18, 1997.
389. Davis ME, Grumbach IM, Fukai T, Cutchins A, Harrison DG. Shear stress regulates
endothelial nitric-oxide synthase promoter activity through nuclear factor kappaB
binding. J Biol Chem 279: 163168, 2004.
407. Deliri H, Meller N, Kadakkal A, Malhotra R, Brewster J, Doran AC, Pei H, Oldham
SN, Skaflen MD, Garmey JC, McNamara CA. Increased 12/15-lipoxygenase enhances cell growth, fibronectin deposition, and neointimal formation in response to
carotid injury. Arterioscler Thromb Vasc Biol 31: 110 116, 2011.
390. Dawson TC, Kuziel WA, Osahar TA, Maeda N. Absence of CC chemokine receptor-2 reduces atherosclerosis in apolipoprotein E-deficient mice. Atherosclerosis 143:
205211, 1999.
408. Dellas C, Schremmer C, Hasenfuss G, Konstantinides SV, Schafer K. Lack of urokinase plasminogen activator promotes progression and instability of atherosclerotic
lesions in apolipoprotein E-knockout mice. Thromb Haemost 98: 220 227, 2007.
391. De Caterina R. n-3 Fatty acids in cardiovascular disease. N Engl J Med 364: 2439
2450, 2011.
409. Delsing DJ, Leijten FP, Arts K, van Eenennaam H, Garritsen A, Gijbels MJ, de Winther
MP, van Elsas A. Cannabinoid Receptor 2 deficiency in haematopoietic cells aggravates early atherosclerosis in LDL receptor deficient mice. Open Cardiovasc Med J 5:
1521, 2011.
393. De Jager SC, Bot I, Kraaijeveld AO, Korporaal SJ, Bot M, van Santbrink PJ, van Berkel
TJ, Kuiper J, Biessen EA. Leukocyte-specific CCL3 deficiency inhibits atherosclerotic
lesion development by affecting neutrophil accumulation. Arterioscler Thromb Vasc
Biol 33: e75 e83, 2013.
394. De Keulenaer GW, Chappell DC, Ishizaka N, Nerem RM, Alexander RW, Griendling
KK. Oscillatory and steady laminar shear stress differentially affect human endothelial redox state: role of a superoxide-producing NADH oxidase. Circ Res 82: 1094
1101, 1998.
395. De la Llera-Moya M, Drazul-Schrader D, Asztalos BF, Cuchel M, Rader DJ, Rothblat
GH. The ability to promote efflux via ABCA1 determines the capacity of serum
specimens with similar high-density lipoprotein cholesterol to remove cholesterol
from macrophages. Arterioscler Thromb Vasc Biol 30: 796 801, 2010.
396. De Nooijer R, Verkleij CJ, von der Thusen JH, Jukema JW, van der Wall EE, van
Berkel TJ, Baker AH, Biessen EA. Lesional overexpression of matrix metalloproteinase-9 promotes intraplaque hemorrhage in advanced lesions but not at earlier
stages of atherogenesis. Arterioscler Thromb Vasc Biol 26: 340 346, 2006.
397. De Villiers WJ, Smith JD, Miyata M, Dansky HM, Darley E, Gordon S. Macrophage
phenotype in mice deficient in both macrophage-colony-stimulating factor (op) and
apolipoprotein E. Arterioscler Thromb Vasc Biol 18: 631 640, 1998.
398. De Waard V, Arkenbout EK, Carmeliet P, Lindner V, Pannekoek H. Plasminogen
activator inhibitor 1 and vitronectin protect against stenosis in a murine carotid
artery ligation model. Arterioscler Thromb Vasc Biol 22: 1978 1983, 2002.
399. Deakin NO, Turner CE. Paxillin comes of age. J Cell Sci 121: 24352444, 2008.
400. Degoma EM, deGoma RL, Rader DJ. Beyond high-density lipoprotein cholesterol
levels evaluating high-density lipoprotein function as influenced by novel therapeutic
approaches. J Am Coll Cardiol 51: 2199 2211, 2008.
401. Degoma EM, Rader DJ. Novel HDL-directed pharmacotherapeutic strategies. Nat
Rev Cardiol 2011.
402. Deguchi JO, Aikawa E, Libby P, Vachon JR, Inada M, Krane SM, Whittaker P, Aikawa
M. Matrix metalloproteinase-13/collagenase-3 deletion promotes collagen accumulation and organization in mouse atherosclerotic plaques. Circulation 112: 2708
2715, 2005.
403. Dekker RJ, van Soest S, Fontijn RD, Salamanca S, de Groot PG, VanBavel E, Pannekoek H, Horrevoets AJG. Prolonged fluid shear stress induces a distinct set of
endothelial cell genes, most specifically lung Kruppel-like factor (KLF2). Blood 100:
1689 1698, 2002.
1496
410. Deng Y, Edin ML, Theken KN, Schuck RN, Flake GP, Kannon MA, Degraff LM, Lih
FB, Foley J, Bradbury JA, Graves JP, Tomer KB, Falck JR, Zeldin DC, Lee CR.
Endothelial CYP epoxygenase overexpression and soluble epoxide hydrolase disruption attenuate acute vascular inflammatory responses in mice. FASEB J 2010.
411. Deng Y, Theken KN, Lee CR. Cytochrome P450 epoxygenases, soluble epoxide
hydrolase, and the regulation of cardiovascular inflammation. J Mol Cell Cardiol 48:
331341, 2010.
412. Denis M, Haidar B, Marcil M, Bouvier M, Krimbou L, Genest J Jr. Molecular and
cellular physiology of apolipoprotein A-I lipidation by the ATP-binding cassette transporter A1 (ABCA1). J Biol Chem 279: 7384 7394, 2004.
413. Derwall M, Malhotra R, Lai CS, Beppu Y, Aikawa E, Seehra JS, Zapol WM, Bloch KD,
Yu PB. Inhibition of bone morphogenetic protein signaling reduces vascular calcification and atherosclerosis. Arterioscler Thromb Vasc Biol 32: 613 622, 2012.
414. Deshmukh L, Gorbatyuk V, Vinogradova O. Integrin 3 phosphorylation dictates its
complex with the Shc phosphotyrosine-binding (PTB) domain. J Biol Chem 285:
3487534884, 2010.
415. DeSouza CA, Shapiro LF, Clevenger CM, Dinenno FA, Monahan KD, Tanaka H,
Seals DR. Regular aerobic exercise prevents and restores age-related declines in
endothelium-dependent vasodilation in healthy men. Circulation 102: 13511357,
2000.
416. Deuchar GA, McLean D, Hadoke PW, Brownstein DG, Webb DJ, Mullins JJ, Chapman K, Seckl JR, Kotelevtsev YV. 11beta-hydroxysteroid dehydrogenase type 2
deficiency accelerates atherogenesis and causes proinflammatory changes in the
endothelium in apoe/ mice. Endocrinology 152: 236 246, 2011.
417. Devlin CM, Leventhal AR, Kuriakose G, Schuchman EH, Williams KJ, Tabas I. Acid
sphingomyelinase promotes lipoprotein retention within early atheromata and accelerates lesion progression. Arterioscler Thromb Vasc Biol 28: 17231730, 2008.
418. Dezso Z, Nikolsky Y, Nikolskaya T, Miller J, Cherba D, Webb C, Bugrim A. Identifying disease-specific genes based on their topological significance in protein networks. BMC Syst Biol 3: 36, 2009.
419. Dhillon AS, Yip YY, Grindlay GJ, Pakay JL, Dangers M, Hillmann M, Clark W, Pitt A,
Mischak H, Kolch W. The C-terminus of Raf-1 acts as a 14-3-3-dependent activation
switch. Cell Signal 21: 16451651, 2009.
420. Di Angelantonio E, Sarwar N, Perry P, Kaptoge S, Ray KK, Thompson A, Wood AM,
Lewington S, Sattar N, Packard CJ, Collins R, Thompson SG, Danesh J. Major lipids,
apolipoproteins, and risk of vascular disease. JAMA 302: 19932000, 2009.
421. Di Francesco L, Totani L, Dovizio M, Piccoli A, Di Francesco A, Salvatore T, Pandolfi
A, Evangelista V, Dercho RA, Seta F, Patrignani P. Induction of prostacyclin by steady
392. De Jager SC, Bermudez B, Bot I, Koenen RR, Bot M, Kavelaars A, de Waard V,
Heijnen CJ, Muriana FJ, Weber C, van Berkel TJ, Kuiper J, Lee SJ, Abia R, Biessen EA.
Growth differentiation factor 15 deficiency protects against atherosclerosis by attenuating CCR2-mediated macrophage chemotaxis. J Exp Med 208: 217225, 2011.
422. Dickhout JG, Lhotak S, Hilditch BA, Basseri S, Colgan SM, Lynn EG, Carlisle RE, Zhou
J, Sood SK, Ingram AJ, Austin RC. Induction of the unfolded protein response after
monocyte to macrophage differentiation augments cell survival in early atherosclerotic lesions. FASEB J 25: 576 589, 2011.
423. Diez-Dacal B, Perez-Sala D. Anti-inflammatory prostanoids: focus on the interactions between electrophile signaling and resolution of inflammation. Sci World J 10:
655 675, 2010.
442. Dovas A, Choi Y, Yoneda A, Multhaupt HA, Kwon SH, Kang D, Oh ES, Couchman JR.
Serine 34 phosphorylation of rho guanine dissociation inhibitor (RhoGDIalpha) links
signaling from conventional protein kinase C to RhoGTPase in cell adhesion. J Biol
Chem 285: 23296 23308, 2010.
424. Diez-Juan A, Andres V. The growth suppressor p27(Kip1) protects against dietinduced atherosclerosis. FASEB J 15: 1989 1995, 2001.
443. Doyon P, Servant MJ. Tumor necrosis factor receptor-associated factor-6 and ribosomal S6 kinase intracellular pathways link the angiotensin II AT1 receptor to the
phosphorylation and activation of the IkappaB kinase complex in vascular smooth
muscle cells. J Biol Chem 285: 30708 30718, 2010.
444. Drechsler M, Doring Y, Megens RT, Soehnlein O. Neutrophilic granulocytes - promiscuous accelerators of atherosclerosis. Thromb Haemost 106: 839 848, 2011.
426. Dikic I, Tokiwa G, Lev S, Courtneidge SA, Schlessinger J. A role for Pyk2 and Src in
linking G protein-coupled receptors with MAP kinase activation. Nature 383: 547
550, 1996.
427. Ding T, Li Z, Hailemariam T, Mukherjee S, Maxfield FR, Wu MP, Jiang XC. SMS
overexpression and knockdown: impact on cellular sphingomyelin and diacylglycerol
metabolism, and cell apoptosis. J Lipid Res 49: 376 385, 2008.
445. Drechsler M, Megens RT, van Zandvoort M, Weber C, Soehnlein O. Hyperlipidemia-triggered neutrophilia promotes early atherosclerosis. Circulation 122: 1837
1845, 2010.
446. Drinane M, Mollmark J, Zagorchev L, Moodie K, Sun B, Hall A, Shipman S, Morganelli
P, Simons M, Mulligan-Kehoe MJ. The antiangiogenic activity of rPAI-1(23) inhibits
vasa vasorum and growth of atherosclerotic plaque. Circ Res 104: 337345, 2009.
447. Dryden NH, Sperone A, Martin-Almedina S, Hannah RL, Birdsey GM, Khan ST,
Layhadi JA, Mason JC, Haskard DO, Gottgens B, Randi AM. The transcription factor
Erg controls endothelial cell quiescence by repressing activity of nuclear factor (NF)kappaB p65. J Biol Chem 287: 1233112342, 2012.
448. Du H, Grabowski GA. Lysosomal acid lipase and atherosclerosis. Curr Opin Lipidol 15:
539 544, 2004.
430. Doi H, Kugiyama K, Oka H, Sugiyama S, Ogata N, Koide SI, Nakamura SI, Yasue H.
Remnant lipoproteins induce proatherothrombogenic molecules in endothelial cells
through a redox-sensitive mechanism. Circulation 102: 670 676, 2000.
449. Du H, Schiavi S, Wan N, Levine M, Witte DP, Grabowski GA. Reduction of atherosclerotic plaques by lysosomal acid lipase supplementation. Arterioscler Thromb Vasc
Biol 24: 147154, 2004.
431. Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev
Immunol 11: 98 107, 2011.
450. Du Y, Kowluru A, Kern TS. PP2A contributes to endothelial death in high glucose:
inhibition by benfotiamine. Am J Physiol Regul Integr Comp Physiol 299: R1610 R1617,
2010.
432. Donato AJ, Eskurza I, Silver AE, Levy AS, Pierce GL, Gates PE, Seals DR. Direct
evidence of endothelial oxidative stress with aging in humans: relation to impaired
endothelium-dependent dilation and upregulation of nuclear factor-kappaB. Circ Res
100: 1659 1666, 2007.
433. Donato AJ, Magerko KA, Lawson BR, Durrant JR, Lesniewski LA, Seals DR. SIRT-1
and vascular endothelial dysfunction with ageing in mice and humans. J Physiol 589:
4545 4554, 2011.
452. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, Abela GS,
Franchi L, Nunez G, Schnurr M, Espevik T, Lien E, Fitzgerald KA, Rock KL, Moore KJ,
Wright SD, Hornung V, Latz E. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 464: 13571361, 2010.
434. Dong J, Liu J, Lou B, Li Z, Ye X, Wu M, Jiang XC. Adenovirus-mediated overexpression of sphingomyelin synthases 1 and 2 increases the atherogenic potential in mice.
J Lipid Res 47: 13071314, 2006.
453. Dufort CC, Paszek MJ, Weaver VM. Balancing forces: architectural control of
mechanotransduction. Nat Rev Mol Cell Biol 12: 308 319, 2011.
435. Dong Y, Zhang M, Liang B, Xie Z, Zhao Z, Asfa S, Choi HC, Zou MH. Reduction of
AMP-activated protein kinase alpha2 increases endoplasmic reticulum stress and
atherosclerosis in vivo. Circulation 121: 792 803, 2010.
436. Dong ZM, Chapman SM, Brown AA, Frenette PS, Hynes RO, Wagner DD. The
combined role of P- and E-selectins in atherosclerosis. J Clin Invest 102: 145152,
1998.
437. Doran AC, Lipinski MJ, Oldham SN, Garmey JC, Campbell KA, Skaflen MD, Cutchins
A, Lee DJ, Glover DK, Kelly KA, Galkina EV, Ley K, Witztum JL, Tsimikas S, Bender
TP, McNamara CA. B-cell aortic homing and atheroprotection depend on Id3. Circ
Res 110: e112, 2012.
438. Doran AC, Meller N, McNamara CA. Role of smooth muscle cells in the initiation
and early progression of atherosclerosis. Arterioscler Thromb Vasc Biol 28: 812 819,
2008.
439. Doring Y, Manthey HD, Drechsler M, Lievens D, Megens RT, Soehnlein O, Busch M,
Manca M, Koenen RR, Pelisek J, Daemen MJ, Lutgens E, Zenke M, Binder CJ, Weber
C, Zernecke A. Auto-antigenic protein-DNA complexes stimulate plasmacytoid
dendritic cells to promote atherosclerosis. Circulation 125: 16731683, 2012.
440. Doring Y, Soehnlein O, Drechsler M, Shagdarsuren E, Chaudhari SM, Meiler S,
Hartwig H, Hristov M, Koenen RR, Hieronymus T, Zenke M, Weber C, Zernecke A.
454. Dufour SP, Patel RP, Brandon A, Teng X, Pearson J, Barker H, Ali L, Yuen AH,
Smolenski RT, Gonzalez-Alonso J. Erythrocyte-dependent regulation of human skeletal muscle blood flow: role of varied oxyhemoglobin and exercise on nitrite, Snitrosohemoglobin, and ATP. Am J Physiol Heart Circ Physiol 299: H1936 H1946,
2010.
455. Duong PT, Collins HL, Nickel M, Lund-Katz S, Rothblat GH, Phillips MC. Characterization of nascent HDL particles and microparticles formed by ABCA1-mediated
efflux of cellular lipids to apoA-I. J Lipid Res 47: 832 843, 2006.
456. Duran S, Gonzalez LA, Alarcon GS. Damage, accelerated atherosclerosis, and mortality in patients with systemic lupus erythematosus: lessons from LUMINA, a multiethnic US cohort. J Clin Rheumatol 13: 350 353, 2007.
457. Durrant JR, Seals DR, Connell ML, Russell MJ, Lawson BR, Folian BJ, Donato AJ,
Lesniewski LA. Voluntary wheel running restores endothelial function in conduit
arteries of old mice: direct evidence for reduced oxidative stress, increased superoxide dismutase activity and down-regulation of NADPH oxidase. J Physiol 587:
32713285, 2009.
458. Ebrahimian T, Li MW, Lemarie CA, Simeone SM, Pagano PJ, Gaestel M, Paradis P,
Wassmann S, Schiffrin EL. Mitogen-activated protein kinase-activated protein kinase
2 in angiotensin II-induced inflammation and hypertension: regulation of oxidative
stress. Hypertension 57: 245254, 2011.
1497
428. Dixit M, Loot AE, Mohamed A, Fisslthaler B, Boulanger CM, Ceacareanu B, Hassid A,
Busse R, Fleming I. Gab1, SHP2, and protein kinase A are crucial for the activation of
the endothelial NO synthase by fluid shear stress. Circ Res 97: 1236 1244, 2005.
PAUL N. HOPKINS
459. Eck MV, Oost J, Goudriaan JR, Hoekstra M, Hildebrand RB, Bos IS, van Dijk KW, Van
Berkel TJ. Role of the macrophage very-low-density lipoprotein receptor in atherosclerotic lesion development. Atherosclerosis 183: 230 237, 2005.
460. Edirisinghe I, Rahman I. Cigarette smoke-mediated oxidative stress, shear stress, and
endothelial dysfunction: role of VEGFR2. Ann NY Acad Sci 1203: 66 72, 2010.
461. Edwards CJ, Syddall H, Goswami R, Goswami P, Dennison EM, Arden NK, Cooper
C. The autoantibody rheumatoid factor may be an independent risk factor for ischaemic heart disease in men. Heart 93: 12631267, 2007.
462. Egan KM, Lawson JA, Fries S, Koller B, Rader DJ, Smyth EM, Fitzgerald GA. COX2-derived prostacyclin confers atheroprotection on female mice. Science 306: 1954
1957, 2004.
463. Egan KM, Wang M, Fries S, Lucitt MB, Zukas AM, Pure E, Lawson JA, FitzGerald GA.
Cyclooxygenases, thromboxane, and atherosclerosis: plaque destabilization by cyclooxygenase-2 inhibition combined with thromboxane receptor antagonism. Circulation 111: 334 342, 2005.
464. Egorova AD, Khedoe PP, Goumans MJ, Yoder BK, Nauli SM, ten Dijke P, Poelmann
RE, Hierck BP. Lack of primary cilia primes shear-induced endothelial-to-mesenchymal transition. Circ Res 108: 10931101, 2011.
465. Eguchi S, Dempsey PJ, Frank GD, Motley ED, Inagami T. Activation of MAPKs by
angiotensin II in vascular smooth muscle cells. Metalloprotease-dependent EGF receptor activation is required for activation of ERK and p38 MAPK but not for JNK. J
Biol Chem 276: 79577962, 2001.
467. El-Osta A, Brasacchio D, Yao D, Pocai A, Jones PL, Roeder RG, Cooper ME, Brownlee M. Transient high glucose causes persistent epigenetic changes and altered gene
expression during subsequent normoglycemia. J Exp Med 205: 2409 2417, 2008.
486. Erlinge D, Burnstock G. P2 receptors in cardiovascular regulation and disease. Purinergic Signal 4: 120, 2008.
469. Elhage R, Gourdy P, Brouchet L, Jawien J, Fouque MJ, Fievet C, Huc X, Barreira Y,
Couloumiers JC, Arnal JF, Bayard F. Deleting TCR alpha beta or CD4 T lymphocytes leads to opposite effects on site-specific atherosclerosis in female apolipoprotein E-deficient mice. Am J Pathol 165: 20132018, 2004.
470. Elhage R, Gourdy P, Jawien J, Brouchet L, Castano C, Fievet C, Hansson GK, Arnal
JF, Bayard F. The atheroprotective effect of 17beta-estradiol depends on complex
interactions in adaptive immunity. Am J Pathol 167: 267274, 2005.
471. Elhage R, Jawien J, Rudling M, Ljunggren HG, Takeda K, Akira S, Bayard F, Hansson
GK. Reduced atherosclerosis in interleukin-18 deficient apolipoprotein E-knockout
mice. Cardiovasc Res 59: 234 240, 2003.
472. Elia L, Quintavalle M, Zhang J, Contu R, Cossu L, Latronico MV, Peterson KL, Indolfi
C, Catalucci D, Chen J, Courtneidge SA, Condorelli G. The knockout of miR-143 and
-145 alters smooth muscle cell maintenance and vascular homeostasis in mice: correlates with human disease. Cell Death Differ 16: 1590 1598, 2009.
473. Elliott MR, Ravichandran KS. Clearance of apoptotic cells: implications in health and
disease. J Cell Biol 189: 1059 1070, 2010.
474. Elliott P, Chambers JC, Zhang W, Clarke R, Hopewell JC, Peden JF, Erdmann J,
Braund P, Engert JC, Bennett D, Coin L, Ashby D, Tzoulaki I, Brown IJ, Mt-Isa S,
McCarthy MI, Peltonen L, Freimer NB, Farrall M, Ruokonen A, Hamsten A, Lim N,
Froguel P, Waterworth DM, Vollenweider P, Waeber G, Jarvelin MR, Mooser V,
Scott J, Hall AS, Schunkert H, Anand SS, Collins R, Samani NJ, Watkins H, Kooner JS.
Genetic loci associated with C-reactive protein levels and risk of coronary heart
disease. JAMA 302: 37 48, 2009.
494. Falk E, Shah PK, Fuster V. Coronary plaque disruption. Circulation 92: 657 671,
1995.
475. Ellsworth ML, Ellis CG, Goldman D, Stephenson AH, Dietrich HH, Sprague RS.
Erythrocytes: oxygen sensors and modulators of vascular tone. Physiology 24: 107
116, 2009.
496. Fan LM, Teng L, Li JM. Knockout of p47 phox uncovers a critical role of p40 phox in
reactive oxygen species production in microvascular endothelial cells. Arterioscler
Thromb Vasc Biol 29: 16511656, 2009.
476. Enesa K, Ito K, Luong le A, Thorbjornsen I, Phua C, To Y, Dean J, Haskard DO, Boyle
J, Adcock I, Evans PC. Hydrogen peroxide prolongs nuclear localization of NFkappaB in activated cells by suppressing negative regulatory mechanisms. J Biol Chem
283: 1858218590, 2008.
497. Fan Y, Shi F, Liu J, Dong J, Bui HH, Peake DA, Kuo MS, Cao G, Jiang XC. Selective
reduction in the sphingomyelin content of atherogenic lipoproteins inhibits their
retention in murine aortas and the subsequent development of atherosclerosis.
Arterioscler Thromb Vasc Biol 30: 2114 2120, 2010.
1498
495. Fan J, Frey RS, Malik AB. TLR4 signaling induces TLR2 expression in endothelial cells
via neutrophil NADPH oxidase. J Clin Invest 112: 1234 1243, 2003.
466. Eken C, Martin PJ, Sadallah S, Treves S, Schaller M, Schifferli JA. Ectosomes released
by polymorphonuclear neutrophils induce a MerTK-dependent anti-inflammatory
pathway in macrophages. J Biol Chem 285: 39914 39921, 2010.
499. Fang F, Fall K, Mittleman MA, Sparn P, Ye W, Adami HO, Valdimarsdttir U. Suicide
and cardiovascular death after a cancer diagnosis. N Engl J Med 366: 1310 1318,
2012.
518. Feng Y, Gordts SC, Chen F, Hu Y, Van Craeyveld E, Jacobs F, Carlier V, Zhang Z, Xu
Q, Ni Y, De Geest B. Topical HDL administration reduces vein graft atherosclerosis
in ApoE deficient mice. Atherosclerosis 214: 271278, 2011.
501. Fang Y, Mohler ER, III, Hsieh E, Osman H, Hashemi SM, Davies PF, Rothblat GH,
Wilensky RL, Levitan I. Hypercholesterolemia suppresses inwardly rectifying K
channels in aortic endothelium in vitro and in vivo. Circ Res 98: 1064 1071, 2006.
502. Fazio S, Babaev VR, Murray AB, Hasty AH, Carter KJ, Gleaves LA, Atkinson JB,
Linton MF. Increased atherosclerosis in mice reconstituted with apolipoprotein E
null macrophages. Proc Natl Acad Sci USA 94: 4647 4652, 1997.
503. Fazio S, Hasty AH, Carter KJ, Murray AB, Price JO, Linton MF. Leukocyte low
density lipoprotein receptor (LDL-R) does not contribute to LDL clearance in vivo:
bone marrow transplantation studies in the mouse. J Lipid Res 38: 391 400, 1997.
504. Fazio S, Major AS, Swift LL, Gleaves LA, Accad M, Linton MF, Farese RV Jr. Increased
atherosclerosis in LDL receptor-null mice lacking ACAT1 in macrophages. J Clin
Invest 107: 163171, 2001.
506. Feaver RE, Hastings NE, Pryor A, Blackman BR. GRP78 upregulation by atheroprone
shear stress via p38-, alpha2beta1-dependent mechanism in endothelial cells. Arterioscler Thromb Vasc Biol 28: 1534 1541, 2008.
507. Febbraio M, Podrez EA, Smith JD, Hajjar DP, Hazen SL, Hoff HF, Sharma K, Silverstein RL. Targeted disruption of the class B scavenger receptor CD36 protects
against atherosclerotic lesion development in mice. J Clin Invest 105: 1049 1056,
2000.
508. Feig JE, Pineda-Torra I, Sanson M, Bradley MN, Vengrenyuk Y, Bogunovic D, Gautier
EL, Rubinstein D, Hong C, Liu J, Wu C, van Rooijen N, Bhardwaj N, Garabedian M,
Tontonoz P, Fisher EA. LXR promotes the maximal egress of monocyte-derived
cells from mouse aortic plaques during atherosclerosis regression. J Clin Invest 120:
4415 4424, 2010.
509. Feil S, Hofmann F, Feil R. SM22alpha modulates vascular smooth muscle cell phenotype during atherogenesis. Circ Res 94: 863 865, 2004.
510. Feletou M, Vanhoutte PM, Verbeuren TJ. The thromboxane/endoperoxide receptor
(TP): the common villain. J Cardiovasc Pharmacol 55: 317332, 2010.
511. Felton CV, Crook D, Davies MJ, Oliver MF. Relation of plaque lipid composition and
morphology to the stability of human aortic plaques. Arterioscler Thromb Vasc Biol 17:
13371345, 1997.
512. Feng B, Tabas I. ABCA1-mediated cholesterol efflux is defective in free cholesterolloaded macrophages. Mechanism involves enhanced ABCA1 degradation in a process requiring full NPC1 activity. J Biol Chem 277: 43271 43280, 2002.
513. Feng B, Yao PM, Li Y, Devlin CM, Zhang D, Harding HP, Sweeney M, Rong JX,
Kuriakose G, Fisher EA, Marks AR, Ron D, Tabas I. The endoplasmic reticulum is the
site of cholesterol-induced cytotoxicity in macrophages. Nat Cell Biol 5: 781792,
2003.
514. Feng B, Zhang D, Kuriakose G, Devlin CM, Kockx M, Tabas I. Niemann-Pick C
heterozygosity confers resistance to lesional necrosis and macrophage apoptosis in
murine atherosclerosis. Proc Natl Acad Sci USA 100: 1042310428, 2003.
515. Feng J, Damrauer SM, Lee M, Sellke FW, Ferran C, Abid MR. Endothelium-dependent coronary vasodilatation requires NADPH oxidase-derived reactive oxygen species. Arterioscler Thromb Vasc Biol 30: 17031710, 2010.
516. Feng W, Xing D, Hua P, Zhang Y, Chen YF, Oparil S, Jaimes EA. The transcription
factor ETS-1 mediates proinflammatory responses and neointima formation in carotid artery endoluminal vascular injury. Hypertension 55: 13811388, 2010.
1499
505. Feaver RE, Gelfand BD, Wang C, Schwartz MA, Blackman BR. Atheroprone hemodynamics regulate fibronectin deposition to create positive feedback that sustains
endothelial inflammation. Circ Res 106: 17031711, 2010.
521. Fernandez-Hernando C, Yu J, Davalos A, Prendergast J, Sessa WC. Endothelialspecific overexpression of caveolin-1 accelerates atherosclerosis in apolipoprotein
E-deficient mice. Am J Pathol 177: 998 1003, 2010.
PAUL N. HOPKINS
536. Francia P, delli Gatti C, Bachschmid M, Martin-Padura I, Savoia C, Migliaccio E,
Pelicci PG, Schiavoni M, Luscher TF, Volpe M, Cosentino F. Deletion of p66shc gene
protects against age-related endothelial dysfunction. Circulation 110: 2889 2895,
2004.
537. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev 236: 219 242, 2010.
538. Franco C, Hou G, Ahmad PJ, Fu EY, Koh L, Vogel WF, Bendeck MP. Discoidin
domain receptor 1 (ddr1) deletion decreases atherosclerosis by accelerating matrix
accumulation and reducing inflammation in low-density lipoprotein receptor-deficient mice. Circ Res 102: 12021211, 2008.
539. Frank GD, Saito S, Motley ED, Sasaki T, Ohba M, Kuroki T, Inagami T, Eguchi S.
Requirement of Ca2 and PKCdelta for Janus kinase 2 activation by angiotensin II:
involvement of PYK2. Mol Endocrinol 16: 367377, 2002.
540. Franscini N, Bachli EB, Blau N, Leikauf MS, Schaffner A, Schoedon G. Gene expression profiling of inflamed human endothelial cells and influence of activated protein
C. Circulation 110: 29032909, 2004.
541. Frasca L, Lande R. Overlapping, additive and counterregulatory effects of type II and
I interferons on myeloid dendritic cell functions. Scientific World J 11: 20712090,
2011.
542. Frasch SC, Fernandez-Boyanapalli RF, Berry KZ, Leslie CC, Bonventre JV, Murphy
RC, Henson PM, Bratton DL. Signaling via macrophage G2A enhances efferocytosis
of dying neutrophils by augmentation of Rac activity. J Biol Chem 286: 12108 12122,
2011.
544. Fredman G, Serhan CN. Specialized proresolving mediator targets for RvE1 and
RvD1 in peripheral blood and mechanisms of resolution. Biochem J 437: 185197,
2011.
545. Fredrikson GN, Andersson L, Soderberg I, Dimayuga P, Chyu KY, Shah PK, Nilsson
J. Atheroprotective immunization with MDA-modified apo B-100 peptide sequences is associated with activation of Th2 specific antibody expression. Autoimmunity 38: 171179, 2005.
546. Freigang S, Ampenberger F, Spohn G, Heer S, Shamshiev AT, Kisielow J, Hersberger
M, Yamamoto M, Bachmann MF, Kopf M. Nrf2 is essential for cholesterol crystalinduced inflammasome activation and exacerbation of atherosclerosis. Eur J Immunol
41: 2040 2051, 2011.
547. Freigang S, Horkko S, Miller E, Witztum JL, Palinski W. Immunization of LDL receptor-deficient mice with homologous malondialdehyde-modified and native LDL reduces progression of atherosclerosis by mechanisms other than induction of high
titers of antibodies to oxidative neoepitopes. Arterioscler Thromb Vasc Biol 18: 1972
1982, 1998.
548. French CJ, Zaman AK, Sobel BE. The angiotensin receptor blocker, Azilsartan medoxomil (TAK-491), suppresses vascular wall expression of plasminogen activator
inhibitor type-I (PAI-I) protein potentially facilitating the stabilization of atherosclerotic plaques. J Cardiovasc Pharmacol 2011.
549. Freudenberger T, Oppermann M, Marzoll A, Heim HK, Mayer P, Kojda G, Weber
AA, Schror K, Fischer JW. Differential effects of medroxyprogesterone acetate on
thrombosis and atherosclerosis in mice. Br J Pharmacol 158: 19511960, 2009.
550. Frey RS, Gao X, Javaid K, Siddiqui SS, Rahman A, Malik AB. Phosphatidylinositol
3-kinase signaling through protein kinase C induces NADPH oxidase-mediated
oxidant generation and NF-B activation in endothelial cells. J Biol Chem 281:
16128 16138, 2006.
551. Frey RS, Ushio-Fukai M, Malik AB. NADPH oxidase-dependent signaling in endothelial cells: role in physiology and pathophysiology. Antioxid Redox Signal 11: 791
810, 2009.
552. Friedland JC, Lee MH, Boettiger D. Mechanically activated integrin switch controls
alpha5beta1 function. Science 323: 642 644, 2009.
553. Frolova EG, Pluskota E, Krukovets I, Burke T, Drumm C, Smith JD, Blech L, Febbraio
M, Bornstein P, Plow EF, Stenina OI. Thrombospondin-4 regulates vascular inflammation and atherogenesis. Circ Res 107: 13131325, 2010.
1500
543. Fraser DA, Tenner AJ. Innate immune proteins C1q and mannan-binding lectin
enhance clearance of atherogenic lipoproteins by human monocytes and macrophages. J Immunol 185: 39323939, 2010.
554. Frostegard J. Low level natural antibodies against phosphorylcholine: a novel risk
marker and potential mechanism in atherosclerosis and cardiovascular disease. Clin
Immunol 134: 4754, 2010.
592. Gareus R, Kotsaki E, Xanthoulea S, van der Made I, Gijbels MJ, Kardakaris R, Polykratis A, Kollias G, de Winther MP, Pasparakis M. Endothelial cell-specific NF-kappaB
inhibition protects mice from atherosclerosis. Cell Metab 8: 372383, 2008.
573. Gabrielsen JS, Gao Y, Simcox JA, Huang J, Thorup D, Jones D, Cooksey RC, Gabrielsen D, Adams TD, Hunt SC, Hopkins PN, Cefalu WT, McClain DA. Adipocyte iron
regulates adiponectin and insulin sensitivity. J Clin Invest 122: 3529 3540, 2012.
574. Gaffen SL. Structure and signalling in the IL-17 receptor family. Nat Rev Immunol 9:
556 567, 2009.
575. Gage J, Hasu M, Thabet M, Whitman SC. Caspase-1 deficiency decreases atherosclerosis in apolipoprotein E-null mice. Can J Cardiol. In press.
576. Gage M, Yuldasheva N, Jackson C, Kearney M, Imrie H, Viswambharan H, Rajwani A,
Kahn M, Surr J, Cubbon R, Wheatcroft S. Abstract 17990: endothelial specific insulin
resistance promotes the development of atherosclerosis. Circulation 122: A17990,
2010.
577. Galbusera M, Zoja C, Donadelli R, Paris S, Morigi M, Benigni A, Figliuzzi M, Remuzzi
G, Remuzzi A. Fluid shear stress modulates von Willebrand factor release from
human vascular endothelium. Blood 90: 1558 1564, 1997.
578. Galisteo ML, Yang Y, Urena J, Schlessinger J. Activation of the nonreceptor protein
tyrosine kinase Ack by multiple extracellular stimuli. Proc Natl Acad Sci USA 103:
9796 9801, 2006.
594. Garin G, Abe Ji Mohan A, Lu W, Yan C, Newby AC, Rhaman A, Berk BC. Flow
antagonizes TNF- signaling in endothelial cells by inhibiting caspase-dependent
PKC processing. Circ Res 101: 97105, 2007.
595. Gautier EL, Huby T, Saint-Charles F, Ouzilleau B, Pirault J, Deswaerte V, Ginhoux F,
Miller ER, Witztum JL, Chapman MJ, Lesnik P. Conventional dendritic cells at the
crossroads between immunity and cholesterol homeostasis in atherosclerosis. Circulation 119: 23672375, 2009.
596. Gautier EL, Huby T, Witztum JL, Ouzilleau B, Miller ER, Saint-Charles F, Aucouturier
P, Chapman MJ, Lesnik P. Macrophage apoptosis exerts divergent effects on atherogenesis as a function of lesion stage. Circulation 119: 17951804, 2009.
597. Gautreau L, Chabannes D, Heslan M, Josien R. Modulation of regulatory T cell-Th17
balance by plasmacytoid dendritic cells. J Leukoc Biol 90: 521527, 2011.
598. Geiger B, Yamada KM. Molecular architecture and function of matrix adhesions. Cold
Spring Harb Perspect Biol 3: 2011.
599. Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K. Development of
monocytes, macrophages, and dendritic cells. Science 327: 656 661, 2010.
600. Gelfand BD, Meller J, Pryor AW, Kahn M, Bortz PD, Wamhoff BR, Blackman BR.
Hemodynamic activation of beta-catenin and T-cell-specific transcription factor signaling in vascular endothelium regulates fibronectin expression. Arterioscler Thromb
Vasc Biol 31: 16251633, 2011.
601. Gelfand JM, Neimann AL, Shin DB, Wang X, Margolis DJ, Troxel AB. Risk of myocardial infarction in patients with psoriasis. JAMA 296: 17351741, 2006.
602. Geloen A, Helin L, Geeraert B, Malaud E, Holvoet P, Marguerie G. CD36 inhibitors
reduce postprandial hypertriglyceridemia and protect against diabetic dyslipidemia
and atherosclerosis. PLoS ONE 7: e37633, 2012.
603. George J. Mechanisms of disease: the evolving role of regulatory T cells in atherosclerosis. Nat Clin Pract Cardiovasc Med 5: 531540, 2008.
604. George J, Afek A, Gilburd B, Shoenfeld Y, Harats D. Cellular and humoral immune
responses to heat shock protein 65 are both involved in promoting fatty-streak
formation in LDL-receptor deficient mice. J Am Coll Cardiol 38: 900 905, 2001.
605. George J, Afek A, Shaish A, Levkovitz H, Bloom N, Cyrus T, Zhao L, Funk CD, Sigal
E, Harats D. 12/15-Lipoxygenase gene disruption attenuates atherogenesis in LDL
receptor-deficient mice. Circulation 104: 1646 1650, 2001.
606. George J, Yacov N, Breitbart E, Bangio L, Shaish A, Gilburd B, Shoenfeld Y, Harats D.
Suppression of early atherosclerosis in LDL-receptor deficient mice by oral tolerance with beta 2-glycoprotein I. Cardiovasc Res 62: 603 609, 2004.
607. Gerits N, Kostenko S, Moens U. In vivo functions of mitogen-activated protein
kinases: conclusions from knock-in and knock-out mice. Transgenic Res 16: 281314,
2007.
608. Gertz M, Steegborn C. The lifespan-regulator p66Shc in mitochondria: redox enzyme or redox sensor? Antioxid Redox Signal 13: 14171428, 2010.
609. Getz GS, Reardon CA. Animal models of atherosclerosis. Arterioscler Thromb Vasc
Biol 32: 1104 1115, 2012.
610. Gharavi NM, Alva JA, Mouillesseaux KP, Lai C, Yeh M, Yeung W, Johnson J, Szeto
WL, Hong L, Fishbein M, Wei L, Pfeffer LM, Berliner JA. Role of the Jak/STAT
pathway in the regulation of interleukin-8 transcription by oxidized phospholipids in
vitro and in atherosclerosis in vivo. J Biol Chem 282: 31460 31468, 2007.
611. Ghazalpour A, Wang X, Lusis AJ, Mehrabian M. Complex inheritance of the 5-lipoxygenase locus influencing atherosclerosis in mice. Genetics 173: 943951, 2006.
612. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circ Res 107:
1058 1070, 2010.
1501
579. Galkina E, Harry BL, Ludwig A, Liehn EA, Sanders JM, Bruce A, Weber C, Ley K.
CXCR6 promotes atherosclerosis by supporting T-cell homing, interferon-gamma
production, and macrophage accumulation in the aortic wall. Circulation 116: 1801
1811, 2007.
PAUL N. HOPKINS
613. Giaever G, Chu AM, Ni L, Connelly C, Riles L, Veronneau S, Dow S, Lucau-Danila A,
Anderson K, Andre B, Arkin AP, Astromoff A, El-Bakkoury M, Bangham R, Benito R,
Brachat S, Campanaro S, Curtiss M, Davis K, Deutschbauer A, Entian KD, Flaherty P,
Foury F, Garfinkel DJ, Gerstein M, Gotte D, Guldener U, Hegemann JH, Hempel S,
Herman Z, Jaramillo DF, Kelly DE, Kelly SL, Kotter P, LaBonte D, Lamb DC, Lan N,
Liang H, Liao H, Liu L, Luo C, Lussier M, Mao R, Menard P, Ooi SL, Revuelta JL,
Roberts CJ, Rose M, Ross-Macdonald P, Scherens B, Schimmack G, Shafer B, Shoemaker DD, Sookhai-Mahadeo S, Storms RK, Strathern JN, Valle G, Voet M, Volckaert G, Wang CY, Ward TR, Wilhelmy J, Winzeler EA, Yang Y, Yen G, Youngman E,
Yu K, Bussey H, Boeke JD, Snyder M, Philippsen P, Davis RW, Johnston M. Functional
profiling of the Saccharomyces cerevisiae genome. Nature 418: 387391, 2002.
614. Giannarelli C, Zafar MU, Badimon JJ. Prostanoid and TP-receptors in atherothrombosis: is there a role for their antagonism? Thromb Haemost 104: 949 954, 2010.
615. Giles JT, Szklo M, Post W, Petri M, Blumenthal RS, Lam G, Gelber AC, Detrano R,
Scott WW Jr, Kronmal RA, Bathon JM. Coronary arterial calcification in rheumatoid
arthritis: comparison with the Multi-Ethnic Study of Atherosclerosis. Arthritis Res
Ther 11: R36, 2009.
616. Ginnan R, Pfleiderer PJ, Pumiglia K, Singer HA. PKC-delta and CaMKII-delta 2 mediate ATP-dependent activation of ERK1/2 in vascular smooth muscle. Am J Physiol
Cell Physiol 286: C1281C1289, 2004.
617. Ginnan R, Singer HA. CaM kinase II-dependent activation of tyrosine kinases and
ERK1/2 in vascular smooth muscle. Am J Physiol Cell Physiol 282: C754 C761, 2002.
619. Gitlin JM, Homeister JW, Bulgrien J, Counselman J, Curtiss LK, Lowe JB, Boisvert
WA. Disruption of tissue-specific fucosyltransferase VII, an enzyme necessary for
selectin ligand synthesis, suppresses atherosclerosis in mice. Am J Pathol 174: 343
350, 2009.
620. Glagov S, Ozoa AK. Significance of the relatively low incidence of atherosclerosis in
the pumonary, renal and mesenteric arteries. Ann NY Acad Sci 149: 940 955, 1968.
621. Gleissner CA. Macrophage phenotype modulation by CXCL4 in atherosclerosis.
Front Physiol 3: 1, 2012.
622. Gleissner CA, Shaked I, Erbel C, Bockler D, Katus HA, Ley K. CXCL4 downregulates
the atheroprotective hemoglobin receptor CD163 in human macrophages. Circ Res
106: 203211, 2010.
643. Gordts PLSM, Reekmans S, Lauwers A, Van Dongen A, Verbeek L, Roebroek AJM.
Inactivation of the LRP1 intracellular NPxYxxL motif in LDLR-deficient mice enhances postprandial dyslipidemia and atherosclerosis. Arterioscler Thromb Vasc Biol
29: 1258 1264, 2009.
624. Go YM, Halvey PJ, Hansen JM, Reed M, Pohl J, Jones DP. Reactive aldehyde modification of thioredoxin-1 activates early steps of inflammation and cell adhesion. Am
J Pathol 171: 1670 1681, 2007.
644. Gosling J, Slaymaker S, Gu L, Tseng S, Zlot CH, Young SG, Rollins BJ, Charo IF.
MCP-1 deficiency reduces susceptibility to atherosclerosis in mice that overexpress
human apolipoprotein B. J Clin Invest 103: 773778, 1999.
645. Gotsman I, Grabie N, Dacosta R, Sukhova G, Sharpe A, Lichtman AH. Proatherogenic immune responses are regulated by the PD-1/PD-L pathway in mice. J Clin
Invest 117: 2974 2982, 2007.
626. Goel R, Schrank BR, Arora S, Boylan B, Fleming B, Miura H, Newman PJ, Molthen
RC, Newman DK. Site-specific effects of PECAM-1 on atherosclerosis in LDL receptor-deficient mice. Arterioscler Thromb Vasc Biol 28: 1996 2002, 2008.
1502
647. Gotsman I, Sharpe AH, Lichtman AH. T-cell costimulation and coinhibition in atherosclerosis. Circ Res 103: 1220 1231, 2008.
648. Gough PJ, Gomez IG, Wille PT, Raines EW. Macrophage expression of active MMP-9
induces acute plaque disruption in apoE-deficient mice. J Clin Invest 116: 59 69,
2006.
649. Grabner R, Lotzer K, Dopping S, Hildner M, Radke D, Beer M, Spanbroek R, Lippert
B, Reardon CA, Getz GS, Fu YX, Hehlgans T, Mebius RE, van der Wall M, Kruspe D,
Englert C, Lovas A, Hu D, Randolph GJ, Weih F, Habenicht AJ. Lymphotoxin beta
receptor signaling promotes tertiary lymphoid organogenesis in the aorta adventitia
of aged ApoE/ mice. J Exp Med 206: 233248, 2009.
650. Gradl D, Kuhl M, Wedlich D. The Wnt/Wg signal transducer beta-catenin controls
fibronectin expression. Mol Cell Biol 19: 5576 5587, 1999.
618. Gislason GH, Rasmussen JN, Abildstrom SZ, Schramm TK, Hansen ML, Fosbol EL,
Sorensen R, Folke F, Buch P, Gadsboll N, Rasmussen S, Poulsen HE, Kober L,
Madsen M, Torp-Pedersen C. Increased mortality and cardiovascular morbidity
associated with use of nonsteroidal anti-inflammatory drugs in chronic heart failure.
Arch Intern Med 169: 141149, 2009.
631. Goldfine AB, Fonseca V, Shoelson SE. Therapeutic approaches to target inflammation in type 2 diabetes. Clin Chem 57: 162167, 2011.
672. Gunduz D, Thom J, Hussain I, Lopez D, Hartel FV, Erdogan A, Grebe M, Sedding D,
Piper HM, Tillmanns H, Noll T, Aslam M. Insulin stabilizes microvascular endothelial
barrier function via phosphatidylinositol 3-kinase/Akt-mediated Rac1 activation. Arterioscler Thromb Vasc Biol 30: 12371245, 2010.
652. Grainger DJ, Reckless J, McKilligin E. Apolipoprotein E modulates clearance of apoptotic bodies in vitro and in vivo, resulting in a systemic proinflammatory state in
apolipoprotein E-deficient mice. J Immunol 173: 6366 6375, 2004.
673. Guns PJ, Hendrickx J, Van Assche T, Fransen P, Bult H. P2Y receptors and atherosclerosis in apolipoprotein E-deficient mice. Br J Pharmacol 159: 326 336, 2010.
653. Grashoff C, Hoffman BD, Brenner MD, Zhou R, Parsons M, Yang MT, McLean MA,
Sligar SG, Chen CS, Ha T, Schwartz MA. Measuring mechanical tension across
vinculin reveals regulation of focal adhesion dynamics. Nature 466: 263266, 2010.
674. Guo GL, Santamarina-Fojo S, Akiyama TE, Amar MJ, Paigen BJ, Brewer B Jr, Gonzalez FJ. Effects of FXR in foam-cell formation and atherosclerosis development.
Biochim Biophys Acta 1761: 14011409, 2006.
654. Greaves DR, Gordon S. The macrophage scavenger receptor at 30 years of age:
current knowledge and future challenges. J Lipid Res 50 Suppl: S282286, 2009.
675. Guo J, Van Eck M, Twisk J, Maeda N, Benson GM, Groot PH, Van Berkel TJ.
Transplantation of monocyte CC-chemokine receptor 2-deficient bone marrow
into ApoE3-Leiden mice inhibits atherogenesis. Arterioscler Thromb Vasc Biol 23:
447 453, 2003.
660. Groeger AL, Freeman BA. Signaling actions of electrophiles: anti-inflammatory therapeutic candidates. Mol Interv 10: 39 50, 2010.
661. Gross S, Tilly P, Hentsch D, Vonesch JL, Fabre JE. Vascular wall-produced prostaglandin E2 exacerbates arterial thrombosis and atherothrombosis through platelet
EP3 receptors. J Exp Med 204: 311320, 2007.
662. Grosser T, Yu Y, Fitzgerald GA. Emotion recollected in tranquility: lessons learned
from the COX-2 saga. Annu Rev Med 61: 1733, 2010.
663. Grosso DM, Ferderbar S, Wanschel AC, Krieger MH, Higushi ML, Abdalla DS.
Antibodies against electronegative LDL inhibit atherosclerosis in LDLr/ mice.
Braz J Med Biol Res 41: 1086 1092, 2008.
664. Gruber M, Christ-Crain M, Stolz D, Keller U, Muller C, Bingisser R, Tamm M,
Mueller B, Schuetz P. Prognostic impact of plasma lipids in patients with lower
respiratory tract infections: an observational study. Swiss Medical Weekly 139: 166
172, 2009.
665. Grunewald JK, Ridley AJ. CD73 represses pro-inflammatory responses in human
endothelial cells. J Inflamm 7: 10, 2010.
666. Gu L, Okada Y, Clinton SK, Gerard C, Sukhova GK, Libby P, Rollins BJ. Absence of
monocyte chemoattractant protein-1 reduces atherosclerosis in low density lipoprotein receptor-deficient mice. Mol Cell 2: 275281, 1998.
667. Gudi S, Huvar I, White CR, McKnight NL, Dusserre N, Boss GR, Frangos JA. Rapid
activation of Ras by fluid flow is mediated by Galpha(q) and Gbetagamma subunits of
heterotrimeric G proteins in human endothelial cells. Arterioscler Thromb Vasc Biol
23: 994 1000, 2003.
668. Guetta J, Strauss M, Levy NS, Fahoum L, Levy AP. Haptoglobin genotype modulates
the balance of Th1/Th2 cytokines produced by macrophages exposed to free hemoglobin. Atherosclerosis 191: 48 53, 2007.
677. Guo Z, Ran Q, Roberts LJ, 2nd Zhou L, Richardson A, Sharan C, Wu D, Yang H.
Suppression of atherogenesis by overexpression of glutathione peroxidase-4 in apolipoprotein E-deficient mice. Free Radic Biol Med 44: 343352, 2008.
678. Gupta S, Pablo AM, Jiang X, Wang N, Tall AR, Schindler C. IFN-gamma potentiates
atherosclerosis in ApoE knock-out mice. J Clin Invest 99: 27522761, 1997.
679. Guyton AC. Blood pressure controlspecial role of the kidneys and body fluids.
Science 252: 18131816, 1991.
680. Guzik TJ, Chen W, Gongora MC, Guzik B, Lob HE, Mangalat D, Hoch N, Dikalov S,
Rudzinski P, Kapelak B, Sadowski J, Harrison DG. Calcium-dependent NOX5 nicotinamide adenine dinucleotide phosphate oxidase contributes to vascular oxidative
stress in human coronary artery disease. J Am Coll Cardiol 52: 18031809, 2008.
681. Guzik TJ, Mussa S, Gastaldi D, Sadowski J, Ratnatunga C, Pillai R, Channon KM.
Mechanisms of increased vascular superoxide production in human diabetes mellitus: role of NAD(P)H oxidase and endothelial nitric oxide synthase. Circulation 105:
1656 1662, 2002.
682. Guzik TJ, Sadowski J, Guzik B, Jopek A, Kapelak B, Przybylowski P, Wierzbicki K,
Korbut R, Harrison DG, Channon KM. Coronary artery superoxide production and
NOX isoform expression in human coronary artery disease. Arterioscler Thromb Vasc
Biol 26: 333339, 2006.
683. Guzik TJ, West NE, Black E, McDonald D, Ratnatunga C, Pillai R, Channon KM.
Vascular superoxide production by NAD(P)H oxidase: association with endothelial
dysfunction and clinical risk factors. Circ Res 86: E8590, 2000.
684. Ha CH, Bennett AM, Jin ZG. A novel role of vascular endothelial cadherin in modulating c-Src activation and downstream signaling of vascular endothelial growth
factor. J Biol Chem 283: 72617270, 2008.
685. Haase CL, Frikke-Schmidt R, Nordestgaard BG, Tybjaerg-Hansen A. Populationbased resequencing of APOA1 in 10,330 individuals: spectrum of genetic variation,
phenotype, and comparison with extreme phenotype approach. PLoS Genet 8:
e1003063, 2012.
686. Habelhah H. Emerging complexity of protein ubiquitination in the NF-B pathway.
Genes & Cancer 1: 735747, 2010.
687. Haberzettl P, Vladykovskaya E, Srivastava S, Bhatnagar A. Role of endoplasmic reticulum stress in acrolein-induced endothelial activation. Toxicol Appl Pharmacol 234:
14 24, 2009.
669. Guevara NV, Kim HS, Antonova EI, Chan L. The absence of p53 accelerates atherosclerosis by increasing cell proliferation in vivo. Nat Med 5: 335339, 1999.
688. Habets KL, van Puijvelde GH, van Duivenvoorde LM, van Wanrooij EJ, de Vos P,
Tervaert JW, van Berkel TJ, Toes RE, Kuiper J. Vaccination using oxidized lowdensity lipoprotein-pulsed dendritic cells reduces atherosclerosis in LDL receptordeficient mice. Cardiovasc Res 85: 622 630, 2010.
690. Hageman J, Herrema H, Groen AK, Kuipers F. A role of the bile salt receptor FXR in
atherosclerosis. Arterioscler Thromb Vasc Biol 30: 1519 1528, 2010.
1503
659. Grimsrud PA, Xie H, Griffin TJ, Bernlohr DA. Oxidative stress and covalent modification of protein with bioactive aldehydes. J Biol Chem 283: 2183721841, 2008.
676. Guo Y, Zhang W, Giroux C, Cai Y, Ekambaram P, Dilly AK, Hsu A, Zhou S, Maddipati
KR, Liu J, Joshi S, Tucker SC, Lee MJ, Honn KV. Identification of the orphan G
protein-coupled receptor GPR31 as a receptor for 12-(S)-hydroxyeicosatetraenoic
acid. J Biol Chem 286: 3383233840, 2011.
PAUL N. HOPKINS
691. Haghighat A, Weiss D, Whalin MK, Cowan DP, Taylor WR. Granulocyte colonystimulating factor and granulocyte macrophage colony-stimulating factor exacerbate
atherosclerosis in apolipoprotein E-deficient mice. Circulation 115: 2049 2054,
2007.
709. Han Z, Varadharaj S, Giedt RJ, Zweier JL, Szeto HH, Alevriadou BR. Mitochondriaderived reactive oxygen species mediate heme oxygenase-1 expression in sheared
endothelial cells. J Pharmacol Exp Ther 329: 94 101, 2009.
692. Hahn C, Orr AW, Sanders JM, Jhaveri KA, Schwartz MA. The subendothelial extracellular matrix modulates JNK activation by flow. Circ Res 104: 9951003, 2009.
710. Hanniman EA, Lambert G, McCarthy TC, Sinal CJ. Loss of functional farnesoid X
receptor increases atherosclerotic lesions in apolipoprotein E-deficient mice. J Lipid
Res 46: 25952604, 2005.
693. Hahn C, Schwartz MA. Mechanotransduction in vascular physiology and atherogenesis. Nat Rev Mol Cell Biol 10: 53 62, 2009.
711. Hannun YA, Obeid LM. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol 9: 139 150, 2008.
694. Haidar B, Denis M, Marcil M, Krimbou L, Genest J Jr. Apolipoprotein A-I activates
cellular cAMP signaling through the ABCA1 transporter. J Biol Chem 279: 9963
9969, 2004.
712. Hanson AJ, Wallace HA, Freeman TJ, Beauchamp RD, Lee LA, Lee E. XIAP monoubiquitylates Groucho/TLE to promote canonical Wnt signaling. Mol Cell 45: 619
628, 2012.
695. Haigis MC, Yankner BA. The aging stress response. Mol Cell 40: 333344, 2010.
696. Hajra L, Evans AI, Chen M, Hyduk SJ, Collins T, Cybulsky MI. The NF-kappa B signal
transduction pathway in aortic endothelial cells is primed for activation in regions
predisposed to atherosclerotic lesion formation. Proc Natl Acad Sci USA 97: 9052
9057, 2000.
697. Hakonarson H, Thorvaldsson S, Helgadottir A, Gudbjartsson D, Zink F, Andresdottir M, Manolescu A, Arnar DO, Andersen K, Sigurdsson A, Thorgeirsson G, Jonsson
A, Agnarsson U, Bjornsdottir H, Gottskalksson G, Einarsson A, Gudmundsdottir H,
Adalsteinsdottir AE, Gudmundsson K, Kristjansson K, Hardarson T, Kristinsson A,
Topol EJ, Gulcher J, Kong A, Gurney M, Thorgeirsson G, Stefansson K. Effects of a
5-lipoxygenase-activating protein inhibitor on biomarkers associated with risk of
myocardial infarction: a randomized trial. JAMA 293: 22452256, 2005.
699. Haller CA, Smith S, Wen J, Xiao J, Chaikof EL. Abstract 5193: Syndecan-1 is protective in atherosclerosis. Circulation 120: S1066-c, 2009.
700. Halterman JA, Kwon HM, Leitinger N, Wamhoff BR. NFAT5 expression in bone
marrow-derived cells enhances atherosclerosis and drives macrophage migration.
Front Physiol 3: 313, 2012.
701. Hamada N, Miyata M, Eto H, Ikeda Y, Shirasawa T, Akasaki Y, Miyauchi T, Furusho
Y, Nagaki A, Aronow BJ, Tei C. Loss of clusterin limits atherosclerosis in apolipoprotein E-deficient mice via reduced expression of Egr-1 and TNF-alpha. J Atheroscler Thromb 18: 209 216, 2011.
702. Hamers AA, Vos M, Rassam F, Marincovic G, Kurakula K, van Gorp PJ, de Winther
MP, Gijbels MJ, de Waard V, de Vries CJ. Bone marrow-specific deficiency of nuclear
receptor Nur77 enhances atherosclerosis. Circ Res 110: 428 438, 2012.
703. Hamilton JR, Cornelissen I, Mountford JK, Coughlin SR. Atherosclerosis proceeds
independently of thrombin-induced platelet activation in ApoE/ mice. Atherosclerosis 205: 427 432, 2009.
704. Hammad SM, Twal WO, Barth JL, Smith KJ, Saad AF, Virella G, Argraves WS,
Lopes-Virella MF. Oxidized LDL immune complexes and oxidized LDL differentially
affect the expression of genes involved with inflammation and survival in human
U937 monocytic cells. Atherosclerosis 202: 394 404, 2009.
705. Han C, Liu J, Liu X, Li M. Angiotensin II induces C-reactive protein expression
through ERK1/2 and JNK signaling in human aortic endothelial cells. Atherosclerosis
212: 206 212, 2010.
715. Harats D, Shaish A, George J, Mulkins M, Kurihara H, Levkovitz H, Sigal E. Overexpression of 15-lipoxygenase in vascular endothelium accelerates early atherosclerosis in LDL receptor-deficient mice. Arterioscler Thromb Vasc Biol 20: 2100 2105,
2000.
716. Harb D, Bujold K, Febbraio M, Sirois MG, Ong H, Marleau S. The role of the
scavenger receptor CD36 in regulating mononuclear phagocyte trafficking to atherosclerotic lesions and vascular inflammation. Cardiovasc Res 83: 4251, 2009.
717. Harismendy O, Notani D, Song X, Rahim NG, Tanasa B, Heintzman N, Ren B, Fu
XD, Topol EJ, Rosenfeld MG, Frazer KA. 9p21 DNA variants associated with coronary artery disease impair interferon-gamma signalling response. Nature 470: 264
268, 2011.
718. Harja E, Bu DX, Hudson BI, Chang JS, Shen X, Hallam K, Kalea AZ, Lu Y, Rosario RH,
Oruganti S, Nikolla Z, Belov D, Lalla E, Ramasamy R, Yan SF, Schmidt AM. Vascular
and inflammatory stresses mediate atherosclerosis via RAGE and its ligands in
apoE/ mice. J Clin Invest 118: 183194, 2008.
719. Harja E, Bucciarelli LG, Lu Y, Stern DM, Zou YS, Schmidt AM, Yan SF. Early growth
response-1 promotes atherogenesis: mice deficient in early growth response-1 and
apolipoprotein E display decreased atherosclerosis and vascular inflammation. Circ
Res 94: 333339, 2004.
720. Harja E, Chang JS, Lu Y, Leitges M, Zou YS, Schmidt AM, Yan SF. Mice deficient in
PKC and apolipoprotein E display decreased atherosclerosis. FASEB J 23: 1081
1091, 2009.
721. Harkewicz R, Hartvigsen K, Almazan F, Dennis EA, Witztum JL, Miller YI. Cholesteryl ester hydroperoxides are biologically active components of minimally oxidized
low density lipoprotein. J Biol Chem 283: 1024110251, 2008.
722. Harrington SC, Simari RD, Conover CA. Genetic deletion of pregnancy-associated
plasma protein-A is associated with resistance to atherosclerotic lesion development
in apolipoprotein E-deficient mice challenged with a high-fat diet. Circ Res 100:
1696 1702, 2007.
723. Harris TA, Yamakuchi M, Ferlito M, Mendell JT, Lowenstein CJ. MicroRNA-126
regulates endothelial expression of vascular cell adhesion molecule 1. Proc Natl Acad
Sci USA 105: 1516 1521, 2008.
706. Han KH, Tangirala RK, Green SR, Quehenberger O. Chemokine receptor CCR2
expression and monocyte chemoattractant protein-1-mediated chemotaxis in human monocytes. A regulatory role for plasma LDL. Arterioscler Thromb Vasc Biol 18:
19831991, 1998.
724. Harrison CM, Pompilius M, Pinkerton KE, Ballinger SW. Mitochondrial oxidative
stress significantly influences atherogenic risk and cytokine-induced oxidant production. Environ Health Perspect 119: 676 681, 2011.
725. Harrison M, Smith E, Ross E, Krams R, Segers D, Buckley CD, Nash GB, Rainger GE.
The role of platelet-endothelial cell adhesion molecule-1 in atheroma formation
varies depending on the site-specific hemodynamic environment. Arterioscler
Thromb Vasc Biol 33: 694 701, 2013.
708. Han Z, Chen YR, Jones CI, 3rd Meenakshisundaram G, Zweier JL, Alevriadou BR.
Shear-induced reactive nitrogen species inhibit mitochondrial respiratory complex
activities in cultured vascular endothelial cells. Am J Physiol Cell Physiol 292: C1103
C1112, 2007.
726. Harry BL, Sanders JM, Feaver RE, Lansey M, Deem TL, Zarbock A, Bruce AC, Pryor
AW, Gelfand BD, Blackman BR, Schwartz MA, Ley K. Endothelial cell PECAM-1
promotes atherosclerotic lesions in areas of disturbed flow in apoE-deficient mice.
Arterioscler Thromb Vasc Biol 28: 20032008, 2008.
1504
714. Hara H, Ishihara C, Takeuchi A, Imanishi T, Xue L, Morris SW, Inui M, Takai T,
Shibuya A, Saijo S, Iwakura Y, Ohno N, Koseki H, Yoshida H, Penninger JM, Saito T.
The adaptor protein CARD9 is essential for the activation of myeloid cells through
ITAM-associated and Toll-like receptors. Nat Immunol 8: 619 629, 2007.
746. Hemdahl AL, Falk E, Thoren P, Hansson GK. Thrombin inhibitor reduces myocardial
infarction in apoE/ LDLR/ mice. Am J Physiol Heart Circ Physiol 287: H872
H877, 2004.
728. Hartvigsen K, Chou MY, Hansen LF, Shaw PX, Tsimikas S, Binder CJ, Witztum JL.
The role of innate immunity in atherogenesis. J Lipid Res 50: S388 393, 2009.
735. Haver VG, Slart RH, Zeebregts CJ, Peppelenbosch MP, Tio RA. Rupture of vulnerable atherosclerotic plaques: microRNAs conducting the orchestra? Trends Cardiovasc Med 20: 6571, 2010.
736. Hayden MS, Ghosh S. Shared principles in NF-kappaB signaling. Cell 132: 344 362,
2008.
737. Hayek T, Pavlotzky E, Hamoud S, Coleman R, Keidar S, Aviram M, Kaplan M. Tissue
angiotensin-converting-enzyme (ACE) deficiency leads to a reduction in oxidative
stress and in atherosclerosis: studies in ACE-knockout mice type 2. Arterioscler
Thromb Vasc Biol 23: 2090 2096, 2003.
738. Hayes JD, Flanagan JU, Jowsey IR. Glutathione transferases. Annu Rev Pharmacol
Toxicol 45: 51 88, 2005.
739. Hazen SL. Oxidized phospholipids as endogenous pattern recognition ligands in
innate immunity. J Biol Chem 283: 1552715531, 2008.
749. Heo KS, Chang E, Le NT, Cushman H, Yeh ETH, Fujiwara K, Abe Ji. De-SUMOylation enzyme of sentrin/SUMO-specific protease 2 regulates disturbed flow-induced
SUMOylation of ERK5 and p53 that leads to endothelial dysfunction and atherosclerosis. Circ Res 112: 911923, 2013.
750. Heo KS, Lee H, Nigro P, Thomas T, Le NT, Chang E, McClain C, Reinhart-King
CA, King MR, Berk BC, Fujiwara K, Woo CH, Abe J. PKC mediates disturbed
flow-induced endothelial apoptosis via p53 SUMOylation. J Cell Biol 193: 867
884, 2011.
751. Herbin O, Ait-Oufella H, Yu W, Fredrikson GN, Aubier B, Perez N, Barateau V,
Nilsson J, Tedgui A, Mallat Z. Regulatory T-cell response to apolipoprotein B100derived peptides reduces the development and progression of atherosclerosis in
mice. Arterioscler Thromb Vasc Biol 32: 605 612, 2012.
752. Hergenreider E, Boon RA, Heydt S, Treguer K, Bottger T, Horrevoets AJ, Zeiher
AM, Braun T, Urbich C, Dimmeler S. Abstract 11676: atheroprotective communication between endothelial cells and smooth muscle cells via KLF2-dependent enrichment of miRNAs in microvesicles. Circulation 124: A11676, 2011.
753. Herijgers N, Van Eck M, Groot PH, Hoogerbrugge PM, Van Berkel TJ. Effect of bone
marrow transplantation on lipoprotein metabolism and atherosclerosis in LDL receptor-knockout mice. Arterioscler Thromb Vasc Biol 17: 19952003, 1997.
754. Herijgers N, Van Eck M, Groot PH, Hoogerbrugge PM, Van Berkel TJ. Low density
lipoprotein receptor of macrophages facilitates atherosclerotic lesion formation in
C57Bl/6 mice. Arterioscler Thromb Vasc Biol 20: 19611967, 2000.
755. Hermansson A, Johansson DK, Ketelhuth DF, Andersson J, Zhou X, Hansson GK.
Immunotherapy with tolerogenic apolipoprotein B-100-loaded dendritic cells attenuates atherosclerosis in hypercholesterolemic mice. Circulation 123: 10831091,
2011.
756. Hermansson A, Ketelhuth DF, Strodthoff D, Wurm M, Hansson EM, Nicoletti A,
Paulsson-Berne G, Hansson GK. Inhibition of T cell response to native low-density
lipoprotein reduces atherosclerosis. J Exp Med 207: 10811093, 2010.
757. Hernandez-Vargas P, Ortiz-Munoz G, Lopez-Franco O, Suzuki Y, Gallego-Delgado
J, Sanjuan G, Lazaro A, Lopez-Parra V, Ortega L, Egido J, Gomez-Guerrero C.
Fcgamma receptor deficiency confers protection against atherosclerosis in apolipoprotein E knockout mice. Circ Res 99: 1188 1196, 2006.
740. Hazen SL, Heinecke JW. 3-Chlorotyrosine, a specific marker of myeloperoxidasecatalyzed oxidation, is markedly elevated in low density lipoprotein isolated from
human atherosclerotic intima. J Clin Invest 99: 20752081, 1997.
759. Hess CN, Kou R, Johnson RP, Li GK, Michel T. ADP signaling in vascular endothelial
cells: ADP-dependent activation of the endothelial isoform of nitric-oxide synthase
requires the expression but not the kinase activity of AMP-activated protein kinase.
J Biol Chem 284: 32209 32224, 2009.
742. Heinecke JW. The protein cargo of HDL: implications for vascular wall biology and
therapeutics. J Clin Lipidol 4: 371375, 2010.
760. Hess D, Chisholm JW, Igal RA. Inhibition of stearoylCoA desaturase activity blocks
cell cycle progression and induces programmed cell death in lung cancer cells. PLoS
ONE 5: e11394, 2010.
743. Heitzeneder S, Seidel M, Forster-Waldl E, Heitger A. Mannan-binding lectin deficiency: good news, bad news, doesnt matter? Clin Immunol 143: 2238, 2012.
744. Heller EA, Liu E, Tager AM, Sinha S, Roberts JD, Koehn SL, Libby P, Aikawa ER, Chen
JQ, Huang P, Freeman MW, Moore KJ, Luster AD, Gerszten RE. Inhibition of atherogenesis in BLT1-deficient mice reveals a role for LTB4 and BLT1 in smooth muscle
cell recruitment. Circulation 112: 578 586, 2005.
745. Heller EA, Liu E, Tager AM, Yuan Q, Lin AY, Ahluwalia N, Jones K, Koehn SL, Lok
VM, Aikawa E, Moore KJ, Luster AD, Gerszten RE. Chemokine CXCL10 promotes
atherogenesis by modulating the local balance of effector and regulatory T cells.
Circulation 113: 23012312, 2006.
761. Hibino H, Inanobe A, Furutani K, Murakami S, Findlay I, Kurachi Y. Inwardly rectifying potassium channels: their structure, function, and physiological roles. Physiol
Rev 90: 291366, 2010.
762. Higashi Y, Shai Sy Sukhanov S, Kim C, Delafontaine P. Abstract 10717: monocyte/
macrophage specific knockout of insulin-like growth factor-1 receptor promotes
atherosclerosis and induces an unstable plaque phenotype. Circulation 124: A10717,
2011.
763. Higashi Y, Sukhanov S, Anwar A, Shai SY, Delafontaine P. IGF-1, oxidative stress and
atheroprotection. Trends Endocrinol Metab 21: 245254, 2010.
1505
734. Hauer AD, van Puijvelde GH, Peterse N, de Vos P, van Weel V, van Wanrooij EJ,
Biessen EA, Quax PH, Niethammer AG, Reisfeld RA, van Berkel TJ, Kuiper J. Vaccination against VEGFR2 attenuates initiation and progression of atherosclerosis. Arterioscler Thromb Vasc Biol 27: 2050 2057, 2007.
748. Henning MF, Herlax V, Bakas L. Contribution of the C-terminal end of apolipoprotein AI to neutralization of lipopolysaccharide endotoxic effect. Innate Immunity 17:
327337, 2011.
PAUL N. HOPKINS
764. Higashimori M, Tatro JB, Moore KJ, Mendelsohn ME, Galper JB, Beasley D. Role of
toll-like receptor 4 in intimal foam cell accumulation in apolipoprotein E-deficient
mice. Arterioscler Thromb Vasc Biol 31: 50 57, 2011.
782. Hopkins PN, Nanjee MN, Wu LL, McGinty MG, Brinton EA, Hunt SC, Anderson JL.
Altered composition of triglyceride-rich lipoproteins and coronary artery disease in
a large case-control study. Atherosclerosis 207: 559 566, 2009.
783. Hopkins PN, Stephenson S, Wu LL, Riley WA, Xin Y, Hunt SC. Evaluation of coronary risk factors in patients with heterozygous familial hypercholesterolemia. Am J
Cardiol 87: 547553, 2001.
784. Hopkins PN, Williams RR. A survey of 246 suggested coronary risk factors. Atherosclerosis 40: 152, 1981.
785. Hopkins PN, Wu LL, Hunt SC, Brinton EA. Plasma triglycerides and type III hyperlipidemia are independently associated with premature familial coronary artery disease. J Am Coll Cardiol 45: 10031012, 2005.
786. Hopkins PN, Wu LL, Schumacher MC, Emi M, Hegele RM, Hunt SC, Lalouel JM,
Williams RR. Type III dyslipoproteinemia in patients heterozygous for familial hypercholesterolemia and apolipoprotein E2. Evidence for a gene-gene interaction. Arterioscler Thromb 11: 11371146, 1991.
769. Hjerpe C, Johansson D, Hermansson A, Hansson GK, Zhou X. Dendritic cells pulsed
with malondialdehyde modified low density lipoprotein aggravate atherosclerosis in
Apoe(/) mice. Atherosclerosis 209: 436 441, 2010.
1506
789. Hu JH, Du L, Chu T, Otsuka G, Dronadula N, Jaffe M, Gill SE, Parks WC, Dichek DA.
Overexpression of urokinase by plaque macrophages causes histological features of
plaque rupture and increases vascular matrix metalloproteinase activity in aged apolipoprotein e-null mice. Circulation 121: 16371644, 2010.
790. Hu L, Boesten LS, May P, Herz J, Bovenschen N, Huisman MV, Berbee JF, Havekes
LM, van Vlijmen BJ, Tamsma JT. Macrophage low-density lipoprotein receptorrelated protein deficiency enhances atherosclerosis in ApoE/LDLR double knockout
mice. Arterioscler Thromb Vasc Biol 26: 2710 2715, 2006.
791. Hu X, Chung AY, Wu I, Foldi J, Chen J, Ji JD, Tateya T, Kang YJ, Han J, Gessler M,
Kageyama R, Ivashkiv LB. Integrated regulation of Toll-like receptor responses by
Notch and interferon-gamma pathways. Immunity 29: 691703, 2008.
792. Hua P, Feng W, Rezonzew G, Chumley P, Jaimes EA. The transcription factor ETS-1
regulates angiotensin II-stimulated fibronectin production in mesangial cells. Am J
Physiol Renal Physiol 302: F1418 F1429, 2012.
793. Huang Q, Qin L, Dai S, Zhang H, Pasula S, Zhou H, Chen H, Min W. AIP1 suppresses
atherosclerosis by limiting hyperlipidemia-induced inflammation and vascular endothelial dysfunction. Arterioscler Thromb Vasc Biol 33: 795 804, 2013.
794. Huang W, Glass CK. Nuclear receptors and inflammation control: molecular mechanisms and pathophysiological relevance. Arterioscler Thromb Vasc Biol 30: 1542
1549, 2010.
795. Huang WC, Chen JJ, Chen CC. c-Src-dependent tyrosine phosphorylation of IKKbeta is involved in tumor necrosis factor-alpha-induced intercellular adhesion molecule-1 expression. J Biol Chem 278: 9944 9952, 2003.
796. Huang X, Chen LY, Doerner AM, Pan WW, Smith L, Huang S, Papadimos TJ, Pan ZK.
An atypical protein kinase C (PKC) plays a critical role in lipopolysaccharide-activated NF-B in human peripheral blood monocytes and macrophages. J Immunol
182: 5810 5815, 2009.
797. Huang ZL, Qu WM, Eguchi N, Chen JF, Schwarzschild MA, Fredholm BB, Urade Y,
Hayaishi O. Adenosine A2A, but not A1, receptors mediate the arousal effect of
caffeine. Nat Neurosci 8: 858 859, 2005.
798. Huber SA, Sakkinen P, David C, Newell MK, Tracy RP. T helper-cell phenotype
regulates atherosclerosis in mice under conditions of mild hypercholesterolemia.
Circulation 103: 2610 2616, 2001.
799. Huddleson JP, Ahmad N, Srinivasan S, Lingrel JB. Induction of KLF2 by fluid shear
stress requires a novel promoter element activated by a phosphatidylinositol 3-kinase-dependent chromatin-remodeling pathway. J Biol Chem 280: 2337123379,
2005.
770. Ho FM, Liu SH, Liau CS, Huang PJ, Lin-Shiau SY. High glucose-induced apoptosis in
human endothelial cells is mediated by sequential activations of c-Jun NH2-terminal
kinase and caspase-3. Circulation 101: 2618 2624, 2000.
821. Imanishi T, Han DK, Hofstra L, Hano T, Nishio I, Liles WC, Gown AM, Schwartz SM.
Apoptosis of vascular smooth muscle cells is induced by Fas ligand derived from
monocytes/macrophage. Atherosclerosis 161: 143151, 2002.
822. Imbert V, Rupec RA, Livolsi A, Pahl HL, Traenckner EB, Mueller-Dieckmann C,
Farahifar D, Rossi B, Auberger P, Baeuerle PA, Peyron JF. Tyrosine phosphorylation
of I kappa B-alpha activates NF-kappa B without proteolytic degradation of I kappa
B-alpha. Cell 86: 787798, 1996.
803. Humphries JD, Byron A, Humphries MJ. Integrin ligands at a glance. J Cell Sci 119:
39013903, 2006.
804. Hunt SC, Hopkins PN, Williams RR. Hypertension: genetics and mechanisms. In:
Atherosclerosis and Coronary Artery Disease, edited by Fuster V, Ross R, and Topol EJ.
Philadelphia, PA: Lippincott-Raven, 1996, p. 209 235.
805. Hunyady L, Catt KJ. Pleiotropic AT1 receptor signaling pathways mediating physiological and pathogenic actions of angiotensin II. Mol Endocrinol 20: 953970, 2006.
806. Huo Y, Schober A, Forlow SB, Smith DF, Hyman MC, Jung S, Littman DR, Weber C,
Ley K. Circulating activated platelets exacerbate atherosclerosis in mice deficient in
apolipoprotein E. Nat Med 9: 61 67, 2003.
808. Hutchins PM, Murphy RC. Cholesteryl ester acyl oxidation and remodeling in murine
macrophages: formation of oxidized phosphatidylcholine. J Lipid Res 53: 1588 1597,
2012.
827. Ionita MG, van den Borne P, Catanzariti LM, Moll FL, de Vries JP, Pasterkamp G, Vink
A, de Kleijn DP. High neutrophil numbers in human carotid atherosclerotic plaques
are associated with characteristics of rupture-prone lesions. Arterioscler Thromb Vasc
Biol 30: 18421848, 2010.
809. Huyer G, Liu S, Kelly J, Moffat J, Payette P, Kennedy B, Tsaprailis G, Gresser MJ,
Ramachandran C. Mechanism of inhibition of protein-tyrosine phosphatases by vanadate and pervanadate. J Biol Chem 272: 843 851, 1997.
810. Hwa JS, Mun L, Kim HJ, Seo HG, Lee JH, Kwak JH, Lee DU, Chang KC. Genipin
selectively inhibits TNF-alpha-activated VCAM-1 but not ICAM-1 expression by
upregulation of PPAR-gamma in human endothelial cells. Korean J Physiol Pharmacol
15: 157162, 2011.
828. Ishida T, Choi SY, Kundu RK, Spin J, Yamashita T, Hirata K, Kojima Y, Yokoyama M,
Cooper AD, Quertermous T. Endothelial lipase modulates susceptibility to atherosclerosis in apolipoprotein-E-deficient mice. J Biol Chem 279: 45085 45092, 2004.
829. Ishigaki Y, Katagiri H, Gao J, Yamada T, Imai J, Uno K, Hasegawa Y, Kaneko K,
Ogihara T, Ishihara H, Sato Y, Takikawa K, Nishimichi N, Matsuda H, Sawamura T,
Oka Y. Impact of plasma oxidized low-density lipoprotein removal on atherosclerosis. Circulation 118: 75 83, 2008.
811. Hwang JW, Rajendrasozhan S, Yao H, Chung S, Sundar IK, Huyck HL, Pryhuber GS,
Kinnula VL, Rahman I. FOXO3 deficiency leads to increased susceptibility to cigarette smoke-induced inflammation, airspace enlargement, and chronic obstructive
pulmonary disease. J Immunol 187: 987998, 2011.
812. Hyman MC, Mazer SP, Yanamadala S, Neral MK, Bouis D, Petrovic-Djergovic D,
Broekman MJ, Marcus AJ, Pinsky DJ. Abstract 624: ecto-enzymatic suppression of
atherogenesis by CD39. Circulation 116: II-114, 2007.
831. Ito J, Nagayasu Y, Ueno S, Yokoyama S. Apolipoprotein-mediated cellular lipid release requires replenishment of sphingomyelin in a phosphatidylcholine-specific
phospholipase C-dependent manner. J Biol Chem 277: 44709 44714, 2002.
832. Ito S, Osaka M, Higuchi Y, Nishijima F, Ishii H, Yoshida M. Indoxyl sulfate induces
leukocyte-endothelial interactions through up-regulation of E-selectin. J Biol Chem
285: 38869 38875, 2010.
833. Ivandic B, Castellani LW, Wang XP, Qiao JH, Mehrabian M, Navab M, Fogelman AM,
Grass DS, Swanson ME, de Beer MC, de Beer F, Lusis AJ. Role of group II secretory
phospholipase A2 in atherosclerosis: 1. Increased atherogenesis and altered lipoproteins in transgenic mice expressing group IIa phospholipase A2. Arterioscler Thromb
Vasc Biol 19: 1284 1290, 1999.
834. Ivashkiv LB. Cross-regulation of signaling by ITAM-associated receptors. Nat Immunol 10: 340 347, 2009.
835. Iwaki T, Sandoval-Cooper MJ, Brechmann M, Ploplis VA, Castellino FJ. A fibrinogen
deficiency accelerates the initiation of LDL cholesterol-driven atherosclerosis via
thrombin generation and platelet activation in genetically predisposed mice. Blood
107: 38833891, 2006.
836. Iwasaki A, Medzhitov R. Regulation of adaptive immunity by the innate immune
system. Science 327: 291295, 2010.
818. Ihrig M, Dangler CA, Fox JG. Mice lacking inducible nitric oxide synthase develop
spontaneous hypercholesterolaemia and aortic atheromas. Atherosclerosis 156: 103
107, 2001.
837. Jackson CL, Bennett MR, Biessen EA, Johnson JL, Krams R. Assessment of unstable
atherosclerosis in mice. Arterioscler Thromb Vasc Biol 27: 714 720, 2007.
819. Im SS, Osborne TF. Liver X receptors in atherosclerosis and inflammation. Circ Res
108: 996 1001, 2011.
838. Jackson SM, Parhami F, Xi XP, Berliner JA, Hsueh WA, Law RE, Demer LL. Peroxisome proliferator-activated receptor activators target human endothelial cells to
1507
807. Huo Y, Zhao L, Hyman MC, Shashkin P, Harry BL, Burcin T, Forlow SB, Stark MA,
Smith DF, Clarke S, Srinivasan S, Hedrick CC, Pratico D, Witztum JL, Nadler JL, Funk
CD, Ley K. Critical role of macrophage 12/15-lipoxygenase for atherosclerosis in
apolipoprotein E-deficient mice. Circulation 110: 2024 2031, 2004.
PAUL N. HOPKINS
inhibit leukocyte-endothelial cell interaction. Arterioscler Thromb Vasc Biol 19: 2094
2104, 1999.
839. Jaeschke A, Davis RJ. Metabolic stress signaling mediated by mixed-lineage kinases.
Mol Cell 27: 498 508, 2007.
840. Jagavelu K, Tietge UJ, Gaestel M, Drexler H, Schieffer B, Bavendiek U. Systemic
deficiency of the MAP kinase-activated protein kinase 2 reduces atherosclerosis in
hypercholesterolemic mice. Circ Res 101: 1104 1112, 2007.
841. Jakus Z, Fodor S, Abram CL, Lowell CA, Mocsai A. Immunoreceptor-like signaling by
beta 2 and beta 3 integrins. Trends Cell Biol 17: 493501, 2007.
842. Jamaluddin M, Wang S, Boldogh I, Tian B, Brasier AR. TNF-alpha-induced NFkappaB/RelA Ser(276) phosphorylation and enhanceosome formation is mediated
by an ROS-dependent PKAc pathway. Cell Signal 19: 1419 1433, 2007.
843. Janardhanan R, Henry Z, Hur DJ, Lin CM, Lopez D, Reagan PM, Rudnick SR, Koshko
TJ, Keeley EC. The snow-shovelers ST elevation myocardial infarction. Am J Cardiol
106: 596 600, 2010.
844. Janeway CA Jr. Approaching the asymptote? Evolution and revolution in immunology. Cold Spring Harb Symp Quant Biol 54: 113, 1989.
845. Jawien J, Gajda M, Rudling M, Mateuszuk L, Olszanecki R, Guzik TJ, Cichocki T,
Chlopicki S, Korbut R. Inhibition of five lipoxygenase activating protein (FLAP) by
MK-886 decreases atherosclerosis in apoE/LDLR-double knockout mice. Eur J Clin
Invest 36: 141146, 2006.
847. Jeon HJ, Choi JH, Jung IH, Park JG, Lee MR, Lee MN, Kim B, Yoo JY, Jeong SJ, Kim
DY, Park JE, Park HY, Kwack K, Choi BK, Kwon BS, Oh GT. CD137 (4 1BB)
deficiency reduces atherosclerosis in hyperlipidemic mice. Circulation 121: 1124
1133, 2010.
848. Jeong Y, Chaupin DF, Matsushita K, Yamakuchi M, Cameron SJ, Morrell CN, Lowenstein CJ. Aldosterone activates endothelial exocytosis. Proc Natl Acad Sci USA 106:
37823787, 2009.
849. Jersmann HP, Hii CS, Hodge GL, Ferrante A. Synthesis and surface expression of
CD14 by human endothelial cells. Infect Immun 69: 479 485, 2001.
850. Jespersen CM, Als-Nielsen B, Damgaard M, Hansen JF, Hansen S, Helo OH, Hildebrandt P, Hilden J, Jensen GB, Kastrup J, Kolmos HJ, Kjoller E, Lind I, Nielsen H,
Petersen L, Gluud C. Randomised placebo controlled multicentre trial to assess
short term clarithromycin for patients with stable coronary heart disease: CLARICOR trial. BMJ 332: 2227, 2006.
851. Jia L, Wang J, Wang X, Chen J, Zhang C, Hui R. Abstract 8783: methylation of FOXP3
In regulatory T cells is related to the severity of coronary atherosclerosis. Circulation
124: A8783, 2011.
852. Jiang XC, Qin S, Qiao C, Kawano K, Lin M, Skold A, Xiao X, Tall AR. Apolipoprotein
B secretion and atherosclerosis are decreased in mice with phospholipid-transfer
protein deficiency. Nat Med 7: 847 852, 2001.
853. Jin X, Jin HR, Jung HS, Lee SJ, Lee JH, Lee JJ. An atypical E3 ligase zinc finger protein
91 stabilizes and activates NF-kappaB-inducing kinase via Lys63-linked ubiquitination. J Biol Chem 285: 30539 30547, 2010.
854. Jin ZG, Wong C, Wu J, Berk BC. Flow shear stress stimulates Gab1 tyrosine phosphorylation to mediate protein kinase B and endothelial nitric-oxide synthase activation in endothelial cells. J Biol Chem 280: 1230512309, 2005.
855. Jo H, Song H, Mowbray A. Role of NADPH oxidases in disturbed flow- and BMP4induced inflammation and atherosclerosis. Antioxid Redox Signal 8: 1609 1619, 2006.
856. Johnson AD, Hwang SJ, Voorman A, Morrison A, Peloso GM, Hu YH, Thanassoulis
G, Newton-Cheh C, Rogers IS, Hoffman U, Freedman JE, Fox CS, Psaty BM, Boerwinkle E, Cupples LA, ODonnell CJ. Resequencing and clinical associations of the
9p21.3 region: a comprehensive investigation in the Framingham Heart Study. Circulation 127: 799 810, 2013.
857. Johnson J, Carson K, Williams H, Karanam S, Newby A, Angelini G, George S, Jackson
C. Plaque rupture after short periods of fat feeding in the apolipoprotein E-knockout
mouse: model characterization and effects of pravastatin treatment. Circulation 111:
14221430, 2005.
1508
859. Johnson JL, Devel L, Czarny B, George SJ, Jackson CL, Rogakos V, Beau F, Yiotakis A,
Newby AC, Dive V. A selective matrix metalloproteinase-12 inhibitor retards atherosclerotic plaque development in apolipoprotein E-knockout mice. Arterioscler
Thromb Vasc Biol 31: 528 535, 2011.
860. Johnson JL, George SJ, Newby AC, Jackson CL. Divergent effects of matrix metalloproteinases 3, 7, 9, and 12 on atherosclerotic plaque stability in mouse brachiocephalic arteries. Proc Natl Acad Sci USA 102: 1557515580, 2005.
861. Johnson JM, Castle J, Garrett-Engele P, Kan Z, Loerch PM, Armour CD, Santos R,
Schadt EE, Stoughton R, Shoemaker DD. Genome-wide survey of human alternative
pre-mRNA splicing with exon junction microarrays. Science 302: 21412144, 2003.
862. Johnson RC, Chapman SM, Dong ZM, Ordovas JM, Mayadas TN, Herz J, Hynes RO,
Schaefer EJ, Wagner DD. Absence of P-selectin delays fatty streak formation in mice.
J Clin Invest 99: 10371043, 1997.
863. Jones CA, Nishiya N, London NR, Zhu W, Sorensen LK, Chan AC, Lim CJ, Chen H,
Zhang Q, Schultz PG, Hayallah AM, Thomas KR, Famulok M, Zhang K, Ginsberg MH,
Li DY. Slit2-Robo4 signalling promotes vascular stability by blocking Arf6 activity. Nat
Cell Biol 11: 13251331, 2009.
864. Jongstra-Bilen J, Haidari M, Zhu SN, Chen M, Guha D, Cybulsky MI. Low-grade
chronic inflammation in regions of the normal mouse arterial intima predisposed to
atherosclerosis. J Exp Med 203: 20732083, 2006.
865. Judge DP, Biery NJ, Keene DR, Geubtner J, Myers L, Huso DL, Sakai LY, Dietz HC.
Evidence for a critical contribution of haploinsufficiency in the complex pathogenesis
of Marfan syndrome. J Clin Invest 114: 172181, 2004.
866. Judkins CP, Diep H, Broughton BRS, Mast AE, Hooker EU, Miller AA, Selemidis S,
Dusting GJ, Sobey CG, Drummond GR. Direct evidence of a role for Nox2 in
superoxide production, reduced nitric oxide bioavailability, and early atherosclerotic
plaque formation in ApoE/ mice. Am J Physiol Heart Circ Physiol 298: H24 H32,
2010.
867. Jun JY, Ma Z, Segar L. Spontaneously diabetic Ins2(/Akita):apoE-deficient mice
exhibit exaggerated hypercholesterolemia and atherosclerosis. Am J Physiol Endocrinol Metab 301: E145E154, 2011.
868. Jung KJ, Lee EK, Yu BP, Chung HY. Significance of protein tyrosine kinase/protein
tyrosine phosphatase balance in the regulation of NF-kappaB signaling in the inflammatory process and aging. Free Radic Biol Med 47: 983991, 2009.
869. Jung SB, Kim CS, Naqvi A, Yamamori T, Mattagajasingh I, Hoffman TA, Cole MP,
Kumar A, Dericco JS, Jeon BH, Irani K. Histone deacetylase 3 antagonizes aspirinstimulated endothelial nitric oxide production by reversing aspirin-induced lysine
acetylation of endothelial nitric oxide synthase. Circ Res 107: 877 887, 2010.
870. Jung Y, Kim H, Min SH, Rhee SG, Jeong W. Dynein light chain LC8 negatively
regulates NF-B through the redox-dependent interaction with IB. J Biol Chem
283: 2386323871, 2008.
871. Jyrkkanen HK, Kansanen E, Inkala M, Kivela AM, Hurttila H, Heinonen SE, Goldsteins
G, Jauhiainen S, Tiainen S, Makkonen H, Oskolkova O, Afonyushkin T, Koistinaho J,
Yamamoto M, Bochkov VN, Yla-Herttuala S, Levonen AL. Nrf2 regulates antioxidant
gene expression evoked by oxidized phospholipids in endothelial cells and murine
arteries in vivo. Circ Res 103: e19, 2008.
872. Kadl A, Meher AK, Sharma PR, Lee MY, Doran AC, Johnstone SR, Elliott MR, Gruber
F, Han J, Chen W, Kensler T, Ravichandran KS, Isakson BE, Wamhoff BR, Leitinger N.
Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circ Res 107: 737746, 2010.
873. Kakiashvili E, Speight P, Waheed F, Seth R, Lodyga M, Tanimura S, Kohno M, Rotstein OD, Kapus A, Szaszi K. GEF-H1 mediates tumor necrosis factor-alpha-induced
Rho activation and myosin phosphorylation: role in the regulation of tubular paracellular permeability. J Biol Chem 284: 11454 11466, 2009.
874. Kales SN, Soteriades ES, Christophi CA, Christiani DC. Emergency duties and
deaths from heart disease among firefighters in the United States. N Engl J Med 356:
12071215, 2007.
846. Jenkins SJ, Ruckerl D, Cook PC, Jones LH, Finkelman FD, van Rooijen N, MacDonald
AS, Allen JE. Local macrophage proliferation, rather than recruitment from the
blood, is a signature of TH2 inflammation. Science 332: 1284 1288, 2011.
858. Johnson JL, Baker AH, Oka K, Chan L, Newby AC, Jackson CL, George SJ. Suppression of atherosclerotic plaque progression and instability by tissue inhibitor of metalloproteinase-2: involvement of macrophage migration and apoptosis. Circulation
113: 24352444, 2006.
883. Karper JC, de Jager SC, Ewing M, Jukema JW, Kuiper J, Quax PH. Abstract 14685:
RP105, a cell surface TLR4 signaling regulator, enhances atherosclerotic plaque
formation. Circulation 124: A14685, 2011.
884. Karra R, Vemullapalli S, Dong C, Herderick EE, Song X, Slosek K, Nevins JR, West M,
Goldschmidt-Clermont PJ, Seo D. Molecular evidence for arterial repair in atherosclerosis. Proc Natl Acad Sci USA 102: 16789 16794, 2005.
885. Karwatsky J, Ma L, Dong F, Zha X. Cholesterol efflux to apoA-I in ABCA1-expressing
cells is regulated by Ca2-dependent calcineurin signaling. J Lipid Res 51: 1144 1156,
2010.
886. Kasuga H, Hosogane N, Matsuoka K, Mori I, Sakura Y, Shimakawa K, Shinki T, Suda
T, Taketomi S. Characterization of transgenic rats constitutively expressing vitamin
D-24-hydroxylase gene. Biochem Biophys Res Commun 297: 13321338, 2002.
887. Kasza Z, Fetalvero KM, Ding M, Wagner RJ, Acs K, Guzman AK, Douville KL, Powell
RJ, Hwa J, Martin KA. Novel signaling pathways promote a paracrine wave of prostacyclin-induced vascular smooth muscle differentiation. J Mol Cell Cardiol 46: 682
694, 2009.
888. Katakami N, Kim YS, Kawamori R, Yamasaki Y. The phosphodiesterase inhibitor
cilostazol induces regression of carotid atherosclerosis in subjects with type 2 diabetes mellitus: principal results of the Diabetic Atherosclerosis Prevention by
Cilostazol (DAPC) study: a randomized trial. Circulation 121: 2584 2591, 2010.
889. Kato H, Ishida J, Nagano K, Honjo K, Sugaya T, Takeda N, Sugiyama F, Yagami K,
Fujita T, Nangaku M, Fukamizu A. Deterioration of atherosclerosis in mice lacking
angiotensin II type 1A receptor in bone marrow-derived cells. Lab Invest 88: 731
739, 2008.
893. Keller TT, van Leuven SI, Meuwese MC, Wareham NJ, Luben R, Stroes ES, Hack CE,
Levi M, Khaw KT, Boekholdt SM. Serum levels of mannose-binding lectin and the risk
of future coronary artery disease in apparently healthy men and women. Arterioscler
Thromb Vasc Biol 26: 23452350, 2006.
894. Kelly EK, Wang L, Ivashkiv LB. Calcium-activated pathways and oxidative burst
mediate zymosan-induced signaling and IL-10 production in human macrophages. J
Immunol 184: 55455552, 2010.
895. Kelly JA, Griffin ME, Fava RA, Wood SG, Bessette KA, Miller ER, Huber SA, Binder
CJ, Witztum JL, Morganelli PM. Inhibition of arterial lesion progression in CD16deficient mice: evidence for altered immunity and the role of IL-10. Cardiovasc Res
85: 224 231, 2010.
896. Keramati AR, Singh R, Lin A, Faramarzi S, Ye ZJ, Mane S, Tellides G, Lifton RP, Mani
A. Wild-type LRP6 inhibits, whereas atherosclerosis-linked LRP6R611C increases
PDGF-dependent vascular smooth muscle cell proliferation. Proc Natl Acad Sci USA
108: 1914 1918, 2011.
897. Ketelhuth DF, Rios FJ, Wang Y, Liu H, Johansson ME, Fredrikson GN, Hedin U,
Gidlund M, Nilsson J, Hansson GK, Yan ZQ. Identification of a danger-associated
peptide from apolipoprotein B100 (ApoBDS-1) that triggers innate proatherogenic
responses. Circulation 124: 24332443, 2011.
898. Keul P, Lucke S, von Wnuck Lipinski K, Bode C, Graler M, Heusch G, Levkau B.
Sphingosine-1-phosphate receptor 3 promotes recruitment of monocyte/macrophages in inflammation and atherosclerosis. Circ Res 108: 314 323, 2011.
899. Khallou-Laschet J, Caligiuri G, Tupin E, Gaston AT, Poirier B, Groyer E, Urbain D,
Maisnier-Patin S, Sarkar R, Kaveri SV, Lacroix-Desmazes S, Nicoletti A. Role of the
intrinsic coagulation pathway in atherogenesis assessed in hemophilic apolipoprotein
E knockout mice. Arterioscler Thromb Vasc Biol 25: e123126, 2005.
900. Khallou-Laschet J, Varthaman A, Fornasa G, Compain C, Gaston AT, Clement M,
Dussiot M, Levillain O, Graff-Dubois S, Nicoletti A, Caligiuri G. Macrophage plasticity in experimental atherosclerosis. PLoS ONE 5: e8852, 2010.
901. Khan JA, Cao M, Kang BY, Liu Y, Mehta JL, Hermonat PL. AAV/hSTAT3-gene delivery lowers aortic inflammatory cell infiltration in LDLR KO mice on high cholesterol.
Atherosclerosis 213: 59 66, 2010.
902. Khera AV, Cuchel M, de la Llera-Moya M, Rodrigues A, Burke MF, Jafri K, French BC,
Phillips JA, Mucksavage ML, Wilensky RL, Mohler ER, Rothblat GH, Rader DJ. Cholesterol efflux capacity, high-density lipoprotein function, and atherosclerosis. N Engl
J Med 364: 127135, 2011.
903. Khurana R, Moons L, Shafi S, Luttun A, Collen D, Martin JF, Carmeliet P, Zachary IC.
Placental growth factor promotes atherosclerotic intimal thickening and macrophage accumulation. Circulation 111: 2828 2836, 2005.
904. Khurma V, Meyer C, Park GS, McMahon M, Lin J, Singh RR, Khanna D. A pilot study
of subclinical coronary atherosclerosis in systemic sclerosis: coronary artery calcification in cases and controls. Arthritis Rheum 59: 591597, 2008.
905. Kim F, Pham M, Luttrell I, Bannerman DD, Tupper J, Thaler J, Hawn TR, Raines EW,
Schwartz MW. Toll-like receptor-4 mediates vascular inflammation and insulin resistance in diet-induced obesity. Circ Res 100: 1589 1596, 2007.
906. Kim HB, Kumar A, Wang L, Liu GH, Keller SR, Lawrence JC Jr, Finck BN, Harris TE.
Lipin 1 represses NFATc4 transcriptional activity in adipocytes to inhibit secretion of
inflammatory factors. Mol Cell Biol 30: 3126 3139, 2010.
907. Kim J, Zhang L, Peppel K, Wu JH, Zidar DA, Brian L, DeWire SM, Exum ST, Lefkowitz RJ, Freedman NJ. Beta-arrestins regulate atherosclerosis and neointimal hyperplasia by controlling smooth muscle cell proliferation and migration. Circ Res 103:
70 79, 2008.
908. Kim JB, Deluna A, Mungrue IN, Vu C, Pouldar D, Civelek M, Orozco L, Wu J, Wang
X, Charugundla S, Castellani LW, Rusek M, Jakobowski H, Lusis AJ. Effect of 9p21.3
coronary artery disease locus neighboring genes on atherosclerosis in mice. Circulation 126: 1896 1906, 2012.
909. Kim KY, Ahn JH, Cheon HG. Anti-angiogenic action of PPARgamma ligand in human
umbilical vein endothelial cells is mediated by PTEN upregulation and VEGFR-2
downregulation. Mol Cell Biochem 358: 375385, 2011.
1509
PAUL N. HOPKINS
910. Kimura T, Tomura H, Mogi C, Kuwabara A, Damirin A, Ishizuka T, Sekiguchi A,
Ishiwara M, Im DS, Sato K, Murakami M, Okajima F. Role of scavenger receptor class
B type I and sphingosine 1-phosphate receptors in high density lipoprotein-induced
inhibition of adhesion molecule expression in endothelial cells. J Biol Chem 281:
3745737467, 2006.
911. Kinderlerer AR, Ali F, Johns M, Lidington EA, Leung V, Boyle JJ, Hamdulay SS, Evans
PC, Haskard DO, Mason JC. KLF2-dependent, shear stress-induced expression of
CD59: a novel cytoprotective mechanism against complement-mediated injury in
the vasculature. J Biol Chem 283: 14636 14644, 2008.
912. King SM, McNamee RA, Houng AK, Patel R, Brands M, Reed GL. Platelet densegranule secretion plays a critical role in thrombosis and subsequent vascular remodeling in atherosclerotic mice. Circulation 120: 785791, 2009.
931. Kojima Y, Kundu RK, Cox CM, Leeper NJ, Anderson JA, Chun HJ, Ali ZA, Ashley EA,
Krieg PA, Quertermous T. Upregulation of the apelin-APJ pathway promotes neointima formation in the carotid ligation model in mouse. Cardiovasc Res 87: 156 165,
2010.
913. King VL, Cassis LA, Daugherty A. Interleukin-4 does not influence development of
hypercholesterolemia or angiotensin II-induced atherosclerotic lesions in mice. Am J
Pathol 171: 2040 2047, 2007.
914. Kirk EA, Dinauer MC, Rosen H, Chait A, Heinecke JW, LeBoeuf RC. Impaired
superoxide production due to a deficiency in phagocyte NADPH oxidase fails to
inhibit atherosclerosis in mice. Arterioscler Thromb Vasc Biol 20: 1529 1535, 2000.
933. Kolch W. Coordinating ERK/MAPK signalling through scaffolds and inhibitors. Nat
Rev Mol Cell Biol 6: 827 837, 2005.
915. Kisucka J, Chauhan AK, Patten IS, Yesilaltay A, Neumann C, Van Etten RA, Krieger M,
Wagner DD. Peroxiredoxin1 prevents excessive endothelial activation and early
atherosclerosis. Circ Res 103: 598 605, 2008.
916. Kita Y, Ohto T, Uozumi N, Shimizu T. Biochemical properties and pathophysiological roles of cytosolic phospholipase A2s. Biochim Biophys Acta 1761: 13171322,
2006.
918. Kleemann R, Zadelaar S, Kooistra T. Cytokines and atherosclerosis: a comprehensive review of studies in mice. Cardiovasc Res 79: 360 376, 2008.
919. Kleinbongard P, Heusch G, Schulz R. TNFalpha in atherosclerosis, myocardial ischemia/reperfusion and heart failure. Pharmacol Ther 127: 295314, 2010.
920. Kloner RA, McDonald SA, Leeka J, Poole WK. Role of age, sex, and race on cardiac
and total mortality associated with Super Bowl wins and losses. Clin Cardiol 34:
102107, 2011.
921. Klysik J, Theroux SJ, Sedivy JM, Moffit JS, Boekelheide K. Signaling crossroads: the
function of Raf kinase inhibitory protein in cancer, the central nervous system and
reproduction. Cell Signal 20: 19, 2008.
922. Knight-Lozano CA, Young CG, Burow DL, Hu ZY, Uyeminami D, Pinkerton KE,
Ischiropoulos H, Ballinger SW. Cigarette smoke exposure and hypercholesterolemia
increase mitochondrial damage in cardiovascular tissues. Circulation 105: 849 854,
2002.
923. Knowles JW, Maeda N. Genetic modifiers of atherosclerosis in mice. Arterioscler
Thromb Vasc Biol 20: 2336 2345, 2000.
924. Kobayashi A, Takanezawa Y, Hirata T, Shimizu Y, Misasa K, Kioka N, Arai H, Ueda K,
Matsuo M. Efflux of sphingomyelin, cholesterol, and phosphatidylcholine by ABCG1.
J Lipid Res 47: 17911802, 2006.
925. Kobayashi T, Tahara Y, Matsumoto M, Iguchi M, Sano H, Murayama T, Arai H, Oida
H, Yurugi-Kobayashi T, Yamashita JK, Katagiri H, Majima M, Yokode M, Kita T,
Narumiya S. Roles of thromboxane A2 and prostacyclin in the development of atherosclerosis in apoE-deficient mice. J Clin Invest 114: 784 794, 2004.
926. Kober F, Canault M, Peiretti F, Mueller C, Kopp F, Alessi MC, Cozzone PJ, Nalbone
G, Bernard M. MRI follow-up of TNF-dependent differential progression of atherosclerotic wall-thickening in mouse aortic arch from early to advanced stages. Atherosclerosis 195: e9399, 2007.
927. Kockx MM, De Meyer GR, Buyssens N, Knaapen MW, Bult H, Herman AG. Cell
composition, replication, and apoptosis in atherosclerotic plaques after 6 months of
cholesterol withdrawal. Circ Res 83: 378 387, 1998.
928. Kofranek J, Rusz J. Restoration of Guyton s diagram for regulation of the circulation
as a basis for quantitative physiological model development. Physiol Res 59: 897908,
2010.
1510
935. Kolodgie FD, Virmani R, Burke AP, Farb A, Weber DK, Kutys R, Finn AV, Gold HK.
Pathologic assessment of the vulnerable human coronary plaque. Heart 90: 1385
1391, 2004.
936. Kontush A, Therond P, Zerrad A, Couturier M, Negre-Salvayre A, de Souza JA,
Chantepie S, Chapman MJ. Preferential sphingosine-1-phosphate enrichment and
sphingomyelin depletion are key features of small dense HDL3 particles: relevance
to antiapoptotic and antioxidative activities. Arterioscler Thromb Vasc Biol 27: 1843
1849, 2007.
937. Korporaal SJ, Meurs I, Calpe-Berdiel L, Habets KL, Josselin E, Hildebrand RB, Ye D,
Out R, Van Berkel TJ, Chimini G, Van Eck M. Abstract 20428: the impact of deletion
of ABCA1 and ABCA7 in bone marrow-derived cells on platelet function and atherosclerosis in LDL receptor knockout mice. Circulation 122: A20428, 2010.
938. Korporaal SJ, Meurs I, Hauer AD, Hildebrand RB, Hoekstra M, Cate HT, Pratico D,
Akkerman JW, Van Berkel TJ, Kuiper J, Van Eck M. Deletion of the high-density
lipoprotein receptor scavenger receptor BI in mice modulates thrombosis susceptibility and indirectly affects platelet function by elevation of plasma free cholesterol.
Arterioscler Thromb Vasc Biol 31: 34 42, 2011.
939. Koscielny J, Klussendorf D, Latza R, Schmitt R, Radtke H, Siegel G, Kiesewetter H.
The antiatherosclerotic effect of Allium sativum. Atherosclerosis 144: 237249, 1999.
940. Koskinas KC, Feldman CL, Chatzizisis YS, Coskun AU, Jonas M, Maynard C, Baker
AB, Papafaklis MI, Edelman ER, Stone PH. Natural history of experimental coronary
atherosclerosis and vascular remodeling in relation to endothelial shear stress: a
serial, in vivo intravascular ultrasound study. Circulation 121: 20922101, 2010.
941. Koupenova M, Johnston-Cox H, Vezeridis A, Gavras H, Yang D, Zannis V, Ravid K.
A2b adenosine receptor regulates hyperlipidemia and atherosclerosis. Circulation
125: 354 363, 2012.
942. Kovacic JC, Moreno P, Hachinski V, Nabel EG, Fuster V. Cellular senescence, vascular disease, and aging: part 1 of a 2-part review. Circulation 123: 1650 1660, 2011.
943. Kovanen PT. Mast cells: multipotent local effector cells in atherothrombosis. Immunol Rev 217: 105122, 2007.
944. Kowluru A, Matti A. Hyperactivation of protein phosphatase 2A in models of glucolipotoxicity and diabetes: potential mechanisms and functional consequences.
Biochem Pharmacol 84: 591597, 2012.
945. Kozaki K, Kaminski WE, Tang J, Hollenbach S, Lindahl P, Sullivan C, Yu JC, Abe K,
Martin PJ, Ross R, Betsholtz C, Giese NA, Raines EW. Blockade of platelet-derived
growth factor or its receptors transiently delays but does not prevent fibrous cap
formation in ApoE null mice. Am J Pathol 161: 13951407, 2002.
946. Kramer A, Jansen A, van Aalst-Cohen E, Tanck M, Kastelein J, Zwinderman A.
Relative risk for cardiovascular atherosclerotic events after smoking cessation: 6 9
years excess risk in individuals with familial hypercholesterolemia. BMC Public Health
6: 262, 2006.
917. Kitamoto S, Sukhova GK, Sun J, Yang M, Libby P, Love V, Duramad P, Sun C, Zhang
Y, Yang X, Peters C, Shi GP. Cathepsin L deficiency reduces diet-induced atherosclerosis in low-density lipoprotein receptor-knockout mice. Circulation 115: 2065
2075, 2007.
934. Kolodgie FD, Burke AP, Nakazawa G, Cheng Q, Xu X, Virmani R. Free cholesterol
in atherosclerotic plaques: where does it come from? Curr Opin Lipidol 18: 500 507,
2007.
965. Kuziel WA, Dawson TC, Quinones M, Garavito E, Chenaux G, Ahuja SS, Reddick RL,
Maeda N. CCR5 deficiency is not protective in the early stages of atherogenesis in
apoE knockout mice. Atherosclerosis 167: 2532, 2003.
966. Kuzuya M, Nakamura K, Sasaki T, Cheng XW, Itohara S, Iguchi A. Effect of MMP-2
deficiency on atherosclerotic lesion formation in apoE-deficient mice. Arterioscler
Thromb Vasc Biol 26: 1120 1125, 2006.
967. Kwak BR, Veillard N, Pelli G, Mulhaupt F, James RW, Chanson M, Mach F. Reduced
connexin43 expression inhibits atherosclerotic lesion formation in low-density lipoprotein receptor-deficient mice. Circulation 107: 10331039, 2003.
950. Kriete A, Bosl WJ, Booker G. Rule-based cell systems model of aging using feedback
loop motifs mediated by stress responses. PLoS Comput Biol 6: e1000820, 2010.
951. Krishnamoorthy S, Recchiuti A, Chiang N, Yacoubian S, Lee CH, Yang R, Petasis NA,
Serhan CN. Resolvin D1 binds human phagocytes with evidence for proresolving
receptors. Proc Natl Acad Sci USA 107: 1660 1665, 2010.
952. Krishnan A, Almen MS, Fredriksson R, Schioth HB. The origin of GPCRs: identification of mammalian like rhodopsin, adhesion, glutamate and frizzled GPCRs in fungi.
PLoS ONE 7: e29817, 2012.
953. Krishnan R, Kremen M, Hu JH, Emery I, Farris SD, Slezicki KI, Chu T, Du L, Dichek
HL, Dichek DA. Level of macrophage uPA expression is an important determinant of
atherosclerotic lesion growth in Apoe/ mice. Arterioscler Thromb Vasc Biol 29:
17371744, 2009.
970. Kyaw T, Tay C, Krishnamurthi S, Kanellakis P, Agrotis A, Tipping P, Bobik A, Toh BH.
B1a B lymphocytes are atheroprotective by secreting natural IgM that increases IgM
deposits and reduces necrotic cores in atherosclerotic lesions. Circ Res 109: 830
840, 2011.
971. Kyaw T, Tipping P, Toh BH, Bobik A. Current understanding of the role of B cell
subsets and intimal and adventitial B cells in atherosclerosis. Curr Opin Lipidol 22:
373379, 2011.
972. Kyaw T, Winship A, Tay C, Kanellakis P, Hosseini H, Cao A, Li P, Tipping P, Bobik A,
Toh BH. Cytotoxic and proinflammatory CD8 T lymphocytes promote development of vulnerable atherosclerotic plaques in apoE/ mice. Circulation 127:
1028 1039, 2013.
973. Kyriakis JM, Avruch J. Mammalian MAPK signal transduction pathways activated by
stress and inflammation: a 10-year update. Physiol Rev 92: 689 737, 2012.
974. Laczy B, Hill BG, Wang K, Paterson AJ, White CR, Xing D, Chen YF, Darley-Usmar
V, Oparil S, Chatham JC. Protein O-GlcNAcylation: a new signaling paradigm for the
cardiovascular system. Am J Physiol Heart Circ Physiol 296: H13H28, 2009.
975. Lagor WR, Rader DJ. Phospholipidation of HDL how much is too much? Clin Chem
57: 4 6, 2011.
976. Lakatta EG. Arterial and cardiac aging: major shareholders in cardiovascular disease
enterprises. Part III: cellular and molecular clues to heart and arterial aging. Circulation 107: 490 497, 2003.
977. Lambert G, Sakai N, Vaisman BL, Neufeld EB, Marteyn B, Chan CC, Paigen B, Lupia
E, Thomas A, Striker LJ, Blanchette-Mackie J, Csako G, Brady JN, Costello R, Striker
GE, Remaley AT, Brewer HB Jr, Santamarina-Fojo S. Analysis of glomerulosclerosis
and atherosclerosis in lecithin cholesterol acyltransferase-deficient mice. J Biol Chem
276: 15090 15098, 2001.
978. Lammers B, Chandak PG, Aflaki E, Van Puijvelde GH, Radovic B, Hildebrand RB,
Meurs I, Out R, Kuiper J, Van Berkel TJ, Kolb D, Haemmerle G, Zechner R, LevakFrank S, Van Eck M, Kratky D. Macrophage adipose triglyceride lipase deficiency
attenuates atherosclerotic lesion development in low-density lipoprotein receptor
knockout mice. Arterioscler Thromb Vasc Biol 31: 6773, 2011.
979. Landar A, Zmijewski JW, Dickinson DA, Le Goffe C, Johnson MS, Milne GL, Zanoni
G, Vidari G, Morrow JD, Darley-Usmar VM. Interaction of electrophilic lipid oxidation products with mitochondria in endothelial cells and formation of reactive oxygen species. Am J Physiol Heart Circ Physiol 290: H1777H1787, 2006.
980. Lande R, Ganguly D, Facchinetti V, Frasca L, Conrad C, Gregorio J, Meller S, Chamilos G, Sebasigari R, Riccieri V, Bassett R, Amuro H, Fukuhara S, Ito T, Liu YJ, Gilliet
M. Neutrophils activate plasmacytoid dendritic cells by releasing self-DNA-peptide
complexes in systemic lupus erythematosus. Science Translational Med 3: 73ra19,
2011.
981. Lande R, Gregorio J, Facchinetti V, Chatterjee B, Wang YH, Homey B, Cao W, Su B,
Nestle FO, Zal T, Mellman I, Schroder JM, Liu YJ, Gilliet M. Plasmacytoid dendritic
cells sense self-DNA coupled with antimicrobial peptide. Nature 449: 564 569,
2007.
982. Laramee M, Chabot C, Cloutier M, Stenne R, Holgado-Madruga M, Wong AJ, Royal
I. The scaffolding adapter Gab1 mediates vascular endothelial growth factor signaling
1511
PAUL N. HOPKINS
and is required for endothelial cell migration and capillary formation. J Biol Chem 282:
7758 7769, 2007.
983. Lauer N, Suvorava T, Ruther U, Jacob R, Meyer W, Harrison DG, Kojda G. Critical
involvement of hydrogen peroxide in exercise-induced up-regulation of endothelial
NO synthase. Cardiovasc Res 65: 254 262, 2005.
1002. Lemaitre V, Soloway PD, DArmiento J. Increased medial degradation with pseudoaneurysm formation in apolipoprotein E-knockout mice deficient in tissue inhibitor
of metalloproteinases-1. Circulation 107: 333338, 2003.
1003. Lemarie CA, Tharaux PL, Esposito B, Tedgui A, Lehoux S. Transforming growth
factor-alpha mediates nuclear factor kappaB activation in strained arteries. Circ Res
99: 434 441, 2006.
984. Laurat E, Poirier B, Tupin E, Caligiuri G, Hansson GK, Bariety J, Nicoletti A. In vivo
downregulation of T helper cell 1 immune responses reduces atherogenesis in apolipoprotein E-knockout mice. Circulation 104: 197202, 2001.
1004. Lemmon MA, Schlessinger J. Cell signaling by receptor tyrosine kinases. Cell 141:
11171134, 2010.
985. Law MR, Wald NJ, Rudnicka AR. Quantifying effect of statins on low density lipoprotein cholesterol, ischaemic heart disease, and stroke: systematic review and metaanalysis. BMJ 326: 1423, 2003.
986. Le NT, Heo KS, Takei Y, Lee H, Woo CH, Chang E, McClain C, Hurley C, Wang X,
Li F, Xu H, Morrell C, Sullivan MA, Cohen MS, Serafimova IM, Taunton J, Fujiwara K,
Abe Ji. A crucial role for p90RSK-mediated reduction of ERK5 transcriptional activity
in endothelial dysfunction and atherosclerosis. Circulation 127: 486 499, 2013.
1006. Leonard SE, Reddie KG, Carroll KS. Mining the thiol proteome for sulfenic acid
modifications reveals new targets for oxidation in cells. ACS Chem Biol 4: 783799,
2009.
1007. Leor J, Poole WK, Kloner RA. Sudden cardiac death triggered by an earthquake. N
Engl J Med 334: 413 419, 1996.
1008. Leppanen P, Koota S, Kholova I, Koponen J, Fieber C, Eriksson U, Alitalo K, YlaHerttuala S. Gene transfers of vascular endothelial growth factor-A, vascular endothelial growth factor-B, vascular endothelial growth factor-C, and vascular endothelial growth factor-D have no effects on atherosclerosis in hypercholesterolemic
low-density lipoprotein-receptor/apolipoprotein B48-deficient mice. Circulation
112: 13471352, 2005.
1009. Leto D, Saltiel AR. Regulation of glucose transport by insulin: traffic control of
GLUT4. Nat Rev Mol Cell Biol 13: 383396, 2012.
990. Lee EY, Klementowicz PT, Hegele RA, Asztalos BF, Schaefer EJ. HDL deficiency due
to a new insertion mutation [ApoA-I(Nashua)] and review of the literature. J Clin
Lipidol 7: 169 173, 2013.
1010. Leung VW, Yun S, Botto M, Mason JC, Malik TH, Song W, Paixao-Cavalcante D,
Pickering MC, Boyle JJ, Haskard DO. Decay-accelerating factor suppresses complement C3 activation and retards atherosclerosis in low-density lipoprotein receptordeficient mice. Am J Pathol 175: 17571767, 2009.
991. Lee HJ, Koh GY. Shear stress activates Tie2 receptor tyrosine kinase in human
endothelial cells. Biochem Biophys Res Commun 304: 399 404, 2003.
992. Lee J, Cooke JP. The role of nicotine in the pathogenesis of atherosclerosis. Atherosclerosis 215: 281283, 2011.
993. Lee MY, Wang Y, Vanhoutte PM. Senescence of cultured porcine coronary arterial
endothelial cells is associated with accelerated oxidative stress and activation of
NFkB. J Vasc Res 47: 287298, 2010.
994. Lee PY, Li Y, Richards HB, Chan FS, Zhuang H, Narain S, Butfiloski EJ, Sobel ES,
Reeves WH, Segal MS. Type I interferon as a novel risk factor for endothelial progenitor cell depletion and endothelial dysfunction in systemic lupus erythematosus.
Arthritis Rheum 56: 3759 3769, 2007.
1011. Lev S, Moreno H, Martinez R, Canoll P, Peles E, Musacchio JM, Plowman GD, Rudy
B, Schlessinger J. Protein tyrosine kinase PYK2 involved in Ca2-induced regulation
of ion channel and MAP kinase functions. Nature 376: 737745, 1995.
1012. Levin MC, Jirholt P, Wramstedt A, Johansson ME, Lundberg AM, Trajkovska MG,
Stahlman M, Fogelstrand P, Brisslert M, Fogelstrand L, Yan ZQ, Hansson GK, Bjorkbacka H, Olofsson SO, Boren J. Rip2 deficiency leads to increased atherosclerosis
despite decreased inflammation. Circ Res 109: 1210 1218, 2011.
1013. Levy AP, Levy JE, Kalet-Litman S, Miller-Lotan R, Levy NS, Asaf R, Guetta J, Yang C,
Purushothaman KR, Fuster V, Moreno PR. Haptoglobin genotype is a determinant of
iron, lipid peroxidation, and macrophage accumulation in the atherosclerotic plaque.
Arterioscler Thromb Vasc Biol 27: 134 140, 2007.
995. Lee TS, Shiao MS, Pan CC, Chau LY. Iron-deficient diet reduces atherosclerotic
lesions in apoE-deficient mice. Circulation 99: 12221229, 1999.
996. Leeuwenburgh C, Hardy MM, Hazen SL, Wagner P, Oh-ishi S, Steinbrecher UP,
Heinecke JW. Reactive nitrogen intermediates promote low density lipoprotein
oxidation in human atherosclerotic intima. J Biol Chem 272: 14331436, 1997.
1015. Lewis MJ, Malik TH, Ehrenstein MR, Boyle JJ, Botto M, Haskard DO. Immunoglobulin M is required for protection against atherosclerosis in low-density lipoprotein
receptor-deficient mice. Circulation 120: 417 426, 2009.
997. Leeuwenburgh C, Rasmussen JE, Hsu FF, Mueller DM, Pennathur S, Heinecke JW.
Mass spectrometric quantification of markers for protein oxidation by tyrosyl radical, copper, and hydroxyl radical in low density lipoprotein isolated from human
atherosclerotic plaques. J Biol Chem 272: 3520 3526, 1997.
998. Lehoux S. Redox signalling in vascular responses to shear and stretch. Cardiovasc Res
71: 269 279, 2006.
1017. Lewis RD, Jackson CL, Morgan BP, Hughes TR. The membrane attack complex of
complement drives the progression of atherosclerosis in apolipoprotein E knockout
mice. Mol Immunol 47: 1098 1105, 2010.
1512
1018. Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation:
the leukocyte adhesion cascade updated. Nat Rev Immunol 7: 678 689, 2007.
1019. Li D, Liu L, Chen H, Sawamura T, Mehta JL. LOX-1, an oxidized LDL endothelial
receptor, induces CD40/CD40L signaling in human coronary artery endothelial cells.
Arterioscler Thromb Vasc Biol 23: 816 821, 2003.
1020. Li D, Mehta JL. Intracellular signaling of LOX-1 in endothelial cell apoptosis. Circ Res
104: 566 568, 2009.
989. Lee DY, Lee CI, Lin TE, Lim SH, Zhou J, Tseng YC, Chien S, Chiu JJ. Role of histone
deacetylases in transcription factor regulation and cell cycle modulation in endothelial cells in response to disturbed flow. Proc Natl Acad Sci USA 109: 19671972, 2012.
1027. Li K, Yao W, Zheng X, Liao K. Berberine promotes the development of atherosclerosis and foam cell formation by inducing scavenger receptor A expression in macrophage. Cell Res 19: 1006 1017, 2009.
1046. Lim SY, Tennant GM, Kennedy S, Wainwright CL, Kane KA. Activation of mouse
protease-activated receptor-2 induces lymphocyte adhesion and generation of reactive oxygen species. Br J Pharmacol 149: 591599, 2006.
1028. Li L, Rose P, Moore PK. Hydrogen sulfide and cell signaling. Annu Rev Pharmacol
Toxicol 51: 169 187, 2011.
1047. Lim WS, Timmins JM, Seimon TA, Sadler A, Kolodgie FD, Virmani R, Tabas I. Signal
transducer and activator of transcription-1 is critical for apoptosis in macrophages
subjected to endoplasmic reticulum stress in vitro and in advanced atherosclerotic
lesions in vivo. Circulation 117: 940 951, 2008.
1031. Li RC, Haribabu B, Mathis SP, Kim J, Gozal D. Leukotriene B4 receptor-1 mediates
intermittent hypoxia-induced atherogenesis. Am J Respir Crit Care Med 184: 124
131, 2011.
1050. Lingrel JB, Pilcher-Roberts R, Basford JE, Manoharan P, Neumann J, Konaniah ES,
Srinivasan R, Bogdanov VY, Hui DY. Myeloid-specific Kruppel-like factor 2 inactivation increases macrophage and neutrophil adhesion and promotes atherosclerosis.
Circ Res 110: 1294 1302, 2012.
1032. Li S, Wang L, Berman M, Kong YY, Dorf ME. Mapping a dynamic innate immunity
protein interaction network regulating type I interferon production. Immunity 35:
426 440, 2011.
1051. Linton MF, Major AS, Fazio S. Proatherogenic role for NK cells revealed. Arterioscler
Thromb Vasc Biol 24: 992994, 2004.
1034. Li SL, Dwarakanath RS, Cai Q, Lanting L, Natarajan R. Effects of silencing leukocytetype 12/15-lipoxygenase using short interfering RNAs. J Lipid Res 46: 220 229, 2005.
1035. Li T, Tian H, Zhao Y, An F, Zhang L, Zhang J, Peng J, Zhang Y, Guo Y. Heme
oxygenase-1 inhibits progression and destabilization of vulnerable plaques in a rabbit
model of atherosclerosis. Eur J Pharmacol 672: 143152, 2011.
1036. Li W, Bengtson MH, Ulbrich A, Matsuda A, Reddy VA, Orth A, Chanda SK, Batalov
S, Joazeiro CA. Genome-wide and functional annotation of human E3 ubiquitin
ligases identifies MULAN, a mitochondrial E3 that regulates the organelles dynamics
and signaling. PLoS ONE 3: e1487, 2008.
1037. Li W, Tang C, Jin H, Du J. Regulatory effects of sulfur dioxide on the development of
atherosclerotic lesions and vascular hydrogen sulfide in atherosclerotic rats. Atherosclerosis 215: 323330, 2011.
1038. Li XM, Tang WH, Mosior MK, Huang Y, Wu Y, Matter W, Gao V, Schmitt D,
Didonato JA, Fisher EA, Smith JD, Hazen SL. Paradoxical association of enhanced
cholesterol efflux with increased incident cardiovascular risks. Arterioscler Thromb
Vasc Biol 2013.
1039. Li Y, Ge M, Ciani L, Kuriakose G, Westover EJ, Dura M, Covey DF, Freed JH,
Maxfield FR, Lytton J, Tabas I. Enrichment of endoplasmic reticulum with cholesterol
inhibits sarcoplasmic-endoplasmic reticulum calcium ATPase-2b activity in parallel
with increased order of membrane lipids: implications for depletion of endoplasmic
1053. Litman GW, Rast JP, Fugmann SD. The origins of vertebrate adaptive immunity. Nat
Rev Immunol 10: 543553, 2010.
1054. Little PJ, Chait A, Bobik A. Cellular and cytokine-based inflammatory processes as
novel therapeutic targets for the prevention and treatment of atherosclerosis. Pharmacol Ther 131: 255268, 2011.
1055. Little PJ, Tannock L, Olin KL, Chait A, Wight TN. Proteoglycans synthesized by
arterial smooth muscle cells in the presence of transforming growth factor-beta1
exhibit increased binding to LDLs. Arterioscler Thromb Vasc Biol 22: 55 60, 2002.
1056. Liu J, Afroza H, Rader DJ, Jin W. Angiopoietin-like protein 3 inhibits lipoprotein lipase
activity through enhancing its cleavage by proprotein convertases. J Biol Chem 285:
2756127570, 2010.
1057. Liu J, Agarwal S. Mechanical signals activate vascular endothelial growth factor receptor-2 to upregulate endothelial cell proliferation during inflammation. J Immunol
185: 12151221, 2010.
1058. Liu J, Huan C, Chakraborty M, Zhang H, Lu D, Kuo MS, Cao G, Jiang XC. Macrophage sphingomyelin synthase 2 deficiency decreases atherosclerosis in mice. Circ
Res 105: 295303, 2009.
1059. Liu J, Liao Z, Camden J, Griffin KD, Garrad RC, Santiago-Perez LI, Gonzalez FA, Seye
CI, Weisman GA, Erb L. Src homology 3 binding sites in the P2Y2 nucleotide recep-
1513
1026. Li JM, Fan LM, Christie MR, Shah AM. Acute tumor necrosis factor alpha signaling via
NADPH oxidase in microvascular endothelial cells: role of p47phox phosphorylation
and binding to TRAF4. Mol Cell Biol 25: 2320 2330, 2005.
PAUL N. HOPKINS
tor interact with Src and regulate activities of Src, proline-rich tyrosine kinase 2, and
growth factor receptors. J Biol Chem 279: 8212 8218, 2004.
1060. Liu J, Thewke DP, Su YR, Linton MF, Fazio S, Sinensky MS. Reduced macrophage
apoptosis is associated with accelerated atherosclerosis in low-density lipoprotein
receptor-null mice. Arterioscler Thromb Vasc Biol 25: 174 179, 2005.
1061. Liu P, Yu YRA, Spencer JA, Johnson AE, Vallanat CT, Fong AM, Patterson C, Patel
DD. CX3CR1 deficiency impairs dendritic cell accumulation in arterial intima and
reduces atherosclerotic burden. Arterioscler Thromb Vasc Biol 28: 243250, 2008.
1062. Liu X, Ukai T, Yumoto H, Davey M, Goswami S, Gibson FC 3rd, Genco CA. Toll-like
receptor 2 plays a critical role in the progression of atherosclerosis that is independent of dietary lipids. Atherosclerosis 196: 146 154, 2008.
1063. Liu Y, Sanoff HK, Cho H, Burd CE, Torrice C, Mohlke KL, Ibrahim JG, Thomas NE,
Sharpless NE. INK4/ARF transcript expression is associated with chromosome 9p21
variants linked to atherosclerosis. PLoS ONE 4: e5027, 2009.
1064. Liu Y, Sweet DT, Irani-Tehrani M, Maeda N, Tzima E. Shc coordinates signals from
intercellular junctions and integrins to regulate flow-induced inflammation. J Cell Biol
182: 185196, 2008.
1065. Liu Y, Tang C. Regulation of ABCA1 functions by signaling pathways. Biochim Biophys
Acta 1821: 522529, 2012.
1066. Lo Sasso G, Murzilli S, Salvatore L, DErrico I, Petruzzelli M, Conca P, Jiang ZY,
Calabresi L, Parini P, Moschetta A. Intestinal specific LXR activation stimulates reverse cholesterol transport and protects from atherosclerosis. Cell Metab 12: 187
193, 2010.
1068. London NR, Zhu W, Bozza FA, Smith MC, Greif DM, Sorensen LK, Chen L, Kaminoh
Y, Chan AC, Passi SF, Day CW, Barnard DL, Zimmerman GA, Krasnow MA, Li DY.
Targeting Robo4-dependent Slit signaling to survive the cytokine storm in sepsis and
influenza. Science Translational Med 2: 23ra19, 2010.
1069. Long EK, Picklo MJ Sr. Trans-4-hydroxy-2-hexenal, a product of n-3 fatty acid peroxidation: make some room HNE. Free Radic Biol Med 49: 1 8, 2010.
1070. Loot AE, Schreiber JG, Fisslthaler B, Fleming I. Angiotensin II impairs endothelial
function via tyrosine phosphorylation of the endothelial nitric oxide synthase. J Exp
Med 206: 2889 2896, 2009.
1071. Lopes-Virella MF, Baker NL, Hunt KJ, Lachin J, Nathan D, Virella G. Oxidized LDL
immune complexes and coronary artery calcification in type 1 diabetes. Atherosclerosis 214: 462 467, 2011.
1072. Lopes-Virella MF, Baker NL, Hunt KJ, Lyons TJ, Jenkins AJ, Virella G. High concentrations of AGE-LDL and oxidized LDL in circulating immune complexes are associated with progression of retinopathy in type 1 diabetes. Diabetes Care 35: 1333
1340, 2012.
1073. Lopes-Virella MF, Carter RE, Baker NL, Lachin J, Virella G. High levels of oxidized
LDL in circulating immune complexes are associated with increased odds of developing abnormal albuminuria in Type 1 diabetes. Nephrol Dial Transplant 27: 1416
1423, 2012.
1074. Lopes-Virella MF, Hunt KJ, Baker NL, Moritz T, Virella G. Levels of MDA-LDL
incirculating immune complexes predict myocardial infarction in theVADT study in
the Veterans Affairs Diabetes Trial (VADT). J Clin Lipidol 6: 256, 2012.
1075. Lorant DE, Patel KD, McIntyre TM, McEver RP, Prescott SM, Zimmerman GA.
Coexpression of GMP-140 and PAF by endothelium stimulated by histamine or
thrombin: a juxtacrine system for adhesion and activation of neutrophils. J Cell Biol
115: 223234, 1991.
1079. Loukogeorgakis SP, van den Berg MJ, Sofat R, Nitsch D, Charakida M, Haiyee B, de
Groot E, MacAllister RJ, Kuijpers TW, Deanfield JE. Role of NADPH oxidase in
endothelial ischemia/reperfusion injury in humans. Circulation 121: 2310 2316,
2010.
1080. Lu H, Rateri DL, Feldman RJ DL Jr, Fukamizu A, Ishida J, Oesterling EG, Cassis LA,
Daugherty A. Renin inhibition reduces hypercholesterolemia-induced atherosclerosis in mice. J Clin Invest 118: 984 993, 2008.
1081. Lu J, Mitra S, Wang X, Khaidakov M, Mehta JL. Oxidative stress and lectin-like
ox-LDL-receptor LOX-1 in atherogenesis and tumorigenesis. Antioxid Redox Signal
2011.
1082. Lu J, Yang JH, Burns AR, Chen HH, Tang D, Walterscheid JP, Suzuki S, Yang CY,
Sawamura T, Chen CH. Mediation of electronegative low-density lipoprotein signaling by LOX-1: a possible mechanism of endothelial apoptosis. Circ Res 104: 619
627, 2009.
1083. Lu LF, Rudensky A. Molecular orchestration of differentiation and function of regulatory T cells. Genes Dev 23: 1270 1282, 2009.
1084. Luchtefeld M, Grothusen C, Gagalick A, Jagavelu K, Schuett H, Tietge UJ, Pabst O,
Grote K, Drexler H, Forster R, Schieffer B. Chemokine receptor 7 knockout attenuates atherosclerotic plaque development. Circulation 122: 16211628, 2010.
1085. Luchtefeld M, Schunkert H, Stoll M, Selle T, Lorier R, Grote K, Sagebiel C,
Jagavelu K, Tietge UJ, Assmus U, Streetz K, Hengstenberg C, Fischer M, Mayer
B, Maresso K, El Mokhtari NE, Schreiber S, Muller W, Bavendiek U, Grothusen
C, Drexler H, Trautwein C, Broeckel U, Schieffer B. Signal transducer of inflammation gp130 modulates atherosclerosis in mice and man. J Exp Med 204: 1935
1944, 2007.
1086. Ludwig RJ, Herzog C, Rostock A, Ochsendorf FR, Zollner TM, Thaci D, Kaufmann R,
Vogl TJ, Boehncke WH. Psoriasis: a possible risk factor for development of coronary
artery calcification. Br J Dermatol 156: 271276, 2007.
1087. Lukasova M, Malaval C, Gille A, Kero J, Offermanns S. Nicotinic acid inhibits progression of atherosclerosis in mice through its receptor GPR109A expressed by
immune cells. J Clin Invest 121: 11631173, 2011.
1088. Lundberg AM, Hansson GK. Innate immune signals in atherosclerosis. Clin Immunol
134: 524, 2010.
1089. Luo W, Wang H, Ohman MK, Guo C, Shi K, Wang J, Eitzman DT. P-selectin
glycoprotein ligand-1 deficiency leads to cytokine resistance and protection
against atherosclerosis in apolipoprotein E deficient mice. Atherosclerosis 220:
110 117, 2012.
1090. Luo Y, Xu Z, Wan T, He Y, Jones D, Zhang H, Min W. Endothelial-specific transgenesis of TNFR2 promotes adaptive arteriogenesis and angiogenesis. Arterioscler
Thromb Vasc Biol 30: 13071314, 2010.
1091. Lutgens E, Gijbels M, Smook M, Heeringa P, Gotwals P, Koteliansky VE, Daemen
MJ. Transforming growth factor-beta mediates balance between inflammation
and fibrosis during plaque progression. Arterioscler Thromb Vasc Biol 22: 975
982, 2002.
1092. Lutgens E, Gorelik L, Daemen MJ, de Muinck ED, Grewal IS, Koteliansky VE, Flavell
RA. Requirement for CD154 in the progression of atherosclerosis. Nat Med 5:
13131316, 1999.
1076. Lorenowicz MJ, Fernandez-Borja M, Hordijk PL. cAMP signaling in leukocyte transendothelial migration. Arterioscler Thromb Vasc Biol 27: 1014 1022, 2007.
1094. Lutgens E, Lutgens SP, Faber BC, Heeneman S, Gijbels MM, de Winther MP, Frederik P, van der Made I, Daugherty A, Sijbers AM, Fisher A, Long CJ, Saftig P, Black D,
Daemen MJ, Cleutjens KB. Disruption of the cathepsin K gene reduces atherosclerosis progression and induces plaque fibrosis but accelerates macrophage foam cell
formation. Circulation 113: 98 107, 2006.
1514
1067. Locasale JW, Shaw AS, Chakraborty AK. Scaffold proteins confer diverse regulatory
properties to protein kinase cascades. Proc Natl Acad Sci USA 104: 1330713312,
2007.
1078. Lou XJ, Boonmark NW, Horrigan FT, Degen JL, Lawn RM. Fibrinogen deficiency
reduces vascular accumulation of apolipoprotein(a) and development of atherosclerosis in apolipoprotein(a) transgenic mice. Proc Natl Acad Sci USA 95: 1259112595,
1998.
1102. Ma Z, Liu Z, Wu RF, Terada LS. p66(Shc) restrains Ras hyperactivation and suppresses metastatic behavior. Oncogene 29: 5559 5567, 2010.
1103. Mabb AM, Wuerzberger-Davis SM, Miyamoto S. PIASy mediates NEMO sumoylation and NF-kappaB activation in response to genotoxic stress. Nat Cell Biol 8:
986 993, 2006.
1114. Mahabadi AA, Reinsch N, Lehmann N, Altenbernd J, Kalsch H, Seibel RM, Erbel R,
Mohlenkamp S. Association of pericoronary fat volume with atherosclerotic plaque
burden in the underlying coronary artery: a segment analysis. Atherosclerosis 211:
195199, 2010.
1115. Mahul-Mellier AL, Pazarentzos E, Datler C, Iwasawa R, AbuAli G, Lin B, Grimm S.
De-ubiquitinating protease USP2a targets RIP1 and TRAF2 to mediate cell death by
TNF. Cell Death Differ 19: 891 899, 2012.
1116. Majdalawieh A, Zhang L, Fuki IV, Rader DJ, Ro HS. Adipocyte enhancer-binding
protein 1 is a potential novel atherogenic factor involved in macrophage cholesterol
homeostasis and inflammation. Proc Natl Acad Sci USA 103: 2346 2351, 2006.
1117. Major AS, Fazio S, Linton MF. B-lymphocyte deficiency increases atherosclerosis in
LDL receptor-null mice. Arterioscler Thromb Vasc Biol 22: 18921898, 2002.
1118. Major AS, Harrison DG. What fans the fire: insights into mechanisms of inflammation
in atherosclerosis and diabetes mellitus. Circulation 124: 2809 2811, 2011.
1119. Major AS, Wilson MT, McCaleb JL, Ru Su Y, Stanic AK, Joyce S, Van Kaer L, Fazio S,
Linton MF. Quantitative and qualitative differences in proatherogenic NKT cells in
apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 24: 23512357, 2004.
1120. Major CD, Santulli RJ, Derian CK, Andrade-Gordon P. Extracellular mediators in
atherosclerosis and thrombosis: lessons from thrombin receptor knockout mice.
Arterioscler Thromb Vasc Biol 23: 931939, 2003.
1121. Makinen PI, Lappalainen JP, Heinonen SE, Leppanen P, Lahteenvuo MT, Aarnio JV,
Heikkila J, Turunen MP, Yla-Herttuala S. Silencing of either SR-A or CD36 reduces
atherosclerosis in hyperlipidaemic mice and reveals reciprocal upregulation of these
receptors. Cardiovasc Res 88: 530 538, 2010.
1104. Mabley JG, Soriano FG. Role of nitrosative stress and poly(ADP-ribose) polymerase
activation in diabetic vascular dysfunction. Curr Vasc Pharmacol 3: 247252, 2005.
1122. Makowski L, Boord JB, Maeda K, Babaev VR, Uysal KT, Morgan MA, Parker RA,
Suttles J, Fazio S, Hotamisligil GS, Linton MF. Lack of macrophage fatty-acid-binding
protein aP2 protects mice deficient in apolipoprotein E against atherosclerosis. Nat
Med 7: 699 705, 2001.
1105. MacDonald ML, van Eck M, Hildebrand RB, Wong BW, Bissada N, Ruddle P, Kontush
A, Hussein H, Pouladi MA, Chapman MJ, Fievet C, van Berkel TJ, Staels B, McManus
BM, Hayden MR. Despite antiatherogenic metabolic characteristics, SCD1-deficient
mice have increased inflammation and atherosclerosis. Arterioscler Thromb Vasc Biol
29: 341347, 2009.
1123. Makowski L, Brittingham KC, Reynolds JM, Suttles J, Hotamisligil GS. The fatty
acid-binding protein, aP2, coordinates macrophage cholesterol trafficking and inflammatory activity. Macrophage expression of aP2 impacts peroxisome proliferator-activated receptor gamma and IkappaB kinase activities. J Biol Chem 280: 12888
12895, 2005.
1106. Mack PJ, Zhang Y, Chung S, Vickerman V, Kamm RD, Garcia-Cardena G. Biomechanical regulation of endothelium-dependent events critical for adaptive remodeling. J Biol Chem 284: 8412 8420, 2009.
1124. Malik TH, Cortini A, Carassiti D, Boyle JJ, Haskard DO, Botto M. The alternative
pathway is critical for pathogenic complement activation in endotoxin- and dietinduced atherosclerosis in low-density lipoprotein receptor-deficient mice. Circulation 122: 1948 1956, 2010.
1107. Macneil LT, Walhout AJ. Gene regulatory networks and the role of robustness and
stochasticity in the control of gene expression. Genome Res 2011.
1108. MacRitchie N, Grassia G, Sabir SR, Maddaluno M, Welsh P, Sattar N, Ialenti A,
Kurowska-Stolarska M, McInnes IB, Brewer JM, Garside P, Maffia P. Plasmacytoid
dendritic cells play a key role in promoting atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2012.
1109. Madrigal-Matute J, Lopez-Franco O, Blanco-Colio LM, Munoz-Garcia B, RamosMozo P, Ortega L, Egido J, Martin-Ventura JL. Heat shock protein 90 inhibitors
attenuate inflammatory responses in atherosclerosis. Cardiovasc Res 86: 330 337,
2010.
1110. Madsen HO, Videm V, Svejgaard A, Svennevig JL, Garred P. Association of mannosebinding-lectin deficiency with severe atherosclerosis. Lancet 352: 959 960, 1998.
1111. Maeda N. Animal Models of Diabetic Complications Consortium (U01 HL087946).
In: Annual Report 2008. Dislipidemia, Lipoic Acid and Diabetic Vascular Complications in
Humanized Mice. Chapel Hill, NC: Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 2008.
1112. Maganto-Garcia E, Tarrio ML, Grabie N, Bu DX, Lichtman AH. Dynamic changes in
regulatory T cells are linked to levels of diet-induced hypercholesterolemia. Circulation 124: 185195, 2011.
1125. Malik UZ, Hundley NJ, Romero G, Radi R, Freeman BA, Tarpey MM, Kelley EE.
Febuxostat inhibition of endothelial-bound XO: Implications for targeting vascular
ROS production. Free Radic Biol Med 51: 179 184, 2011.
1126. Mallat Z, Gojova A, Brun V, Esposito B, Fournier N, Cottrez F, Tedgui A, Groux H.
Induction of a regulatory T cell type 1 response reduces the development of atherosclerosis in apolipoprotein E-knockout mice. Circulation 108: 12321237, 2003.
1127. Mallat Z, Gojova A, Marchiol-Fournigault C, Esposito B, Kamate C, Merval R, Fradelizi D, Tedgui A. Inhibition of transforming growth factor-beta signaling accelerates
atherosclerosis and induces an unstable plaque phenotype in mice. Circ Res 89:
930 934, 2001.
1128. Mallat Z, Gojova A, Sauzeau V, Brun V, Silvestre JS, Esposito B, Merval R, Groux H,
Loirand G, Tedgui A. Rho-associated protein kinase contributes to early atherosclerotic lesion formation in mice. Circ Res 93: 884 888, 2003.
1129. Mallat Z, Lambeau G, Tedgui A. Lipoprotein-associated and secreted phospholipases A(2) in cardiovascular disease: roles as biological effectors and biomarkers.
Circulation 122: 21832200, 2010.
1130. Mallat Z, Taleb S, Ait-Oufella H, Tedgui A. The role of adaptive T cell immunity in
atherosclerosis. J Lipid Res 50 Suppl: S364 369, 2009.
1515
1101. Ma X, Qiu S, Luo J, Ma Y, Ngai CY, Shen B, Wong CO, Huang Y, Yao X. Functional
role of vanilloid transient receptor potential 4-canonical transient receptor potential
1 complex in flow-induced Ca2 influx. Arterioscler Thromb Vasc Biol 30: 851 858,
2010.
1113. Magoori K, Kang MJ, Ito MR, Kakuuchi H, Ioka RX, Kamataki A, Kim DH, Asaba H,
Iwasaki S, Takei YA, Sasaki M, Usui S, Okazaki M, Takahashi S, Ono M, Nose M, Sakai
J, Fujino T, Yamamoto TT. Severe hypercholesterolemia, impaired fat tolerance, and
advanced atherosclerosis in mice lacking both low density lipoprotein receptorrelated protein 5 and apolipoprotein E. J Biol Chem 278: 1133111336, 2003.
PAUL N. HOPKINS
1131. Maloney E, Sweet IR, Hockenbery DM, Pham M, Rizzo NO, Tateya S, Handa P,
Schwartz MW, Kim F. Activation of NF-B by palmitate in endothelial cells: a key
role for NADPH oxidase-derived superoxide in response to TLR4 activation. Arterioscler Thromb Vasc Biol 29: 1370 1375, 2009.
1132. Mamontova A, Seguret-Mace S, Esposito B, Chaniale C, Bouly M, DelhayeBouchaud N, Luc G, Staels B, Duverger N, Mariani J, Tedgui A. Severe atherosclerosis and hypoalphalipoproteinemia in the staggerer mouse, a mutant of the nuclear
receptor RORalpha. Circulation 98: 2738 2743, 1998.
1133. Mangat R, Warnakula S, Borthwick F, Hassanali Z, Uwiera RRE, Russell JC, Cheeseman CI, Vine DF, Proctor SD. Arterial retention of remnant lipoproteins ex vivo is
increased in insulin resistance because of increased arterial biglycan and production
of cholesterol-rich atherogenic particles that can be improved by ezetimibe in the
JCR:LA-cp rat. J Am Heart Assoc 1: 2012.
1134. Mani A, Radhakrishnan J, Wang H, Mani A, Mani MA, Nelson-Williams C, Carew KS,
Mane S, Najmabadi H, Wu D, Lifton RP. LRP6 mutation in a family with early
coronary disease and metabolic risk factors. Science 315: 1278 1282, 2007.
1135. Manka D, Chatterjee TK, Moseley N, LaSance K, Lemen L, Hui DY, Weintraub
NL. Abstract 15733: perivascular adipose tissue triggers vasa vasorum formation
and injury-induced neointimal hyperplasia in mice. Circulation 122: A15733,
2010.
1136. Mann J, Davies MJ. Mechanisms of progression in native coronary artery disease: role
of healed plaque disruption. Heart 82: 265268, 1999.
1138. Marciniak SJ, Ron D. Endoplasmic reticulum stress signaling in disease. Physiol Rev 86:
11331149, 2006.
1157. Mattagajasingh I, Kim CS, Naqvi A, Yamamori T, Hoffman TA, Jung SB, DeRicco J,
Kasuno K, Irani K. SIRT1 promotes endothelium-dependent vascular relaxation by
activating endothelial nitric oxide synthase. Proc Natl Acad Sci USA 104: 14855
14860, 2007.
1140. Markevich NI, Hoek JB, Kholodenko BN. Signaling switches and bistability arising
from multisite phosphorylation in protein kinase cascades. J Cell Biol 164: 353359,
2004.
1158. Mattaliano MD, Wooters J, Shih HH, Paulsen JE. ROCK2 associates with lectin-like
oxidized LDL receptor-1 and mediates oxidized LDL-induced IL-8 production. Am J
Physiol Cell Physiol 298: C1180 C1187, 2010.
1159. Matthijsen RA, de Winther MP, Kuipers D, van der Made I, Weber C, Herias MV,
Gijbels MJ, Buurman WA. Macrophage-specific expression of mannose-binding lectin controls atherosclerosis in low-density lipoprotein receptor-deficient mice. Circulation 119: 2188 2195, 2009.
1142. Maron R, Sukhova G, Faria AM, Hoffmann E, Mach F, Libby P, Weiner HL. Mucosal
administration of heat shock protein-65 decreases atherosclerosis and inflammation
in aortic arch of low-density lipoprotein receptor-deficient mice. Circulation 106:
1708 1715, 2002.
1143. Marotti KR, Castle CK, Boyle TP, Lin AH, Murray RW, Melchior GW. Severe atherosclerosis in transgenic mice expressing simian cholesteryl ester transfer protein.
Nature 364: 7375, 1993.
1144. Marro S, Chiabrando D, Messana E, Stolte J, Turco E, Tolosano E, Muckenthaler
MU. Heme controls ferroportin1 (FPN1) transcription involving Bach1, Nrf2 and a
MARE/ARE sequence motif at position -7007 of the FPN1 promoter. Haematologica
95: 12611268, 2010.
1160. Mattison JA, Roth GS, Beasley TM, Tilmont EM, Handy AM, Herbert RL, Longo DL,
Allison DB, Young JE, Bryant M, Barnard D, Ward WF, Qi W, Ingram DK, de Cabo
R. Impact of caloric restriction on health and survival in rhesus monkeys from the
NIA study. Nature 2012.
1161. Maurovich-Horvat P, Kallianos K, Engel LC, Szymonifka J, Fox CS, Hoffmann U,
Truong QA. Influence of pericoronary adipose tissue on local coronary atherosclerosis as assessed by a novel MDCT volumetric method. Atherosclerosis 219: 151
157, 2011.
1162. Mayr M, Sidibe A, Zampetaki A. The paradox of hypoxic and oxidative stress in
atherosclerosis. J Am Coll Cardiol 51: 1266 1267, 2008.
1145. Marsh SA, Coombes JS. Exercise and the endothelial cell. Int J Cardiol 99: 165169,
2005.
1163. Mays LE, Chen YH. Maintaining immunological tolerance with Foxp3. Cell Res 17:
904 918, 2007.
1146. Marson A, Kretschmer K, Frampton GM, Jacobsen ES, Polansky JK, MacIsaac KD,
Levine SS, Fraenkel E, von Boehmer H, Young RA. Foxp3 occupancy and regulation
of key target genes during T-cell stimulation. Nature 445: 931935, 2007.
1164. McAllister-Lucas LM, Jin X, Gu S, Siu K, McDonnell S, Ruland J, Delekta PC, Van Beek
M, Lucas PC. The CARMA3-Bcl10-MALT1 signalosome promotes angiotensin IIdependent vascular inflammation and atherogenesis. J Biol Chem 285: 25880 25884,
2010.
1516
1165. McAlpine CS, Bowes AJ, Khan MI, Shi Y, Werstuck GH. Endoplasmic reticulum
stress and glycogen synthase kinase-3beta activation in apolipoprotein E-deficient
mouse models of accelerated atherosclerosis. Arterioscler Thromb Vasc Biol 32: 82
91, 2012.
1166. McClelland S, Cox C, OConnor R, de Gaetano M, McCarthy C, Cryan L, Fitzgerald
D, Belton O. Conjugated linoleic acid suppresses the migratory and inflammatory
phenotype of the monocyte/macrophage cell. Atherosclerosis 211: 96 102, 2010.
1137. Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase
complement of the human genome. Science 298: 19121934, 2002.
1155. Matsui R, Xu S, Maitland KA, Mastroianni R, Leopold JA, Handy DE, Loscalzo J,
Cohen RA. Glucose-6-phosphate dehydrogenase deficiency decreases vascular superoxide and atherosclerotic lesions in apolipoprotein E(/) mice. Arterioscler
Thromb Vasc Biol 26: 910 916, 2006.
1168. McDonald JE, Padmanabhan N, Petrie MC, Hillier C, Connell JM, McMurray JJ.
Vasoconstrictor effect of the angiotensin-converting enzyme-resistant, chymasespecific substrate [Pro(11)(D)-Ala(12)] angiotensin I in human dorsal hand veins: in
vivo demonstration of non-ace production of angiotensin II in humans. Circulation
104: 18051808, 2001.
1187. Mehta PK, Griendling KK. Angiotensin II cell signaling: physiological and pathological
effects in the cardiovascular system. Am J Physiol Cell Physiol 292: C82C97, 2007.
1169. McDonald TO, Gerrity RG, Jen C, Chen HJ, Wark K, Wight TN, Chait A, OBrien
KD. Diabetes and arterial extracellular matrix changes in a porcine model of atherosclerosis. J Histochem Cytochem 55: 1149 1157, 2007.
1170. McEwen JE, Zimniak P, Mehta JL, Shmookler Reis RJ. Molecular pathology of aging
and its implications for senescent coronary atherosclerosis. Curr Opin Cardiol 20:
399 406, 2005.
1172. McKenzie JA, Ridley AJ. Roles of Rho/ROCK and MLCK in TNF-alpha-induced
changes in endothelial morphology and permeability. J Cell Physiol 213: 221228,
2007.
1192. Mendoza SA, Fang J, Gutterman DD, Wilcox DA, Bubolz AH, Li R, Suzuki M, Zhang
DX. TRPV4-mediated endothelial Ca2 influx and vasodilation in response to shear
stress. Am J Physiol Heart Circ Physiol 298: H466 H476, 2010.
1173. McLaren JE, Calder CJ, McSharry BP, Sexton K, Salter RC, Singh NN, Wilkinson GW,
Wang EC, Ramji DP. The TNF-like protein 1A-death receptor 3 pathway promotes
macrophage foam cell formation in vitro. J Immunol 184: 58275834, 2010.
1174. McLaren JE, Michael DR, Ashlin TG, Ramji DP. Cytokines, macrophage lipid metabolism and foam cells: implications for cardiovascular disease therapy. Prog Lipid Res
50: 331347, 2011.
1194. Mercer J, Figg N, Stoneman V, Braganza D, Bennett MR. Endogenous p53 protects
vascular smooth muscle cells from apoptosis and reduces atherosclerosis in ApoE
knockout mice. Circ Res 96: 667 674, 2005.
1175. McLaren JE, Michael DR, Salter RC, Ashlin TG, Calder CJ, Miller AM, Liew FY, Ramji
DP. IL-33 reduces macrophage foam cell formation. J Immunol 185: 12221229,
2010.
1195. Mercer JR, Cheng KK, Figg N, Gorenne I, Mahmoudi M, Griffin J, Vidal-Puig A, Logan
A, Murphy MP, Bennett M. DNA damage links mitochondrial dysfunction to atherosclerosis and the metabolic syndrome. Circ Res 107: 10211031, 2010.
1176. McMillen TS, Heinecke JW, LeBoeuf RC. Expression of human myeloperoxidase by
macrophages promotes atherosclerosis in mice. Circulation 111: 2798 2804, 2005.
1196. Merched AJ, Chan L. Absence of p21Waf1/Cip1/Sdi1 modulates macrophage differentiation and inflammatory response and protects against atherosclerosis. Circulation 110: 3830 3841, 2004.
1177. McNally JS, Davis ME, Giddens DP, Saha A, Hwang J, Dikalov S, Jo H, Harrison DG.
Role of xanthine oxidoreductase and NAD(P)H oxidase in endothelial superoxide
production in response to oscillatory shear stress. Am J Physiol Heart Circ Physiol 285:
H2290 H2297, 2003.
1197. Merched AJ, Ko K, Gotlinger KH, Serhan CN, Chan L. Atherosclerosis: evidence for
impairment of resolution of vascular inflammation governed by specific lipid mediators. FASEB J 22: 35953606, 2008.
1178. McNally JS, Saxena A, Cai H, Dikalov S, Harrison DG. Regulation of xanthine oxidoreductase protein expression by hydrogen peroxide and calcium. Arterioscler
Thromb Vasc Biol 25: 16231628, 2005.
1179. Megens RT, Vijayan S, Lievens D, Doring Y, van Zandvoort MA, Grommes J, Weber
C, Soehnlein O. Presence of luminal neutrophil extracellular traps in atherosclerosis.
Thromb Haemost 107: 597598, 2012.
1180. Mehrabian M, Allayee H, Stockton J, Lum PY, Drake TA, Castellani LW, Suh M,
Armour C, Edwards S, Lamb J, Lusis AJ, Schadt EE. Integrating genotypic and expression data in a segregating mouse population to identify 5-lipoxygenase as a
susceptibility gene for obesity and bone traits. Nat Genet 37: 1224 1233, 2005.
1181. Mehrabian M, Allayee H, Wong J, Shi W, Wang XP, Shaposhnik Z, Funk CD, Lusis AJ,
Shih W. Identification of 5-lipoxygenase as a major gene contributing to atherosclerosis susceptibility in mice. Circ Res 91: 120 126, 2002.
1182. Mehta JL, Chen J, Hermonat PL, Romeo F, Novelli G. Lectin-like, oxidized lowdensity lipoprotein receptor-1 (LOX-1): A critical player in the development of
atherosclerosis and related disorders. Cardiovasc Res 69: 36 45, 2006.
1183. Mehta JL, Hu B, Chen J, Li D. Pioglitazone inhibits LOX-1 expression in human
coronary artery endothelial cells by reducing intracellular superoxide radical generation. Arterioscler Thromb Vasc Biol 23: 22032208, 2003.
1184. Mehta JL, Li D. Identification, regulation and function of a novel lectin-like oxidized
low-density lipoprotein receptor. J Am Coll Cardiol 39: 1429 1435, 2002.
1185. Mehta JL, Sanada N, Hu CP, Chen J, Dandapat A, Sugawara F, Satoh H, Inoue K,
Kawase Y, Jishage Ki Suzuki H, Takeya M, Schnackenberg L, Beger R, Hermonat PL,
Thomas M, Sawamura T. Deletion of LOX-1 reduces atherogenesis in LDLR knockout mice fed high cholesterol diet. Circ Res 100: 1634 1642, 2007.
1205. Miao H, Hu YL, Shiu YT, Yuan S, Zhao Y, Kaunas R, Wang Y, Jin G, Usami S, Chien
S. Effects of flow patterns on the localization and expression of VE-cadherin at
vascular endothelial cell junctions: in vivo and in vitro investigations. J Vasc Res 42:
77 89, 2005.
1517
1171. McKellar GE, McCarey DW, Sattar N, McInnes IB. Role for TNF in atherosclerosis?
Lessons from autoimmune disease. Nat Rev Cardiol 6: 410 417, 2009.
1191. Mendez-Fernandez YV, Stevenson BG, Diehl CJ, Braun NA, Wade NS, Covarrubias
R, van Leuven S, Witztum JL, Major AS. The inhibitory FcgammaRIIb modulates the
inflammatory response and influences atherosclerosis in male apoE(/) mice.
Atherosclerosis 214: 73 80, 2011.
PAUL N. HOPKINS
1206. Michel JB, Thaunat O, Houard X, Meilhac O, Caligiuri G, Nicoletti A. Topological
determinants and consequences of adventitial responses to arterial wall injury. Arterioscler Thromb Vasc Biol 27: 1259 1268, 2007.
1207. Michelsen KS, Wong MH, Shah PK, Zhang W, Yano J, Doherty TM, Akira S, Rajavashisth TB, Arditi M. Lack of Toll-like receptor 4 or myeloid differentiation factor 88
reduces atherosclerosis and alters plaque phenotype in mice deficient in apolipoprotein E. Proc Natl Acad Sci USA 101: 10679 10684, 2004.
1208. Mill C, George SJ. Wnt signalling in smooth muscle cells and its role in cardiovascular
disorders. Cardiovasc Res 95: 233240, 2012.
1209. Miller AM, Xu D, Asquith DL, Denby L, Li Y, Sattar N, Baker AH, McInnes IB, Liew
FY. IL-33 reduces the development of atherosclerosis. J Exp Med 205: 339 346,
2008.
1210. Miller YI, Choi SH, Wiesner P, Fang L, Harkewicz R, Hartvigsen K, Boullier A, Gonen
A, Diehl CJ, Que X, Montano E, Shaw PX, Tsimikas S, Binder CJ, Witztum JL.
Oxidation-specific epitopes are danger-associated molecular patterns recognized by
pattern recognition receptors of innate immunity. Circ Res 108: 235248, 2011.
1211. Miller YI, Viriyakosol S, Binder CJ, Feramisco JR, Kirkland TN, Witztum JL. Minimally
modified LDL binds to CD14, induces macrophage spreading via TLR4/MD-2, and
inhibits phagocytosis of apoptotic cells. J Biol Chem 278: 15611568, 2003.
1212. Millonig G, Niederegger H, Rabl W, Hochleitner BW, Hoefer D, Romani N, Wick G.
Network of vascular-associated dendritic cells in intima of healthy young individuals.
Arterioscler Thromb Vasc Biol 21: 503508, 2001.
1224. Mocsai A, Ruland J, Tybulewicz VL. The SYK tyrosine kinase: a crucial player in
diverse biological functions. Nat Rev Immunol 10: 387 402, 2010.
1225. Molinero LL, Miller ML, Evaristo C, Alegre ML. High TCR stimuli prevent induced
regulatory T cell differentiation in a NF-B-dependent manner. J Immunol 186:
4609 4617, 2011.
1226. Monaco C, Gregan SM, Navin TJ, Foxwell BM, Davies AH, Feldmann M. Toll-like
receptor-2 mediates inflammation and matrix degradation in human atherosclerosis.
Circulation 120: 24622469, 2009.
1227. Montecucco F, Vuilleumier N, Pagano S, Lenglet S, Bertolotto M, Braunersreuther
V, Pelli G, Kovari E, Pane B, Spinella G, Pende A, Palombo D, Dallegri F, Mach F,
Roux-Lombard P. Anti-Apolipoprotein A-1 auto-antibodies are active mediators of
atherosclerotic plaque vulnerability. Eur Heart J 32: 412 421, 2011.
1228. Montfort A, Martin PG, Levade T, Benoist H, Segui B. FAN (factor associated with
neutral sphingomyelinase activation), a moonlighting protein in TNF-R1 signaling. J
Leukoc Biol 88: 897903, 2010.
1229. Moon SK, Thompson LJ, Madamanchi N, Ballinger S, Papaconstantinou J, Horaist C,
Runge MS, Patterson C. Aging, oxidative responses, and proliferative capacity in
cultured mouse aortic smooth muscle cells. Am J Physiol Heart Circ Physiol 280:
H2779 H2788, 2001.
1230. Moore KJ, Freeman MW. Scavenger receptors in atherosclerosis: beyond lipid uptake. Arterioscler Thromb Vasc Biol 26: 17021711, 2006.
1231. Moore KJ, Kunjathoor VV, Koehn SL, Manning JJ, Tseng AA, Silver JM, McKee M,
Freeman MW. Loss of receptor-mediated lipid uptake via scavenger receptor A or
CD36 pathways does not ameliorate atherosclerosis in hyperlipidemic mice. J Clin
Invest 115: 21922201, 2005.
1214. Ming XF, Rajapakse AG, Yepuri G, Xiong Y, Carvas JM, Ruffieux J, Scerri I, Wu Z,
Popp K, Li J, Sartori C, Scherrer U, Kwak BR, Montani JP, Yang Z. Arginase II
promotes macrophage inflammatory responses through mitochondrial reactive oxygen species, contributing to insulin resistance and atherogenesis. J Am Heart Assoc 1:
e000992, 2012.
1232. Moore RE, Navab M, Millar JS, Zimetti F, Hama S, Rothblat GH, Rader DJ. Increased
atherosclerosis in mice lacking apolipoprotein A-I attributable to both impaired
reverse cholesterol transport and increased inflammation. Circ Res 97: 763771,
2005.
1215. Minick CR. Immunologic arterial injury in atherogenesis. Ann NY Acad Sci 275: 210
227, 1976.
1233. Morgan BP, Gasque P. Extrahepatic complement biosynthesis: where, when and
why? Clin Exp Immunol 107: 17, 1997.
1234. Morgan MJ, Liu ZG. Reactive oxygen species in TNFalpha-induced signaling and cell
death. Mol Cells 30: 112, 2010.
1518
1235. Morgan MR, Humphries MJ, Bass MD. Synergistic control of cell adhesion by integrins and syndecans. Nat Rev Mol Cell Biol 8: 957969, 2007.
1236. Mortuza R, Chen S, Feng B, Sen S, Chakrabarti S. High glucose induced alteration of
SIRTs in endothelial cells causes rapid aging in a p300 and FOXO regulated pathway.
PLoS ONE 8: e54514, 2013.
1237. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat
Rev Immunol 8: 958 969, 2008.
1238. Mostofsky E, Maclure M, Sherwood JB, Tofler GH, Muller JE, Mittleman MA. Risk of
acute myocardial infarction after the death of a significant person in ones life: the
Determinants of Myocardial Infarction Onset Study. Circulation 125: 491 496, 2012.
1239. Motterle A, Pu X, Wood H, Xiao Q, Gor S, Ng FL, Chan K, Cross F, Shohreh B,
Poston RN, Tucker AT, Caulfield MJ, Ye S. Functional analyses of coronary artery
disease associated variation on chromosome 9p21 in vascular smooth muscle cells.
Hum Mol Genet 21: 4021 4029, 2012.
1240. Moukdar F, Robidoux J, Lyght O, Pi J, Daniel KW, Collins S. Reduced antioxidant
capacity and diet-induced atherosclerosis in uncoupling protein-2-deficient mice. J
Lipid Res 50: 59 70, 2009.
1241. Moulton KS, Heller E, Konerding MA, Flynn E, Palinski W, Folkman J. Angiogenesis
inhibitors endostatin or TNP-470 reduce intimal neovascularization and plaque
growth in apolipoprotein E-deficient mice. Circulation 99: 1726 1732, 1999.
1242. Moulton KS, Olsen BR, Sonn S, Fukai N, Zurakowski D, Zeng X. Loss of collagen
XVIII enhances neovascularization and vascular permeability in atherosclerosis. Circulation 110: 1330 1336, 2004.
1243. Moulton KS, Vakili K, Zurakowski D, Soliman M, Butterfield C, Sylvin E, Lo KM,
Gillies S, Javaherian K, Folkman J. Inhibition of plaque neovascularization reduces
macrophage accumulation and progression of advanced atherosclerosis. Proc Natl
Acad Sci USA 100: 4736 4741, 2003.
1264. Mustafa AK, Gadalla MM, Snyder SH. Signaling by gasotransmitters. Sci Signal 2: re2,
2009.
1265. Na S, Collin O, Chowdhury F, Tay B, Ouyang M, Wang Y, Wang N. Rapid signal
transduction in living cells is a unique feature of mechanotransduction. Proc Natl Acad
Sci USA 105: 6626 6631, 2008.
1266. Nachtigal M, Ghaffar A, Mayer EP. Galectin-3 gene inactivation reduces atherosclerotic lesions and adventitial inflammation in ApoE-deficient mice. Am J Pathol 172:
247255, 2008.
1247. Mullick AE, Soldau K, Kiosses WB, Bell TA 3rd, Tobias PS, Curtiss LK. Increased
endothelial expression of Toll-like receptor 2 at sites of disturbed blood flow exacerbates early atherogenic events. J Exp Med 205: 373383, 2008.
1268. Nadtochiy SM, Baker PR, Freeman BA, Brookes PS. Mitochondrial nitroalkene formation and mild uncoupling in ischaemic preconditioning: implications for cardioprotection. Cardiovasc Res 82: 333340, 2009.
1248. Mullick AE, Tobias PS, Curtiss LK. Modulation of atherosclerosis in mice by Toll-like
receptor 2. J Clin Invest 115: 3149 3156, 2005.
1269. Nagarajan S. Anti-OxLDL IgG blocks OxLDL interaction with CD36, but promotes
FcgammaR, CD32A-dependent inflammatory cell adhesion. Immunol Lett 108: 52
61, 2007.
1249. Mulya A, Lee JY, Gebre AK, Thomas MJ, Colvin PL, Parks JS. Minimal lipidation of
pre-beta HDL by ABCA1 results in reduced ability to interact with ABCA1. Arterioscler Thromb Vasc Biol 27: 1828 1836, 2007.
1250. Mungrue IN, Pagnon J, Kohannim O, Gargalovic PS, Lusis AJ. CHAC1/MGC4504 is a
novel proapoptotic component of the unfolded protein response, downstream of
the ATF4-ATF3-CHOP cascade. J Immunol 182: 466 476, 2009.
1252. Murakami N, Yokomizo T, Okuno T, Shimizu T. G2A is a proton-sensing G proteincoupled receptor antagonized by lysophosphatidylcholine. J Biol Chem 279: 42484
42491, 2004.
1253. Murakami T, Felinski EA, Antonetti DA. Occludin phosphorylation and ubiquitination regulate tight junction trafficking and vascular endothelial growth factor-induced
permeability. J Biol Chem 284: 21036 21046, 2009.
1254. Murakami T, Ockinger J, Yu J, Byles V, McColl A, Hofer AM, Horng T. Critical role
for calcium mobilization in activation of the NLRP3 inflammasome. Proc Natl Acad Sci
USA 109: 1128211287, 2012.
1255. Murase T, Kume N, Korenaga R, Ando J, Sawamura T, Masaki T, Kita T. Fluid shear
stress transcriptionally induces lectin-like oxidized LDL receptor-1 in vascular endothelial cells. Circ Res 83: 328 333, 1998.
1256. Muratoglu SC, Mikhailenko I, Newton C, Migliorini M, Strickland DK. Low density
lipoprotein receptor-related protein 1 (LRP1) forms a signaling complex with platelet-derived growth factor receptor-beta in endosomes and regulates activation of
the MAPK pathway. J Biol Chem 285: 14308 14317, 2010.
1257. Murayama T, Yokode M, Kataoka H, Imabayashi T, Yoshida H, Sano H, Nishikawa S,
Nishikawa SI, Kita T. Intraperitoneal administration of Anti-c-fms monoclonal antibody prevents initial events of atherogenesis but does not reduce the size of advanced lesions in apolipoprotein E-deficient mice. Circulation 99: 1740 1746, 1999.
1258. Murphy AJ, Akhtari M, Tolani S, Pagler T, Bijl N, Kuo CL, Wang M, Sanson M,
Abramowicz S, Welch C, Bochem AE, Kuivenhoven JA, Yvan-Charvet L, Tall AR.
ApoE regulates hematopoietic stem cell proliferation, monocytosis, and monocyte
accumulation in atherosclerotic lesions in mice. J Clin Invest 121: 4138 4149, 2011.
1259. Murphy AJ, Westerterp M, Yvan-Charvet L, Tall AR. Anti-atherogenic mechanisms
of high density lipoprotein: effects on myeloid cells. Biochim Biophys Acta 2011.
1260. Murphy AJ, Woollard KJ, Suhartoyo A, Stirzaker RA, Shaw J, Sviridov D, ChinDusting JP. Neutrophil activation is attenuated by high-density lipoprotein and apolipoprotein A-I in in vitro and in vivo models of inflammation. Arterioscler Thromb Vasc
Biol 31: 13331341, 2011.
1261. Murray HW, Spitalny GL, Nathan CF. Activation of mouse peritoneal macrophages
in vitro and in vivo by interferon-gamma. J Immunol 134: 1619 1622, 1985.
1262. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets.
Nat Rev Immunol 11: 723737, 2011.
1263. Mustafa AK, Gadalla MM, Sen N, Kim S, Mu W, Gazi SK, Barrow RK, Yang G, Wang
R, Snyder SH. H2S signals through protein S-sulfhydration. Sci Signal 2: ra72, 2009.
1519
PAUL N. HOPKINS
1283. Napoli C, Martin-Padura I, de Nigris F, Giorgio M, Mansueto G, Somma P, Condorelli M, Sica G, De Rosa G, Pelicci P. Deletion of the p66Shc longevity gene
reduces systemic and tissue oxidative stress, vascular cell apoptosis, and early
atherogenesis in mice fed a high-fat diet. Proc Natl Acad Sci USA 100: 21122116,
2003.
1284. Narasimha AJ, Watanabe J, Ishikawa TO, Priceman SJ, Wu L, Herschman HR, Reddy
ST. Absence of myeloid COX-2 attenuates acute inflammation but does not influence development of atherosclerosis in apolipoprotein E null mice. Arterioscler
Thromb Vasc Biol 30: 260 268, 2010.
1285. Naruko T, Ueda M, Haze K, van der Wal AC, van der Loos CM, Itoh A, Komatsu R,
Ikura Y, Ogami M, Shimada Y, Ehara S, Yoshiyama M, Takeuchi K, Yoshikawa J,
Becker AE. Neutrophil infiltration of culprit lesions in acute coronary syndromes.
Circulation 106: 2894 2900, 2002.
1286. Narumiya S, Furuyashiki T. Fever, inflammation, pain and beyond: prostanoid receptor research during these 25 years. FASEB J 25: 813 818, 2011.
1287. Nauli SM, Kawanabe Y, Kaminski JJ, Pearce WJ, Ingber DE, Zhou J. Endothelial cilia
are fluid shear sensors that regulate calcium signaling and nitric oxide production
through polycystin-1. Circulation 117: 11611171, 2008.
1288. Navab M, Ananthramaiah GM, Reddy ST, Van Lenten BJ, Ansell BJ, Fonarow GC,
Vahabzadeh K, Hama S, Hough G, Kamranpour N, Berliner JA, Lusis AJ, Fogelman
AM. The oxidation hypothesis of atherogenesis: the role of oxidized phospholipids
and HDL. J Lipid Res 45: 9931007, 2004.
1290. Navab M, Reddy ST, Van Lenten BJ, Fogelman AM. HDL and cardiovascular disease:
atherogenic and atheroprotective mechanisms. Nat Rev Cardiol 8: 222232, 2011.
1291. Nayak L, Lin Z, Jain MK. Go with the flow: how Kruppel-like factor 2 regulates the
vasoprotective effects of shear stress. Antioxid Redox Signal 15: 1449 1461, 2011.
1292. Nemenoff RA, Horita H, Ostriker AC, Furgeson SB, Simpson PA, VanPutten V,
Crossno J, Offermanns S, Weiser-Evans MC. SDF-1alpha induction in mature
smooth muscle cells by inactivation of PTEN is a critical mediator of exacerbated
injury-induced neointima formation. Arterioscler Thromb Vasc Biol 31: 1300 1308,
2011.
1293. Netea MG, Kullberg BJ, Demacker PN, Jacobs LE, Verver-Jansen TJ, Hijmans A, van
Tits LH, Hoenderop JG, Willems PH, Van der Meer JW, Stalenhoef AF. Native LDL
potentiate TNF alpha and IL-8 production by human mononuclear cells. J Lipid Res
43: 10651071, 2002.
1294. Neumann CA, Cao J, Manevich Y. Peroxiredoxin 1 and its role in cell signaling. Cell
Cycle 8: 4072 4078, 2009.
1295. Newby AC, George SJ, Ismail Y, Johnson JL, Sala-Newby GB, Thomas AC. Vulnerable atherosclerotic plaque metalloproteinases and foam cell phenotypes. Thromb
Haemost 101: 1006 1011, 2009.
1296. Newton CS, Loukinova E, Mikhailenko I, Ranganathan S, Gao Y, Haudenschild C,
Strickland DK. Platelet-derived growth factor receptor-beta (PDGFR-beta) activation promotes its association with the low density lipoprotein receptor-related protein (LRP). Evidence for co-receptor function. J Biol Chem 280: 2787227878, 2005.
1297. Ng CJ, Bourquard N, Grijalva V, Hama S, Shih DM, Navab M, Fogelman AM, Lusis AJ,
Young S, Reddy ST. Paraoxonase-2 deficiency aggravates atherosclerosis in mice
despite lower apolipoprotein-B-containing lipoproteins: anti-atherogenic role for
paraoxonase-2. J Biol Chem 281: 2949129500, 2006.
1298. Ng HP, Burris RL, Nagarajan S. Attenuated atherosclerotic lesions in apoEFcgamma-chain-deficient hyperlipidemic mouse model is associated with inhibition
of Th17 cells and promotion of regulatory T cells. J Immunol 187: 6082 6093, 2011.
1299. Ni M, Chen WQ, Zhang Y. Animal models and potential mechanisms of plaque
destabilisation and disruption. Heart 95: 13931398, 2009.
1300. Ni M, Chen WQ, Zhang Y. Animal models and potential mechanisms of plaque
destabilisation and disruption. Heart 95: 13931398, 2009.
1520
1302. Ni W, Zhan Y, He H, Maynard E, Balschi JA, Oettgen P. Ets-1 is a critical transcriptional regulator of reactive oxygen species and p47(phox) gene expression in response to angiotensin II. Circ Res 101: 985994, 2007.
1303. Nicholls SJ, Hazen SL. Myeloperoxidase and cardiovascular disease. Arterioscler
Thromb Vasc Biol 25: 11021111, 2005.
1304. Nicholls SJ, Hazen SL. Myeloperoxidase, modified lipoproteins, atherogenesis. J Lipid
Res 50: S346 351, 2009.
1305. Nievelstein PF, Fogelman AM, Mottino G, Frank JS. Lipid accumulation in rabbit
aortic intima 2 hours after bolus infusion of low density lipoprotein. A deep-etch and
immunolocalization study of ultrarapidly frozen tissue. Arterioscler Thromb 11: 1795
1805, 1991.
1306. Nigro P, Abe Ji Woo CH, Satoh K, McClain C, ODell MR, Lee H, Lim JH, Li Jd Heo
KS, Fujiwara K, Berk BC. PKC decreases eNOS protein stability via inhibitory
phosphorylation of ERK5. Blood 116: 19711979, 2010.
1307. Nihira K, Ando Y, Yamaguchi T, Kagami Y, Miki Y, Yoshida K. Pim-1 controls NFkappaB signalling by stabilizing RelA/p65. Cell Death Differ 17: 689 698, 2010.
1308. Nilsson-Berglund LM, Zetterqvist AV, Nilsson-Ohman J, Sigvardsson M, Gonzalez
Bosc LV, Smith ML, Salehi A, Agardh E, Fredrikson GN, Agardh CD, Nilsson J,
Wamhoff BR, Hultgardh-Nilsson A, Gomez MF. Nuclear factor of activated T cells
regulates osteopontin expression in arterial smooth muscle in response to diabetesinduced hyperglycemia. Arterioscler Thromb Vasc Biol 30: 218 224, 2010.
1309. Nilsson J, Nilsson LM, Chen YW, Molkentin JD, Erlinge D, Gomez MF. High glucose
activates nuclear factor of activated T cells in native vascular smooth muscle. Arterioscler Thromb Vasc Biol 26: 794 800, 2006.
1310. Nilsson LM, Nilsson-Ohman J, Zetterqvist AV, Gomez MF. Nuclear factor of activated T-cells transcription factors in the vasculature: the good guys or the bad guys?
Curr Opin Lipidol 19: 483 490, 2008.
1311. Nishino T, Okamoto K, Eger BT, Pai EF. Mammalian xanthine oxidoreductase:
mechanism of transition from xanthine dehydrogenase to xanthine oxidase. FEBS J
275: 3278 3289, 2008.
1312. Niu J, Profirovic J, Pan H, Vaiskunaite R, Voyno-Yasenetskaya T. G Protein betagamma subunits stimulate p114RhoGEF, a guanine nucleotide exchange factor for
RhoA and Rac1: regulation of cell shape and reactive oxygen species production. Circ
Res 93: 848 856, 2003.
1313. Niu XL, Molnar K, Tchivilev I, Vendrov AE, Madamanchi NR, Runge MS. Abstract
20332: oxidative inactivation of protein tyrosine phosphatases underlies aging-associated increases in inflammation in VSMC and atherosclerosis in ApoE/ mice.
Circulation 122: A20332, 2010.
1314. Nogueira E, Fidalgo M, Molnar A, Kyriakis J, Force T, Zalvide J, Pombo CM. SOK1
translocates from the Golgi to the nucleus upon chemical anoxia and induces apoptotic cell death. J Biol Chem 283: 16248 16258, 2008.
1315. Norata GD, Catapano AL. Molecular mechanisms responsible for the antiinflammatory and protective effect of HDL on the endothelium. Vasc Health Risk Manag 1:
119 129, 2005.
1316. Norata GD, Grigore L, Raselli S, Redaelli L, Hamsten A, Maggi F, Eriksson P, Catapano AL. Postprandial endothelial dysfunction in hypertriglyceridemic subjects: molecular mechanisms and gene expression studies. Atherosclerosis 193: 321327,
2007.
1317. Norata GD, Marchesi P, Pulakazhi Venu VK, Pasqualini F, Anselmo A, Moalli F,
Pizzitola I, Garlanda C, Mantovani A, Catapano AL. Deficiency of the long pentraxin
PTX3 promotes vascular inflammation and atherosclerosis. Circulation 120: 699
708, 2009.
1318. Norata GD, Pirillo A, Ammirati E, Catapano AL. Emerging role of high density
lipoproteins as a player in the immune system. Atherosclerosis 220: 1121, 2012.
1319. Norata GD, Pirillo A, Callegari E, Hamsten A, Catapano AL, Eriksson P. Gene
expression and intracellular pathways involved in endothelial dysfunction induced by
VLDL and oxidised VLDL. Cardiovasc Res 59: 169 180, 2003.
1289. Navab M, Imes SS, Hama SY, Hough GP, Ross LA, Bork RW, Valente AJ, Berliner JA,
Drinkwater DC, Laks H. Monocyte transmigration induced by modification of low
density lipoprotein in cocultures of human aortic wall cells is due to induction of
monocyte chemotactic protein 1 synthesis and is abolished by high density lipoprotein. J Clin Invest 88: 2039 2046, 1991.
1301. Ni M, Zhang M, Ding SF, Chen WQ, Zhang Y. Micro-ultrasound imaging assessment
of carotid plaque characteristics in apolipoprotein-E knockout mice. Atherosclerosis
197: 64 71, 2008.
1329. Oh JY, Giles N, Landar A, Darley-Usmar V. Accumulation of 15-deoxy-delta(12,14)prostaglandin J2 adduct formation with Keap1 over time: effects on potency for
intracellular antioxidant defence induction. Biochem J 411: 297306, 2008.
1330. Oh SF, Pillai PS, Recchiuti A, Yang R, Serhan CN. Pro-resolving actions and stereoselective biosynthesis of 18S E-series resolvins in human leukocytes and murine
inflammation. J Clin Invest 121: 569 581, 2011.
1331. Ohlenschlaeger T, Garred P, Madsen HO, Jacobsen S. Mannose-binding lectin variant alleles and the risk of arterial thrombosis in systemic lupus erythematosus. N Engl
J Med 351: 260 267, 2004.
1349. Oram JF, Heinecke JW. ATP-binding cassette transporter A1: a cell cholesterol
exporter that protects against cardiovascular disease. Physiol Rev 85: 13431372,
2005.
1333. Ohta H, Wada H, Niwa T, Kirii H, Iwamoto N, Fujii H, Saito K, Sekikawa K, Seishima
M. Disruption of tumor necrosis factor-alpha gene diminishes the development of
atherosclerosis in ApoE-deficient mice. Atherosclerosis 180: 1117, 2005.
1334. Ohtsubo T, Matsumura K, Sakagami K, Fujii K, Tsuruya K, Noguchi H, Rovira II,
Finkel T, Iida M. Xanthine oxidoreductase depletion induces renal interstitial fibrosis
through aberrant lipid and purine accumulation in renal tubules. Hypertension 54:
868 876, 2009.
1335. Okabe Ta Shimada K, Hattori M, Murayama T, Yokode M, Kita T, Kishimoto C.
Swimming reduces the severity of atherosclerosis in apolipoprotein E deficient mice
by antioxidant effects. Cardiovasc Res 74: 537545, 2007.
1336. Okahara K, Sun B, Kambayashi J. Upregulation of prostacyclin synthesis-related gene
expression by shear stress in vascular endothelial cells. Arterioscler Thromb Vasc Biol
18: 19221926, 1998.
1337. Okahara K, Sun B, Kawasaki T, Monden M, Kambayashi JI. Expression of plateletactivating factor receptor transcript-2 is induced by shear stress in HUVEC. Prostaglandins 55: 323329, 1998.
1350. Oram JF, Wolfbauer G, Tang C, Davidson WS, Albers JJ. An amphipathic helical
region of the N-terminal barrel of phospholipid transfer protein is critical for
ABCA1-dependent cholesterol efflux. J Biol Chem 283: 1154111549, 2008.
1351. Orozco LD, Kapturczak MH, Barajas B, Wang X, Weinstein MM, Wong J, Deshane
J, Bolisetty S, Shaposhnik Z, Shih DM, Agarwal A, Lusis AJ, Araujo JA. Heme oxygenase-1 expression in macrophages plays a beneficial role in atherosclerosis. Circ Res
100: 17031711, 2007.
1352. Orr AW, Ginsberg MH, Shattil SJ, Deckmyn H, Schwartz MA. Matrix-specific suppression of integrin activation in shear stress signaling. Mol Biol Cell 17: 4686 4697,
2006.
1353. Orr AW, Hahn C, Blackman BR, Schwartz MA. p21-activated kinase signaling regulates oxidant-dependent NF-B activation by flow. Circ Res 103: 671 679, 2008.
1354. Oshima S, Turer EE, Callahan JA, Chai S, Advincula R, Barrera J, Shifrin N, Lee B,
Benedict Yen TS, Woo T, Malynn BA, Ma A. ABIN-1 is a ubiquitin sensor that
restricts cell death and sustains embryonic development. Nature 457: 906 909,
2009.
1355. Osto E, Kouroedov A, Mocharla P, Akhmedov A, Besler C, Rohrer L, von Eckardstein A, Iliceto S, Volpe M, Luscher TF, Cosentino F. Inhibition of protein kinase C
prevents foam cell formation by reducing scavenger receptor A expression in human
macrophages. Circulation 118: 2174 2182, 2008.
1356. Ota H, Eto M, Kano MR, Kahyo T, Setou M, Ogawa S, Iijima K, Akishita M, Ouchi Y.
Induction of endothelial nitric oxide synthase, SIRT1, and catalase by statins inhibits
1521
PAUL N. HOPKINS
endothelial senescence through the Akt pathway. Arterioscler Thromb Vasc Biol 30:
22052211, 2010.
1357. Ott SJ, El Mokhtari NE, Musfeldt M, Hellmig S, Freitag S, Rehman A, Kuhbacher T,
Nikolaus S, Namsolleck P, Blaut M, Hampe J, Sahly H, Reinecke A, Haake N,
Gunther R, Kruger D, Lins M, Herrmann G, Folsch UR, Simon R, Schreiber S.
Detection of diverse bacterial signatures in atherosclerotic lesions of patients with
coronary heart disease. Circulation 113: 929 937, 2006.
1358. Otte LA, Bell KS, Loufrani L, Yeh JC, Melchior B, Dao DN, Stevens HY, White CR,
Frangos JA. Rapid changes in shear stress induce dissociation of a Gq/11-platelet
endothelial cell adhesion molecule-1 complex. J Physiol 587: 23652373, 2009.
1359. Otten JJ, de Jager SC, Bermudez B, Bot I, van Berkel TJ, Kavelaars A, Heijnen CJ,
Biessen EA. Abstract 15514: lymphoid but not phagocytic deficiency in G proteincoupled receptor kinase 2 decreases atherosclerotic plaque formation in LDLr/
mice. Circulation 122: A15514, 2010.
1360. Ouabed A, Hubert FX, Chabannes D, Gautreau L, Heslan M, Josien R. Differential control of T regulatory cell proliferation and suppressive activity by mature
plasmacytoid versus conventional spleen dendritic cells. J Immunol 180: 5862
5870, 2008.
1361. Oumouna-Benachour K, Hans CP, Suzuki Y, Naura A, Datta R, Belmadani S, Fallon
K, Woods C, Boulares AH. Poly(ADP-Ribose) polymerase inhibition reduces atherosclerotic plaque size and promotes factors of plaque stability in apolipoprotein
E-deficient mice: effects on macrophage recruitment, nuclear factor-B nuclear
translocation, and foam cell death. Circulation 115: 24422450, 2007.
1363. Ozsoy HZ, Sivasubramanian N, Wieder ED, Pedersen S, Mann DL. Oxidative stress
promotes ligand-independent and enhanced ligand-dependent tumor necrosis factor receptor signaling. J Biol Chem 283: 23419 23428, 2008.
1364. Pacher P, Nivorozhkin A, Szabo C. Therapeutic effects of xanthine oxidase inhibitors: renaissance half a century after the discovery of allopurinol. Pharmacol Rev 58:
87114, 2006.
1365. Pacher P, Szabo C. Role of poly(ADP-ribose) polymerase 1 (PARP-1) in cardiovascular diseases: the therapeutic potential of PARP inhibitors. Cardiovasc Drug Rev 25:
235260, 2007.
1366. Packard RR, Maganto-Garcia E, Gotsman I, Tabas I, Libby P, Lichtman AH.
CD11c() dendritic cells maintain antigen processing, presentation capabilities, and
CD4() T-cell priming efficacy under hypercholesterolemic conditions associated
with atherosclerosis. Circ Res 103: 965973, 2008.
1367. Packer L, Cadenas E. Lipoic acid: energy metabolism and redox regulation of transcription and cell signaling. J Clin Biochem Nutr 48: 26 32, 2011.
1368. Page JH, Ma J, Chiuve SE, Stampfer MJ, Selhub J, Manson JE, Rimm EB. Plasma total
cysteine and total homocysteine and risk of myocardial infarction in women: A
prospective study. Am Heart J 159: 599 604, 2010.
1369. Pahakis MY, Kosky JR, Dull RO, Tarbell JM. The role of endothelial glycocalyx components in mechanotransduction of fluid shear stress. Biochem Biophys Res Commun
355: 228 233, 2007.
1370. Paigen B, Holmes PA, Novak EK, Swank RT. Analysis of atherosclerosis susceptibility in mice with genetic defects in platelet function. Arteriosclerosis 10: 648
652, 1990.
1371. Palinski W, Miller E, Witztum JL. Immunization of low density lipoprotein (LDL)
receptor-deficient rabbits with homologous malondialdehyde-modified LDL reduces atherogenesis. Proc Natl Acad Sci USA 92: 821 825, 1995.
1372. Palmby TR, Rosenfeldt H, Gutkind JS. G protein-coupled receptors, signal fidelity,
and cell transformation. In: Transduction Mechanisms in Cellular Signaling, edited by
Dennis EA and Bradshaw RA. New York: Academic, 2011, p. 467 480.
1373. Pan JH, Sukhova GK, Yang JT, Wang B, Xie T, Fu H, Zhang Y, Satoskar AR, David JR,
Metz CN, Bucala R, Fang K, Simon DI, Chapman HA, Libby P, Shi GP. Macrophage
migration inhibitory factor deficiency impairs atherosclerosis in low-density lipoprotein receptor-deficient mice. Circulation 109: 3149 3153, 2004.
1522
1375. Pandey D, Fulton DJ. Molecular regulation of NADPH oxidase 5 via the MAPK
pathway. Am J Physiol Heart Circ Physiol 300: H1336 H1344, 2011.
1376. Pandey D, Gratton JP, Rafikov R, Black SM, Fulton DJ. Calcium/calmodulin-dependent kinase II mediates the phosphorylation and activation of NADPH oxidase 5. Mol
Pharmacol 80: 407 415, 2011.
1377. Paneni F, Mocharla P, Akhmedov A, Osto E, Volpe M, Luscher TF, Cosentino F.
Abstract 12438: vascular hyperglycemic memory is abolished by in vivo silencing of
the mitochondrial adaptor p66Shc. Circulation 124: A12438, 2011.
1378. Paneni F, Mocharla P, Osto E, Volpe M, Luscher TF, Cosentino F. Abstract 16655:
role of PKCII/p66Shc pathway as a determinant of metabolic memory in human
endothelial cells. Circulation 122: A16655, 2010.
1379. Papadopoulou C, Corrigall V, Taylor PR, Poston RN. The role of the chemokines
MCP-1, GRO-alpha, IL-8 and their receptors in the adhesion of monocytic cells to
human atherosclerotic plaques. Cytokine 43: 181186, 2008.
1380. Park D, Han CZ, Elliott MR, Kinchen JM, Trampont PC, Das S, Collins S, Lysiak JJ,
Hoehn KL, Ravichandran KS. Continued clearance of apoptotic cells critically depends on the phagocyte Ucp2 protein. Nature 477: 220 224, 2011.
1381. Park JG, Yoo JY, Jeong SJ, Choi JH, Lee MR, Lee MN, Hwa Lee J, Kim HC, Jo H, Yu
DY, Kang SW, Rhee SG, Lee MH, Oh GT. Peroxiredoxin 2 deficiency exacerbates
atherosclerosis in apolipoprotein E-deficient mice. Circ Res 109: 739 749, 2011.
1382. Park SY, Lee JH, Kim YK, Kim CD, Rhim BY, Lee WS, Hong KW. Cilostazol prevents
remnant lipoprotein particle-induced monocyte adhesion to endothelial cells by
suppression of adhesion molecules and monocyte chemoattractant protein-1 expression via lectin-like receptor for oxidized low-density lipoprotein receptor activation. J Pharmacol Exp Ther 312: 12411248, 2005.
1383. Park SY, Schinkmann KA, Avraham S. RAFTK/Pyk2 mediates LPA-induced PC12 cell
migration. Cell Signal 18: 10631071, 2006.
1384. Park TS, Panek RL, Mueller SB, Hanselman JC, Rosebury WS, Robertson AW, Kindt
EK, Homan R, Karathanasis SK, Rekhter MD. Inhibition of sphingomyelin synthesis
reduces atherogenesis in apolipoprotein E-knockout mice. Circulation 110: 3465
3471, 2004.
1385. Park TS, Rosebury W, Kindt EK, Kowala MC, Panek RL. Serine palmitoyltransferase
inhibitor myriocin induces the regression of atherosclerotic plaques in hyperlipidemic ApoE-deficient mice. Pharmacol Res 58: 4551, 2008.
1386. Parks BW, Lusis AJ, Kabarowski JH. Loss of the lysophosphatidylcholine effector,
G2A, ameliorates aortic atherosclerosis in low-density lipoprotein receptor knockout mice. Arterioscler Thromb Vasc Biol 26: 27032709, 2006.
1387. Parks BW, Srivastava R, Yu S, Kabarowski JH. ApoE-dependent modulation of HDL
and atherosclerosis by G2A in LDL receptor-deficient mice independent of bone
marrow-derived cells. Arterioscler Thromb Vasc Biol 29: 539 547, 2009.
1388. Parmar KM, Nambudiri V, Dai G, Larman HB, Gimbrone MA Jr, Garcia-Cardena G.
Statins exert endothelial atheroprotective effects via the KLF2 transcription factor. J
Biol Chem 280: 26714 26719, 2005.
1389. Parsons JT, Horwitz AR, Schwartz MA. Cell adhesion: integrating cytoskeletal dynamics and cellular tension. Nat Rev Mol Cell Biol 11: 633 643, 2010.
1390. Partridge J, Carlsen H, Enesa K, Chaudhury H, Zakkar M, Luong L, Kinderlerer A,
Johns M, Blomhoff R, Mason JC, Haskard DO, Evans PC. Laminar shear stress acts as
a switch to regulate divergent functions of NF-kappaB in endothelial cells. FASEB J 21:
35533561, 2007.
1391. Passerini AG, Polacek DC, Shi C, Francesco NM, Manduchi E, Grant GR, Pritchard
WF, Powell S, Chang GY, Stoeckert CJ Jr, Davies PF. Coexisting proinflammatory
and antioxidative endothelial transcription profiles in a disturbed flow region of the
adult porcine aorta. Proc Natl Acad Sci USA 101: 24822487, 2004.
1392. Pastorino JG, Hoek JB. Regulation of hexokinase binding to VDAC. J Bioenerg
Biomembr 40: 171182, 2008.
1393. Patel S, Thelander EM, Hernandez M, Montenegro J, Hassing H, Burton C, Mundt S,
Hermanowski-Vosatka A, Wright SD, Chao YS, Detmers PA. ApoE(/) mice
1362. Overton CD, Yancey PG, Major AS, Linton MF, Fazio S. Deletion of macrophage
LDL receptor-related protein increases atherogenesis in the mouse. Circ Res 100:
670 677, 2007.
1394. Patel VB, Bodiga S, Fan D, Das SK, Wang Z, Wang W, Basu R, Zhong J, Kassiri Z,
Oudit GY. Cardioprotective effects mediated by angiotensin II type 1 receptor
blockade and enhancing angiotensin 17 in experimental heart failure in angiotensinconverting enzyme 2-null mice. Hypertension 59: 11951203, 2012.
1414. Petzold T, Orr AW, Hahn C, Jhaveri KA, Parsons JT, Schwartz MA. Focal adhesion
kinase modulates activation of NF-kappaB by flow in endothelial cells. Am J Physiol
Cell Physiol 297: C814 C822, 2009.
1395. Patricia MK, Kim JA, Harper CM, Shih PT, Berliner JA, Natarajan R, Nadler JL,
Hedrick CC. Lipoxygenase products increase monocyte adhesion to human aortic
endothelial cells. Arterioscler Thromb Vasc Biol 19: 26152622, 1999.
1396. Patricia MK, Natarajan R, Dooley AN, Hernandez F, Gu JL, Berliner JA, Rossi JJ,
Nadler JL, Meidell RS, Hedrick CC. Adenoviral delivery of a leukocyte-type 12
lipoxygenase ribozyme inhibits effects of glucose and platelet-derived growth factor
in vascular endothelial and smooth muscle cells. Circ Res 88: 659 665, 2001.
1416. Pi X, Lockyer P, Dyer LA, Schisler JC, Russell B, Carey S, Sweet DT, Chen Z, Tzima
E, Willis MS, Homeister JW, Moser M, Patterson C. Bmper inhibits endothelial
expression of inflammatory adhesion molecules and protects against atherosclerosis.
Arterioscler Thromb Vasc Biol 32: 2214 2222, 2012.
1397. Paul A, Chang BHJ, Li L, Yechoor VK, Chan L. Deficiency of adipose differentiationrelated protein impairs foam cell formation and protects against atherosclerosis. Circ
Res 102: 14921501, 2008.
1417. Pierce GL, Lesniewski LA, Lawson BR, Beske SD, Seals DR. Nuclear factor-kappaB
activation contributes to vascular endothelial dysfunction via oxidative stress in overweight/obese middle-aged and older humans. Circulation 119: 1284 1292, 2009.
1398. Paulsen CE, Carroll KS. Orchestrating redox signaling networks through regulatory
cysteine switches. ACS Chem Biol 5: 47 62, 2010.
1418. Pincheira R, Castro AF, Ozes ON, Idumalla PS, Donner DB. Type 1 TNF receptor
forms a complex with and uses Jak2 and c-Src to selectively engage signaling pathways that regulate transcription factor activity. J Immunol 181: 1288 1298, 2008.
1399. Paulson KE, Zhu SN, Chen M, Nurmohamed S, Jongstra-Bilen J, Cybulsky MI. Resident intimal dendritic cells accumulate lipid and contribute to the initiation of atherosclerosis. Circ Res 106: 383390, 2010.
1419. Plaisance EP, Lukasova M, Offermanns S, Zhang Y, Cao G, Judd RL. Niacin stimulates
adiponectin secretion through the GPR109A receptor. Am J Physiol Endocrinol Metab
296: E549 E558, 2009.
1420. Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat
Rev Immunol 5: 375386, 2005.
1401. Peng C, Cho YY, Zhu F, Xu YM, Wen W, Ma WY, Bode AM, Dong Z. RSK2 mediates
NF-B activity through the phosphorylation of IkappaBalpha in the TNF-R1 pathway. FASEB J 24: 3490 3499, 2010.
1421. Plihtari R, Hurt-Camejo E, rni K, Kovanen PT. Proteolysis sensitizes LDL particles
to phospholipolysis by secretory phospholipase A2 group V and secretory sphingomyelinase. J Lipid Res 51: 18011809, 2010.
1402. Peng L, Bhatia N, Parker AC, Zhu Y, Fay WP. Endogenous vitronectin and plasminogen activator inhibitor-1 promote neointima formation in murine carotid arteries.
Arterioscler Thromb Vasc Biol 22: 934 939, 2002.
1422. Pluddemann A, Neyen C, Gordon S. Macrophage scavenger receptors and hostderived ligands. Methods 43: 207217, 2007.
1403. Pennings M, Hildebrand RB, Ye D, Kunne C, Van Berkel TJ, Groen AK, Van Eck M.
Bone marrow-derived multidrug resistance protein ABCB4 protects against atherosclerotic lesion development in LDL receptor knockout mice. Cardiovasc Res 76:
175183, 2007.
1423. Pluskota E, Stenina OI, Krukovets I, Szpak D, Topol EJ, Plow EF. Mechanism and
effect of thrombospondin-4 polymorphisms on neutrophil function. Blood 106:
3970 3978, 2005.
1424. Pober JS, Min W, Bradley JR. Mechanisms of endothelial dysfunction, injury, and
death. Annu Rev Pathol 4: 7195, 2009.
1404. Perez J, Torres RA, Rocic P, Cismowski MJ, Weber DS, Darley-Usmar VM, Lucchesi
PA. PYK2 signaling is required for PDGF-dependent vascular smooth muscle cell
proliferation. Am J Physiol Cell Physiol 301: C242C251, 2011.
1425. Pober JS, Sessa WC. Evolving functions of endothelial cells in inflammation. Nat Rev
Immunol 7: 803 815, 2007.
1405. Perkins ND. Integrating cell-signalling pathways with NF-kappaB and IKK function.
Nat Rev Mol Cell Biol 8: 49 62, 2007.
1426. Podrez EA, Byzova TV, Febbraio M, Salomon RG, Ma Y, Valiyaveettil M, Poliakov E,
Sun M, Finton PJ, Curtis BR, Chen J, Zhang R, Silverstein RL, Hazen SL. Platelet CD36
links hyperlipidemia, oxidant stress and a prothrombotic phenotype. Nat Med 13:
1086 1095, 2007.
1406. Perlmutter DH, Dinarello CA, Punsal PI, Colten HR. Cachectin/tumor necrosis
factor regulates hepatic acute-phase gene expression. J Clin Invest 78: 1349 1354,
1986.
1407. Perretti M, DAcquisto F. Annexin A1 and glucocorticoids as effectors of the resolution of inflammation. Nat Rev Immunol 9: 6270, 2009.
1408. Perrotta C, Clementi E. Biological roles of acid and neutral sphingomyelinases and
their regulation by nitric oxide. Physiology 25: 64 71, 2010.
1409. Persson L, Boren J, Robertson AK, Wallenius V, Hansson GK, Pekna M. Lack of
complement factor C3, but not factor B, increases hyperlipidemia and atherosclerosis in apolipoprotein E/ low-density lipoprotein receptor/ mice. Arterioscler Thromb Vasc Biol 24: 10621067, 2004.
1410. Peter A, Weigert C, Staiger H, Rittig K, Cegan A, Lutz P, Machicao F, Haring HU,
Schleicher E. Induction of stearoyl-CoA desaturase protects human arterial endothelial cells against lipotoxicity. Am J Physiol Endocrinol Metab 295: E339 E349, 2008.
1411. Peter C, Wesselborg S, Herrmann M, Lauber K. Dangerous attraction: phagocyte
recruitment and danger signals of apoptotic and necrotic cells. Apoptosis 15: 1007
1028, 2010.
1412. Petrovan RJ, Kaplan CD, Reisfeld RA, Curtiss LK. DNA vaccination against VEGF
receptor 2 reduces atherosclerosis in LDL receptor-deficient mice. Arterioscler
Thromb Vasc Biol 27: 10951100, 2007.
1427. Podrez EA, Schmitt D, Hoff HF, Hazen SL. Myeloperoxidase-generated reactive
nitrogen species convert LDL into an atherogenic form in vitro. J Clin Invest 103:
15471560, 1999.
1428. Poeckel D, Funk CD. The 5-lipoxygenase/leukotriene pathway in preclinical models
of cardiovascular disease. Cardiovasc Res 86: 243253, 2010.
1429. Poeckel D, Zemski Berry KA, Murphy RC, Funk CD. Dual 12/15- and 5-lipoxygenase
deficiency in macrophages alters arachidonic acid metabolism and attenuates peritonitis and atherosclerosis in ApoE knock-out mice. J Biol Chem 284: 2107721089,
2009.
1430. Ponnuswamy P, Ostermeier E, Schrottle A, Chen J, Huang PL, Ertl G, Nieswandt B,
Kuhlencordt PJ. Oxidative stress and compartment of gene expression determine
proatherosclerotic effects of inducible nitric oxide synthase. Am J Pathol 174: 2400
2410, 2009.
1431. Poon IK, Hulett MD, Parish CR. Molecular mechanisms of late apoptotic/necrotic
cell clearance. Cell Death Differ 17: 381397, 2010.
1432. Potenza MA, Addabbo F, Montagnani M. Vascular actions of insulin with implications
for endothelial dysfunction. Am J Physiol Endocrinol Metab 297: E568 E577, 2009.
1433. Potteaux S, Combadiere C, Esposito B, Lecureuil C, Ait-Oufella H, Merval R, Ardouin P, Tedgui A, Mallat Z. Role of bone marrow-derived CC-chemokine receptor
1523
1400. Pejnovic N, Vratimos A, Lee SH, Popadic D, Takeda K, Akira S, Chan WL. Increased
atherosclerotic lesions and Th17 in interleukin-18 deficient apolipoprotein E-knockout mice fed high-fat diet. Mol Immunol 47: 37 45, 2009.
PAUL N. HOPKINS
5 in the development of atherosclerosis of low-density lipoprotein receptor knockout mice. Arterioscler Thromb Vasc Biol 26: 1858 1863, 2006.
1434. Potteaux S, Gautier EL, Hutchison SB, van Rooijen N, Rader DJ, Thomas MJ, SorciThomas MG, Randolph GJ. Suppressed monocyte recruitment drives macrophage
removal from atherosclerotic plaques of Apoe/ mice during disease regression.
J Clin Invest 121: 20252036, 2011.
1435. Pourcet B, Feig JE, Vengrenyuk Y, Hobbs AJ, Kepka-Lenhart D, Garabedian MJ,
Morris SM Jr, Fisher EA, Pineda-Torra I. LXRalpha regulates macrophage arginase 1
through PU.1 and interferon regulatory factor 8. Circ Res 109: 492501, 2011.
1454. Rajavashisth T, Qiao JH, Tripathi S, Tripathi J, Mishra N, Hua M, Wang XP, Loussararian A, Clinton S, Libby P, Lusis A. Heterozygous osteopetrotic (op) mutation
reduces atherosclerosis in LDL receptor- deficient mice. J Clin Invest 101: 2702
2710, 1998.
1436. Powell-Braxton L, Veniant M, Latvala RD, Hirano KI, Won WB, Ross J, Dybdal N,
Zlot CH, Young SG, Davidson NO. A mouse model of human familial hypercholesterolemia: markedly elevated low density lipoprotein cholesterol levels and severe
atherosclerosis on a low-fat chow diet. Nat Med 4: 934 938, 1998.
1437. Preusch MR, Rattazzi M, Albrecht C, Merle U, Tuckermann J, Schutz G, Blessing E,
Zoppellaro G, Pauletto P, Krempien R, Rosenfeld ME, Katus HA, Bea F. Critical role
of macrophages in glucocorticoid driven vascular calcification in a mouse-model of
atherosclerosis. Arterioscler Thromb Vasc Biol 28: 2158 2164, 2008.
1438. Probst-Kepper M, Buer J. FOXP3 and GARP (LRRC32): the master and its minion.
Biol Direct 5: 8, 2010.
1439. Proctor BM, Ren J, Chen Z, Schneider JG, Coleman T, Lupu TS, Semenkovich CF,
Muslin AJ. Grb2 is required for atherosclerotic lesion formation. Arterioscler Thromb
Vasc Biol 27: 13611367, 2007.
1441. Puri KD, Doggett TA, Huang CY, Douangpanya J, Hayflick JS, Turner M, Penninger
J, Diacovo TG. The role of endothelial PI3Kgamma activity in neutrophil trafficking.
Blood 106: 150 157, 2005.
1442. Qiang L, Tsuchiya K, Kim-Muller JY, Lin HV, Welch C, Accili D. Increased atherosclerosis and endothelial dysfunction in mice bearing constitutively deacetylated
alleles of Foxo1 gene. J Biol Chem 287: 13944 13951, 2012.
1443. Qiao JH, Tripathi J, Mishra NK, Cai Y, Tripathi S, Wang XP, Imes S, Fishbein MC,
Clinton SK, Libby P, Lusis AJ, Rajavashisth TB. Role of macrophage colony-stimulating factor in atherosclerosis: studies of osteopetrotic mice. Am J Pathol 150: 1687
1699, 1997.
1444. Qiu G, Hill JS. Endothelial lipase enhances low density lipoprotein binding and cell
association in THP-1 macrophages. Cardiovasc Res 76: 528 538, 2007.
1445. Quinn MT, Parthasarathy S, Fong LG, Steinberg D. Oxidatively modified low density
lipoproteins: a potential role in recruitment and retention of monocyte/macrophages during atherogenesis. Proc Natl Acad Sci USA 84: 29952998, 1987.
1446. Quinones MP, Martinez HG, Jimenez F, Estrada CA, Dudley M, Willmon O, Kulkarni
H, Reddick RL, Fernandes G, Kuziel WA, Ahuja SK, Ahuja SS. CC chemokine receptor 5 influences late-stage atherosclerosis. Atherosclerosis 195: e92103, 2007.
1447. Rader DJ, Alexander ET, Weibel GL, Billheimer J, Rothblat GH. The role of reverse
cholesterol transport in animals and humans and relationship to atherosclerosis. J
Lipid Res 50: S189 194, 2009.
1448. Raffai RL. Apolipoprotein E regulation of myeloid cell plasticity in atherosclerosis.
Curr Opin Lipidol 23: 471 478, 2012.
1449. Rahaman SO, Swat W, Febbraio M, Silverstein RL. Vav family Rho guanine nucleotide
exchange factors regulate CD36-mediated macrophage foam cell formation. J Biol
Chem 286: 7010 7017, 2011.
1450. Rahimi N. A role for protein ubiquitination in VEGFR-2 signalling and angiogenesis.
Biochem Soc Trans 37: 1189 1192, 2009.
1451. Raife TJ, Dwyre DM, Stevens JW, Erger RA, Leo L, Wilson KM, Fernandez JA, Wilder
J, Kim HS, Griffin JH, Maeda N, Lentz SR. Human thrombomodulin knock-in mice
reveal differential effects of human thrombomodulin on thrombosis and atherosclerosis. Arterioscler Thromb Vasc Biol 31: 2509 2517, 2011.
1452. Raizman JE, Hu TH, Chen YX, Salari S, Seibert T, Ma X, Hibbert B, Simard T, Zhao
X, Rayner K, Moore K, OBrien E. Abstract 15504: heat shock protein 27 mediated
1524
1440. Pueyo ME, Gonzalez W, Nicoletti A, Savoie F, Arnal JF, Michel JB. Angiotensin II
stimulates endothelial vascular cell adhesion molecule-1 via nuclear factor-kappaB
activation induced by intracellular oxidative stress. Arterioscler Thromb Vasc Biol 20:
645 651, 2000.
1455. Rajsheker S, Manka D, Blomkalns AL, Chatterjee TK, Stoll LL, Weintraub NL. Crosstalk between perivascular adipose tissue and blood vessels. Curr Opin Pharmacol 10:
191196, 2010.
1478. Reynaert NL, van der Vliet A, Guala AS, McGovern T, Hristova M, Pantano C, Heintz
NH, Heim J, Ho YS, Matthews DE, Wouters EF, Janssen-Heininger YM. Dynamic
redox control of NF-kappaB through glutaredoxin-regulated S-glutathionylation of
inhibitory kappaB kinase beta. Proc Natl Acad Sci USA 103: 13086 13091, 2006.
1479. Rezaee F, Casetta B, Levels JH, Speijer D, Meijers JC. Proteomic analysis of highdensity lipoprotein. Proteomics 6: 721730, 2006.
1480. Rezaee F, Gijbels M, Offerman E, Verheijen J. Genetic deletion of tissue-type plasminogen activator (t-PA) in APOE3-Leiden mice reduces progression of cholesterolinduced atherosclerosis. Thromb Haemost 90: 710 716, 2003.
1481. Rezaee F, Gijbels MJ, Offerman EH, van der Linden M, De Maat MP, Verheijen JH.
Overexpression of fibrinogen in ApoE*3-Leiden transgenic mice does not influence
the progression of diet-induced atherosclerosis. Thromb Haemost 88: 329 334,
2002.
1482. Rhen T, Cidlowski JA. Antiinflammatory action of glucocorticoids: new mechanisms
for old drugs. N Engl J Med 353: 17111723, 2005.
1483. Ricci R, Sumara G, Sumara I, Rozenberg I, Kurrer M, Akhmedov A, Hersberger M,
Eriksson U, Eberli FR, Becher B, Boren J, Chen M, Cybulsky MI, Moore KJ, Freeman
MW, Wagner EF, Matter CM, Luscher TF. Requirement of JNK2 for scavenger
receptor A-mediated foam cell formation in atherogenesis. Science 306: 1558 1561,
2004.
1484. Richards MR, Black AS, Bonnet DJ, Barish GD, Woo CW, Tabas I, Curtiss LK, Tobias
PS. The LPS2 mutation in TRIF is atheroprotective in hyperlipidemic low density
lipoprotein receptor knockout mice. Innate Immunity 2012.
1503. Romeo GR, Moulton KS, Kazlauskas A. Attenuated expression of profilin-1 confers
protection from atherosclerosis in the LDL receptor null mouse. Circ Res 101:
357367, 2007.
1485. Ridker PM, Thuren T, Zalewski A, Libby P. Interleukin-1beta inhibition and the
prevention of recurrent cardiovascular events: rationale and design of the Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS). Am Heart J
162: 597 605, 2011.
1504. Romeo S, Yin W, Kozlitina J, Pennacchio LA, Boerwinkle E, Hobbs HH, Cohen JC.
Rare loss-of-function mutations in ANGPTL family members contribute to plasma
triglyceride levels in humans. J Clin Invest 119: 70 79, 2009.
1486. Riehl A, Nemeth J, Angel P, Hess J. The receptor RAGE: Bridging inflammation and
cancer. Cell Commun Signaling 7: 12, 2009.
1505. Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein
response. Nat Rev Mol Cell Biol 8: 519 529, 2007.
1487. Riggs D, Yang Z, Kloss J, Loftus JC. The Pyk2 FERM regulates Pyk2 complex formation and phosphorylation. Cell Signal 23: 288 296, 2011.
1506. Rondaij MG, Bierings R, Kragt A, van Mourik JA, Voorberg J. Dynamics and plasticity
of Weibel-Palade bodies in endothelial cells. Arterioscler Thromb Vasc Biol 26: 1002
1007, 2006.
1507. Rong S, Cao Q, Liu M, Seo J, Jia L, Boudyguina E, Gebre AK, Colvin PL, Smith TL,
Murphy RC, Mishra N, Parks JS. Macrophage 12/15 lipoxygenase expression increases plasma and hepatic lipid levels and exacerbates atherosclerosis. J Lipid Res 53:
686 695, 2012.
1525
1477. Reynaert NL, Ckless K, Korn SH, Vos N, Guala AS, Wouters EF, van der Vliet A,
Janssen-Heininger YM. Nitric oxide represses inhibitory kappaB kinase through Snitrosylation. Proc Natl Acad Sci USA 101: 8945 8950, 2004.
1489. Riou S, Mees B, Esposito B, Merval R, Vilar J, Stengel D, Ninio E, van Haperen R, de
Crom R, Tedgui A, Lehoux S. High pressure promotes monocyte adhesion to the
vascular wall. Circ Res 100: 1226 1233, 2007.
PAUL N. HOPKINS
1508. Rosenson RS, Brewer HB Jr, Chapman MJ, Fazio S, Hussain MM, Kontush A, Krauss
RM, Otvos JD, Remaley AT, Schaefer EJ. HDL measures, particle heterogeneity,
proposed nomenclature, and relation to atherosclerotic cardiovascular events. Clin
Chem 57: 392 410, 2011.
1509. Rosenson RS, Hurt-Camejo E. Phospholipase A2 enzymes and the risk of atherosclerosis. Eur Heart J 2012.
1510. Roskoski R Jr. RAF protein-serine/threonine kinases: structure and regulation.
Biochem Biophys Res Commun 399: 313317, 2010.
1528. Sage AP, Tsiantoulas D, Baker L, Harrison J, Masters L, Murphy D, Loinard C, Binder
CJ, Mallat Z. BAFF receptor deficiency reduces the development of atherosclerosis
in mice brief report. Arterioscler Thromb Vasc Biol 32: 15731576, 2012.
1529. Saito R, Matsuzaka T, Karasawa T, Sekiya M, Okada N, Igarashi M, Matsumori R, Ishii
K, Nakagawa Y, Iwasaki H, Kobayashi K, Yatoh S, Takahashi A, Sone H, Suzuki H,
Yahagi N, Yamada N, Shimano H. Macrophage Elovl6 deficiency ameliorates foam
cell formation and reduces atherosclerosis in low-density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol 31: 19731979, 2011.
1512. Ross R, Glomset JA. The pathogenesis of atherosclerosis (first of two parts). N Engl
J Med 295: 369 377, 1976.
1513. Ross R, Glomset JA. The pathogenesis of atherosclerosis (second of two parts). N
Engl J Med 295: 420 425, 1976.
1531. Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3 regulatory T cells in the
human immune system. Nat Rev Immunol 10: 490 500, 2010.
1514. Rother E, Brandl R, Baker DL, Goyal P, Gebhard H, Tigyi G, Siess W. Subtypeselective antagonists of lysophosphatidic acid receptors inhibit platelet activation
triggered by the lipid core of atherosclerotic plaques. Circulation 108: 741747,
2003.
1532. Sakamoto H, Aikawa M, Hill CC, Weiss D, Taylor WR, Libby P, Lee RT. Biomechanical strain induces class a scavenger receptor expression in human monocyte/macrophages and THP-1 cells: a potential mechanism of increased atherosclerosis in hypertension. Circulation 104: 109 114, 2001.
1511. Ross R. Atherosclerosisan inflammatory disease. N Engl J Med 340: 115126, 1999.
1526
1535. Sanchez-Quesada JL, Benitez S, Ordonez-Llanos J. Electronegative low-density lipoprotein. Curr Opin Lipidol 15: 329 335, 2004.
1536. Sangle GV, Zhao R, Shen GX. Transmembrane signaling pathway mediates oxidized low-density lipoprotein-induced expression of plasminogen activator inhibitor-1 in vascular endothelial cells. Am J Physiol Endocrinol Metab 295: E1243
E1254, 2008.
1537. Sari FR, Watanabe K, Widyantoro B, Thandavarayan RA, Harima M, Zhang S, Muslin
AJ, Kodama M, Aizawa Y. Partial inactivation of cardiac 14-3-3 protein in vivo elicits
endoplasmic reticulum stress (ERS) and activates ERS-initiated apoptosis in ERSinduced mice. Cell Physiol Biochem 26: 167178, 2010.
1538. Sarkar D, Fisher PB. Molecular mechanisms of aging-associated inflammation. Cancer
Lett 236: 1323, 2006.
1539. Sasaki T, Kuzuya M, Nakamura K, Cheng XW, Shibata T, Sato K, Iguchi A. A simple
method of plaque rupture induction in apolipoprotein E-deficient mice. Arterioscler
Thromb Vasc Biol 26: 1304 1309, 2006.
1540. Sato H, Kato R, Isogai Y, Saka G, Ohtsuki M, Taketomi Y, Yamamoto K, Tsutsumi K,
Yamada J, Masuda S, Ishikawa Y, Ishii T, Kobayashi T, Ikeda K, Taguchi R, Hatakeyama S, Hara S, Kudo I, Itabe H, Murakami M. Analyses of group III secreted
phospholipase A2 transgenic mice reveal potential participation of this enzyme in
plasma lipoprotein modification, macrophage foam cell formation, and atherosclerosis. J Biol Chem 283: 3348333497, 2008.
1541. Sato K, Okajima F. Role of sphingosine 1-phosphate in anti-atherogenic actions of
high-density lipoprotein. World J Biol Chem 1: 327337, 2010.
1542. Satonaka H, Suzuki E, Nishimatsu H, Oba S, Takeda R, Goto A, Omata M, Fujita T,
Nagai R, Hirata Y. Calcineurin promotes the expression of monocyte chemoattractant protein-1 in vascular myocytes and mediates vascular inflammation. Circ Res 94:
693700, 2004.
1543. Sattler KJ, Elbasan S, Keul P, Elter-Schulz M, Bode C, Graler MH, Brocker-Preuss M,
Budde T, Erbel R, Heusch G, Levkau B. Sphingosine 1-phosphate levels in plasma and
HDL are altered in coronary artery disease. Basic Res Cardiol 105: 821 832, 2010.
1544. Sawada Y, Tamada M, Dubin-Thaler BJ, Cherniavskaya O, Sakai R, Tanaka S, Sheetz
MP. Force sensing by mechanical extension of the Src family kinase substrate
p130Cas. Cell 127: 10151026, 2006.
1545. Sawamura T, Kume N, Aoyama T, Moriwaki H, Hoshikawa H, Aiba Y, Tanaka T,
Miwa S, Katsura Y, Kita T, Masaki T. An endothelial receptor for oxidized lowdensity lipoprotein. Nature 386: 7377, 1997.
1534. Sanan DA, Newland DL, Tao R, Marcovina S, Wang J, Mooser V, Hammer RE, Hobbs
HH. Low density lipoprotein receptor-negative mice expressing human apolipoprotein B-100 develop complex atherosclerotic lesions on a chow diet: no accentuation
by apolipoprotein(a). Proc Natl Acad Sci USA 95: 4544 4549, 1998.
1566. Schroder K, Benkhoff S, Mieth A, Zhang M, Shah AM, Brandes RP. Abstract 13713:
The H2O2-producing NADPH oxidase Nox4 protects against angiotensin II-Induced
vascular hypertrophy and inflammation. Circulation 122: A13713, 2010.
1567. Schroder K, Hertzog PJ, Ravasi T, Hume DA. Interferon-gamma: an overview of
signals, mechanisms and functions. J Leukoc Biol 75: 163189, 2004.
1568. Schrder K, Vecchione C, Jung O, Schreiber JG, Shiri-Sverdlov R, van Gorp PJ, Busse
R, Brandes RP. Xanthine oxidase inhibitor tungsten prevents the development of
atherosclerosis in ApoE knockout mice fed a Western-type diet. Free Radical Biol
Med 41: 13531360, 2006.
1569. Schuett H, Oestreich R, Waetzig GH, Annema W, Luchtefeld M, Hillmer A, Bavendiek U, von Felden J, Divchev D, Kempf T, Wollert KC, Seegert D, Rose-John S,
Tietge UJ, Schieffer B, Grote K. Transsignaling of interleukin-6 crucially contributes
to atherosclerosis in mice. Arterioscler Thromb Vasc Biol 32: 281290, 2012.
1570. Schultz JR, Verstuyft JG, Gong EL, Nichols AV, Rubin EM. Protein composition
determines the anti-atherogenic properties of HDL in transgenic mice. Nature 365:
762764, 1993.
1571. Schulze PC, Liu H, Choe E, Yoshioka J, Shalev A, Bloch KD, Lee RT. Nitric oxidedependent suppression of thioredoxin-interacting protein expression enhances thioredoxin activity. Arterioscler Thromb Vasc Biol 26: 2666 2672, 2006.
1572. Schulze PC, Yoshioka J, Takahashi T, He Z, King GL, Lee RT. Hyperglycemia promotes oxidative stress through inhibition of thioredoxin function by thioredoxininteracting protein. J Biol Chem 279: 30369 30374, 2004.
1573. Schurks M, Glynn RJ, Rist PM, Tzourio C, Kurth T. Effects of vitamin E on stroke
subtypes: meta-analysis of randomised controlled trials. BMJ 341: c5702, 2010.
1574. Schwartz EA, Zhang WY, Karnik SK, Borwege S, Anand VR, Laine PS, Su Y, Reaven
PD. Nutrient modification of the innate immune response: a novel mechanism by
which saturated fatty acids greatly amplify monocyte inflammation. Arterioscler
Thromb Vasc Biol 30: 802 808, 2010.
1575. Schwartz GG, Olsson AG, Abt M, Ballantyne CM, Barter PJ, Brumm J, Chaitman BR,
Holme IM, Kallend D, Leiter LA, Leitersdorf E, McMurray JJ, Mundl H, Nicholls SJ,
Shah PK, Tardif JC, Wright RS. Effects of dalcetrapib in patients with a recent acute
coronary syndrome. N Engl J Med 367: 2089 2099, 2012.
1576. Schwartz SM, Galis ZS, Rosenfeld ME, Falk E. Plaque rupture in humans and mice.
Arterioscler Thromb Vasc Biol 27: 705713, 2007.
1577. Schwenke DC, Carew TE. Initiation of atherosclerotic lesions in cholesterol-fed
rabbits. I. Focal increases in arterial LDL concentration precede development of
fatty streak lesions. Arteriosclerosis 9: 895907, 1989.
1578. Schwenke DC, Carew TE. Initiation of atherosclerotic lesions in cholesterol-fed
rabbits. II. Selective retention of LDL vs selective increases in LDL permeability in
susceptible sites of arteries. Arteriosclerosis 9: 908 918, 1989.
1579. Scotta C, Soligo M, Camperio C, Piccolella E. FOXP3 induced by CD28/B7 interaction regulates CD25 and anergic phenotype in human CD4CD25- T lymphocytes.
J Immunol 181: 10251033, 2008.
1580. Seals DR, Desouza CA, Donato AJ, Tanaka H. Habitual exercise and arterial aging. J
Appl Physiol 105: 13231332, 2008.
1563. Schoneveld AH, Hoefer I, Sluijter JP, Laman JD, de Kleijn DP, Pasterkamp G. Atherosclerotic lesion development and Toll like receptor 2 and 4 responsiveness.
Atherosclerosis 197: 95104, 2008.
1582. Seimon TA, Kim MJ, Blumenthal A, Koo J, Ehrt S, Wainwright H, Bekker LG, Kaplan
G, Nathan C, Tabas I, Russell DG. Induction of ER stress in macrophages of tuberculosis granulomas. PLoS ONE 5: e12772, 2010.
1564. Schoneveld AH, Oude Nijhuis MM, van Middelaar B, Laman JD, de Kleijn DP, Pasterkamp G. Toll-like receptor 2 stimulation induces intimal hyperplasia and atherosclerotic lesion development. Cardiovasc Res 66: 162169, 2005.
1583. Seimon TA, Wang Y, Han S, Senokuchi T, Schrijvers DM, Kuriakose G, Tall AR, Tabas
IA. Macrophage deficiency of p38 MAPK promotes apoptosis and plaque necrosis
in advanced atherosclerotic lesions in mice. J Clin Invest 119: 886 898, 2009.
1527
1565. Schreyer SA, Vick CM, LeBoeuf RC. Loss of lymphotoxin-alpha but not tumor
necrosis factor-alpha reduces atherosclerosis in mice. J Biol Chem 277: 12364
12368, 2002.
PAUL N. HOPKINS
1584. Sekiya M, Osuga J, Nagashima S, Ohshiro T, Igarashi M, Okazaki H, Takahashi M, Tazoe
F, Wada T, Ohta K, Takanashi M, Kumagai M, Nishi M, Takase S, Yahagi N, Yagyu H,
Ohashi K, Nagai R, Kadowaki T, Furukawa Y, Ishibashi S. Ablation of neutral cholesterol
ester hydrolase 1 accelerates atherosclerosis. Cell Metab 10: 219 228, 2009.
1585. Semple K, Nguyen A, Yu Y, Wang H, Anasetti C, Yu XZ. Strong CD28 costimulation
suppresses induction of regulatory T cells from naive precursors through Lck signaling. Blood 117: 3096 3103, 2011.
1586. Sen U, Givvimani S, Abe OA, Lederer ED, Tyagi SC. Cystathionine beta-synthase and
cystathionine gamma-lyase double gene transfer ameliorate homocysteine-mediated mesangial inflammation through hydrogen sulfide generation. Am J Physiol Cell
Physiol 300: C155C163, 2011.
1587. Sen U, Tyagi N, Patibandla PK, Dean WL, Tyagi SC, Roberts AM, Lominadze D.
Fibrinogen-induced endothelin-1 production from endothelial cells. Am J Physiol Cell
Physiol 296: C840 C847, 2009.
1588. Sentman ML, Brannstrom T, Westerlund S, Laukkanen MO, Yla-Herttuala S, Basu S,
Marklund SL. Extracellular superoxide dismutase deficiency and atherosclerosis in
mice. Arterioscler Thromb Vasc Biol 21: 14771482, 2001.
1589. Serezani CH, Lewis C, Jancar S, Peters-Golden M. Leukotriene B4 amplifies NFkappaB activation in mouse macrophages by reducing SOCS1 inhibition of MyD88
expression. J Clin Invest 121: 671 682, 2011.
1590. Serhan CN. Novel lipid mediators and resolution mechanisms in acute inflammation:
to resolve or not? Am J Pathol 177: 1576 1591, 2010.
1592. Serhan CN, Petasis NA. Resolvins and protectins in inflammation resolution. Chem
Rev 111: 59225943, 2011.
1593. Serhan CN, Yacoubian S, Yang R. Anti-inflammatory and proresolving lipid mediators. Annu Rev Pathol 3: 279 312, 2008.
1594. Serruys PW, Garcia-Garcia HM, Buszman P, Erne P, Verheye S, Aschermann M, Duckers H, Bleie O, Dudek D, Botker HE, von Birgelen C, DAmico D, Hutchinson T,
Zambanini A, Mastik F, van Es GA, van der Steen AF, Vince DG, Ganz P, Hamm CW,
Wijns W, Zalewski A. Effects of the direct lipoprotein-associated phospholipase A(2)
inhibitor darapladib on human coronary atherosclerotic plaque. Circulation 118: 1172
1182, 2008.
1595. Sesso HD, Buring JE, Christen WG, Kurth T, Belanger C, MacFadyen J, Bubes V,
Manson JE, Glynn RJ, Gaziano JM. Vitamins E and C in the prevention of cardiovascular disease in men: the Physicians Health Study II randomized controlled trial.
JAMA 300: 21232133, 2008.
1596. Seye CI, Yu N, Jain R, Kong Q, Minor T, Newton J, Erb L, Gonzalez FA, Weisman GA.
The P2Y2 nucleotide receptor mediates UTP-induced vascular cell adhesion molecule-1 expression in coronary artery endothelial cells. J Biol Chem 278: 24960
24965, 2003.
1597. Shagdarsuren E, Djalali-Talab Y, Aurrand-Lions M, Bidzhekov K, Liehn EA, Imhof BA,
Weber C, Zernecke A. Importance of junctional adhesion molecule-C for neointimal
hyperplasia and monocyte recruitment in atherosclerosis-prone mice-brief report.
Arterioscler Thromb Vasc Biol 29: 11611163, 2009.
1598. Shai SY, Sukhanov S, Higashi Y, Vaughn C, Kelly J, Delafontaine P. Smooth muscle
cell-specific insulin-like growth factor-1 overexpression in Apoe/ mice does not
alter atherosclerotic plaque burden but increases features of plaque stability. Arterioscler Thromb Vasc Biol 30: 1916 1924, 2010.
1599. Shai SY, Sukhanov S, Higashi Y, Vaughn C, Rosen CJ, Delafontaine P. Low circulating
insulin-like growth factor I increases atherosclerosis in ApoE-deficient mice. Am J
Physiol Heart Circ Physiol 300: H1898 H1906, 2011.
1600. Shaik SS, Soltau TD, Chaturvedi G, Totapally B, Hagood JS, Andrews WW, Athar
M, Voitenok NN, Killingsworth CR, Patel RP, Fallon MB, Maheshwari A. Low
intensity shear stress increases endothelial ELR CXC chemokine production
via a focal adhesion kinase-p38 MAPK-NF-B pathway. J Biol Chem 284: 5945
5955, 2009.
1601. Shalhoub J, Falck-Hansen MA, Davies AH, Monaco C. Innate immunity and monocyte-macrophage activation in atherosclerosis. J Inflamm 8: 9, 2011.
1528
1603. Sharma J, Turk J, McHowat J. Endothelial cell prostaglandin I(2) and platelet-activating factor production are markedly attenuated in the calcium-independent phospholipase A(2)beta knockout mouse. Biochemistry 49: 54735481, 2010.
1604. Sharma M. Chemokines and their receptors: orchestrating a fine balance between
health and disease. Crit Rev Biotechnol 30: 122, 2010.
1605. Shattil SJ, Kim C, Ginsberg MH. The final steps of integrin activation: the end game.
Nat Rev Mol Cell Biol 11: 288 300, 2010.
1606. Shay KP, Moreau RF, Smith EJ, Smith AR, Hagen TM. Alpha-lipoic acid as a dietary
supplement: molecular mechanisms and therapeutic potential. Biochim Biophys Acta
1790: 1149 1160, 2009.
1607. She ZG, Zheng W, Wei YS, Chen HZ, Wang AB, Li HL, Liu G, Zhang R, Liu JJ, Stallcup
WB, Zhou Z, Liu DP, Liang CC. Human paraoxonase gene cluster transgenic overexpression represses atherogenesis and promotes atherosclerotic plaque stability in
ApoE-null mice. Circ Res 104: 1160 1168, 2009.
1608. Shechtman CF, Henneberry AL, Seimon TA, Tinkelenberg AH, Wilcox LJ, Lee E,
Fazlollahi M, Munkacsi AB, Bussemaker HJ, Tabas I, Sturley SL. Loss of subcellular
lipid transport due to ARV1 deficiency disrupts organelle homeostasis and activates
the unfolded protein response. J Biol Chem 286: 1195111959, 2011.
1609. Sheehan AL, Carrell S, Johnson B, Stanic B, Banfi B, Miller FJ Jr. Role for Nox1
NADPH oxidase in atherosclerosis. Atherosclerosis 216: 321326, 2011.
1610. Shemesh S, Kamari Y, Shaish A, Olteanu S, Kandel-Kfir M, Almog T, Grosskopf I,
Apte RN, Harats D. Interleukin-1 receptor type-1 in non-hematopoietic cells is the
target for the pro-atherogenic effects of interleukin-1 in apoE-deficient mice. Atherosclerosis 222: 329 336, 2012.
1611. Shen H, Bielak LF, Ferguson JF, Streeten EA, Yerges-Armstrong LM, Liu J, Post W,
OConnell JR, Hixson JE, Kardia SL, Sun YV, Jhun MA, Wang X, Mehta NN, Li M,
Koller DL, Hakonarson H, Keating BJ, Rader DJ, Shuldiner AR, Peyser PA, Reilly MP,
Mitchell BD. Association of the vitamin D metabolism gene CYP24A1 with coronary
artery calcification. Arterioscler Thromb Vasc Biol 30: 2648 2654, 2010.
1612. Shen J, Chandrasekharan UM, Ashraf MZ, Long E, Morton RE, Liu Y, Smith JD,
DiCorleto PE. Lack of mitogen-activated protein kinase phosphatase-1 protects
apoE-null mice against atherosclerosis. Circ Res 106: 902910, 2010.
1613. Shen J, Herderick E, Cornhill JF, Zsigmond E, Kim HS, Kuhn H, Guevara NV, Chan L.
Macrophage-mediated 15-lipoxygenase expression protects against atherosclerosis
development. J Clin Invest 98: 22012208, 1996.
1614. Shen J, Yang M, Ju D, Jiang H, Zheng JP, Xu Z, Li L. Disruption of SM22 promotes
inflammation after artery injury via nuclear factor kappaB activation. Circ Res 106:
13511362, 2010.
1615. Shenouda SM, Widlansky ME, Chen K, Xu G, Holbrook M, Tabit CE, Hamburg NM,
Frame AA, Caiano TL, Kluge MA, Duess MA, Levit A, Kim B, Hartman ML, Joseph L,
Shirihai OS, Vita JA. Altered mitochondrial dynamics contributes to endothelial dysfunction in diabetes mellitus. Circulation 124: 444 453, 2011.
1616. Shi R, Re D, Dudl E, Cuddy M, Okolotowicz KJ, Dahl R, Su Y, Hurder A, Kitada S,
Peddibhotla S, Roth GP, Smith LH, Kipps TJ, Cosford N, Cashman J, Reed JC.
Chemical biology strategy reveals pathway-selective inhibitor of NF-kappaB activation induced by protein kinase C. ACS Chem Biol 5: 287299, 2010.
1617. Shi W, Wang NJ, Shih DM, Sun VZ, Wang X, Lusis AJ. Determinants of atherosclerosis susceptibility in the C3H and C57BL/6 mouse model: evidence for involvement
of endothelial cells but not blood cells or cholesterol metabolism. Circ Res 86: 1078
1084, 2000.
1618. Shibata N, Glass CK. Regulation of macrophage function in inflammation and atherosclerosis. J Lipid Res 50: S277281, 2009.
1619. Shiflett AM, Bishop JR, Pahwa A, Hajduk SL. Human high density lipoproteins are
platforms for the assembly of multi-component innate immune complexes. J Biol
Chem 280: 32578 32585, 2005.
1620. Shih DM, Lusis AJ. The roles of PON1 and PON2 in cardiovascular disease and innate
immunity. Curr Opin Lipidol 20: 288 292, 2009.
1591. Serhan CN, Chiang N, Van Dyke TE. Resolving inflammation: dual anti-inflammatory
and pro-resolution lipid mediators. Nat Rev Immunol 8: 349 361, 2008.
1602. Shaposhnik Z, Wang X, Lusis AJ. Arterial colony stimulating factor-1 influences
atherosclerotic lesions by regulating monocyte migration and apoptosis. J Lipid Res
51: 19621970, 2010.
1628. Shpilberg O, Rabi I, Schiller K, Walden R, Harats D, Tyrrell KS, Coller B, Seligsohn U.
Patients with Glanzmann thrombasthenia lacking platelet glycoprotein alpha(IIb)beta(3) (GPIIb/IIIa) and alpha(v)beta(3) receptors are not protected from atherosclerosis. Circulation 105: 1044 1048, 2002.
1629. Shu L, Shayman JA. Caveolin-associated accumulation of globotriaosylceramide in
the vascular endothelium of alpha-galactosidase A null mice. J Biol Chem 282: 20960
20967, 2007.
1630. Siddle K. Molecular basis of signaling specificity of insulin and IGF receptors: neglected corners and recent advances. Front Endocrinol 3: 34, 2012.
1631. Siess W, Zangl KJ, Essler M, Bauer M, Brandl R, Corrinth C, Bittman R, Tigyi G,
Aepfelbacher M. Lysophosphatidic acid mediates the rapid activation of platelets and
endothelial cells by mildly oxidized low density lipoprotein and accumulates in human atherosclerotic lesions. Proc Natl Acad Sci USA 96: 6931 6936, 1999.
1640. Skoura A, Michaud J, Im DS, Thangada S, Xiong Y, Smith JD, Hla T. Sphingosine-1phosphate receptor-2 function in myeloid cells regulates vascular inflammation and
atherosclerosis. Arterioscler Thromb Vasc Biol 31: 81 85, 2011.
1641. Slager CJ, Wentzel JJ, Gijsen FJ, Schuurbiers JC, van der Wal AC, van der Steen AF,
Serruys PW. The role of shear stress in the generation of rupture-prone vulnerable
plaques. Nat Clin Pract Cardiovasc Med 2: 401 407, 2005.
1642. Slager CJ, Wentzel JJ, Gijsen FJ, Thury A, van der Wal AC, Schaar JA, Serruys PW. The
role of shear stress in the destabilization of vulnerable plaques and related therapeutic implications. Nat Clin Pract Cardiovasc Med 2: 456 464, 2005.
1643. Sleiman L, Hasu M, Thabet M, Thorn S, Wells G, deKemp R, Holcik M, Beanlands RS,
Whitman S. Abstract 17207: the role of cIAP2 in atherosclerotic lesion development.
Circulation 122: A17207, 2010.
1644. Sluimer JC, Gasc JM, van Wanroij JL, Kisters N, Groeneweg M, Sollewijn Gelpke MD,
Cleutjens JP, van den Akker LH, Corvol P, Wouters BG, Daemen MJ, Bijnens AP.
Hypoxia, hypoxia-inducible transcription factor, and macrophages in human atherosclerotic plaques are correlated with intraplaque angiogenesis. J Am Coll Cardiol 51:
1258 1265, 2008.
1645. Small DM. George Lyman Duff memorial lecture. Progression and regression of
atherosclerotic lesions Insights from lipid physical biochemistry. Arteriosclerosis 8:
103129, 1988.
1646. Smedlund K, Tano JY, Vazquez G. The constitutive function of native TRPC3 channels modulates vascular cell adhesion molecule-1 expression in coronary endothelial
cells through nuclear factor kappaB signaling. Circ Res 106: 1479 1488, 2010.
1647. Smith EB. Transport, interactions and retention of plasma proteins in the intima: the
barrier function of the internal elastic lamina. Eur Heart J 11 Suppl E: 72 81, 1990.
1648. Smith JD, Trogan E, Ginsberg M, Grigaux C, Tian J, Miyata M. Decreased atherosclerosis in mice deficient in both macrophage colony-stimulating factor (op) and
apolipoprotein E. Proc Natl Acad Sci USA 92: 8264 8268, 1995.
1649. Soehnlein O. Multiple roles for neutrophils in atherosclerosis. Circ Res 110: 875
888, 2012.
1650. Soehnlein O, Wantha S, Simsekyilmaz S, Doring Y, Megens RT, Mause SF, Drechsler
M, Smeets R, Weinandy S, Schreiber F, Gries T, Jockenhoevel S, Moller M, Vijayan S,
van Zandvoort MA, Agerberth B, Pham CT, Gallo RL, Hackeng TM, Liehn EA,
Zernecke A, Klee D, Weber C. Neutrophil-derived cathelicidin protects from neointimal hyperplasia. Sci translational Med 3: 103ra198, 2011.
1632. Silence J, Collen D, Lijnen HR. Reduced atherosclerotic plaque but enhanced aneurysm formation in mice with inactivation of the tissue inhibitor of metalloproteinase-1 (TIMP-1) gene. Circ Res 90: 897903, 2002.
1651. Solomon SD, Wittes J, Finn PV, Fowler R, Viner J, Bertagnolli MM, Arber N, Levin B,
Meinert CL, Martin B, Pater JL, Goss PE, Lance P, Obara S, Chew EY, Kim J, Arndt
G, Hawk E. Cardiovascular risk of celecoxib in 6 randomized placebo-controlled
trials: the cross trial safety analysis. Circulation 117: 2104 2113, 2008.
1633. Silence J, Lupu F, Collen D, Lijnen HR. Persistence of atherosclerotic plaque but
reduced aneurysm formation in mice with stromelysin-1 (MMP-3) gene inactivation.
Arterioscler Thromb Vasc Biol 21: 1440 1445, 2001.
1652. Sommerville LJ, Kelemen SE, Ellison SP, England RN, Autieri MV. Increased atherosclerosis and vascular smooth muscle cell activation in AIF-1 transgenic mice fed a
high-fat diet. Atherosclerosis 220: 4552, 2012.
1635. Singh H, Milner CS, Aguilar Hernandez MM, Patel N, Brindle NP. Vascular endothelial growth factor activates the Tie family of receptor tyrosine kinases. Cell Signal 21:
1346 1350, 2009.
1636. Singh SP, Niemczyk M, Saini D, Sadovov V, Zimniak L, Zimniak P. Disruption of the
mGsta4 gene increases life span of C57BL mice. J Gerontol A Biol Sci Med Sci 65:
14 23, 2010.
1637. Sjoland H, Eitzman DT, Gordon D, Westrick R, Nabel EG, Ginsburg D. Atherosclerosis progression in LDL receptor-deficient and apolipoprotein E-deficient mice is
independent of genetic alterations in plasminogen activator inhibitor-1. Arterioscler
Thromb Vasc Biol 20: 846 852, 2000.
1638. Skalen K, Gustafsson M, Rydberg EK, Hulten LM, Wiklund O, Innerarity TL, Boren J.
Subendothelial retention of atherogenic lipoproteins in early atherosclerosis. Nature
417: 750 754, 2002.
1529
1639. Skaug B, Jiang X, Chen ZJ. The role of ubiquitin in NF-kappaB regulatory pathways.
Annu Rev Biochem 78: 769 796, 2009.
PAUL N. HOPKINS
1659. Soriano FG, Pacher P, Mabley J, Liaudet L, Szabo C. Rapid reversal of the diabetic
endothelial dysfunction by pharmacological inhibition of poly(ADP-ribose) polymerase. Circ Res 89: 684 691, 2001.
1661. Sourris KC, Harcourt BE, Penfold SA, Yap FY, Morley AL, Morgan PE, Davies MJ,
Baker ST, Jerums G, Forbes JM. Modulation of the cellular expression of circulating
advanced glycation end-product receptors in type 2 diabetic nephropathy. Exp Diabetes Res 2010: 974681, 2010.
1662. Spann NJ, Garmire LX, McDonald JG, Myers DS, Milne SB, Shibata N, Reichart D,
Fox JN, Shaked I, Heudobler D, Raetz CR, Wang EW, Kelly SL, Sullards MC, Murphy
RC, Merrill AH Jr, Brown HA, Dennis EA, Li AC, Ley K, Tsimikas S, Fahy E, Subramaniam S, Quehenberger O, Russell DW, Glass CK. Regulated accumulation of
desmosterol integrates macrophage lipid metabolism and inflammatory responses.
Cell 151: 138 152, 2012.
1663. Speidl WS, Kastl SP, Huber K, Wojta J. Complement in atherosclerosis: friend or foe?
J Thromb Haemost 9: 428 440, 2011.
1664. Speidl WS, Kastl SP, Hutter R, Katsaros KM, Kaun C, Bauriedel G, Maurer G, Huber
K, Badimon JJ, Wojta J. The complement component C5a is present in human
coronary lesions in vivo and induces the expression of MMP-1 and MMP-9 in human
macrophages in vitro. FASEB J 25: 35 44, 2011.
1666. Spite M, Serhan CN. Novel lipid mediators promote resolution of acute inflammation: impact of aspirin and statins. Circ Res 107: 1170 1184, 2010.
1667. Sramek A, Kriek M, Rosendaal FR. Decreased mortality of ischaemic heart disease
among carriers of haemophilia. Lancet 362: 351354, 2003.
1668. Sramek A, Reiber JH, Gerrits WB, Rosendaal FR. Decreased coagulability has no
clinically relevant effect on atherogenesis: observations in individuals with a hereditary bleeding tendency. Circulation 104: 762767, 2001.
1669. Sriburi R, Jackowski S, Mori K, Brewer JW. XBP1: a link between the unfolded
protein response, lipid biosynthesis, and biogenesis of the endoplasmic reticulum. J
Cell Biol 167: 35 41, 2004.
1670. Sridharan M, Adderley SP, Bowles EA, Egan TM, Stephenson AH, Ellsworth ML,
Sprague RS. Pannexin 1 is the conduit for low oxygen tension-induced ATP release
from human erythrocytes. Am J Physiol Heart Circ Physiol 299: H1146 H1152, 2010.
1671. Srivastava S, Vladykovskaya E, Barski OA, Spite M, Kaiserova K, Petrash JM, Chung
SS, Hunt G, Dawn B, Bhatnagar A. Aldose reductase protects against early atherosclerotic lesion formation in apolipoprotein E-null mice. Circ Res 105: 793 802,
2009.
1672. St-Onge M, Dumas A, Michaud A, Laflamme C, Dussault AA, Pouliot M. Impact of
anti-inflammatory agents on the gene expression profile of stimulated human neutrophils: unraveling endogenous resolution pathways. PLoS ONE 4: e4902, 2009.
1673. St-Pierre J, Lysechko TL, Ostergaard HL. Hypophosphorylated and inactive Pyk2
associates with paxillin at the microtubule organizing center in hematopoietic cells.
Cell Signal 23: 718 730, 2011.
1692. Su X, Ao L, Shi Y, Johnson TR, Fullerton DA, Meng X. Oxidized low density lipoprotein induces bone morphogenetic protein-2 in coronary artery endothelial cells
via Toll-like receptors 2 and 4. J Biol Chem 286: 1221312220, 2011.
1674. Staal SP, Hartley JW, Rowe WP. Isolation of transforming murine leukemia viruses
from mice with a high incidence of spontaneous lymphoma. Proc Natl Acad Sci USA
74: 30653067, 1977.
1693. Subbarao K, Jala VR, Mathis S, Suttles J, Zacharias W, Ahamed J, Ali H, Tseng MT,
Haribabu B. Role of leukotriene B4 receptors in the development of atherosclerosis:
potential mechanisms. Arterioscler Thromb Vasc Biol 24: 369 375, 2004.
1675. Stachon P, Missiou A, Walter C, Varo N, Colberg C, Wolf D, Buchner M, von Zur
Muhlen C, Zirlik K, Bode C, Zirlik A. Tumor necrosis factor receptor associated
factor 6 is not required for atherogenesis in mice and does not associate with
atherosclerosis in humans. PLoS ONE 5: e11589, 2010.
1676. Stanic AK, Stein CM, Morgan AC, Fazio S, Linton MF, Wakeland EK, Olsen NJ, Major
AS. Immune dysregulation accelerates atherosclerosis and modulates plaque composition in systemic lupus erythematosus. Proc Natl Acad Sci USA 103: 7018 7023,
2006.
1530
1665. Spiegel S, Milstien S. The outs and the ins of sphingosine-1-phosphate in immunity.
Nat Rev Immunol 11: 403 415, 2011.
1679. Stein S, Lohmann C, Handschin C, Stenfeldt E, Boren J, Luscher TF, Matter CM.
ApoE/ PGC-1alpha/ mice display reduced IL-18 levels and do not develop
enhanced atherosclerosis. PLoS ONE 5: e13539, 2010.
1704. Sumpter R Jr, Levine B. Autophagy and innate immunity: triggering, targeting and
tuning. Semin Cell Dev Biol 21: 699 711, 2010.
1705. Sun C, Wu MH, Lee ES, Yuan SY. A Disintegrin and Metalloproteinase 15 contributes
to atherosclerosis by mediating endothelial barrier dysfunction via Src family kinase
activity. Arterioscler Thromb Vasc Biol 32: 2444 2451, 2012.
1706. Sun C, Wu MH, Yuan SY. Nonmuscle myosin light-chain kinase deficiency attenuates
atherosclerosis in apolipoprotein E-deficient mice via reduced endothelial barrier
dysfunction and monocyte migration. Circulation 124: 48 57, 2011.
1707. Sun J, Hartvigsen K, Chou MY, Zhang Y, Sukhova GK, Zhang J, Lopez-Ilasaca M,
Diehl CJ, Yakov N, Harats D, George J, Witztum JL, Libby P, Ploegh H, Shi GP.
Deficiency of antigen-presenting cell invariant chain reduces atherosclerosis in mice.
Circulation 122: 808 820, 2010.
1708. Sun J, Lin X. Beta-arrestin 2 is required for lysophosphatidic acid-induced NF-kappaB activation. Proc Natl Acad Sci USA 105: 1708517090, 2008.
1709. Sun J, Sukhova GK, Wolters PJ, Yang M, Kitamoto S, Libby P, MacFarlane LA, Mallen-St Clair J, Shi GP. Mast cells promote atherosclerosis by releasing proinflammatory cytokines. Nat Med 13: 719 724, 2007.
1710. Sun J, Zhang J, Lindholt JS, Sukhova GK, Liu J, He A, Abrink M, Pejler G, Stevens RL,
Thompson RW, Ennis TL, Gurish MF, Libby P, Shi GP. Critical role of mast cell
chymase in mouse abdominal aortic aneurysm formation. Circulation 120: 973982,
2009.
1730. Tabas I, Marathe S, Keesler GA, Beatini N, Shiratori Y. Evidence that the initial
up-regulation of phosphatidylcholine biosynthesis in free cholesterol-loaded macrophages is an adaptive response that prevents cholesterol-induced cellular necrosis.
Proposed role of an eventual failure of this response in foam cell necrosis in advanced
atherosclerosis. J Biol Chem 271: 2277322781, 1996.
1712. Sun L, Xiao L, Nie J, Liu FY, Ling GH, Zhu XJ, Tang WB, Chen WC, Xia YC, Zhan M,
Ma MM, Peng YM, Liu H, Liu YH, Kanwar YS. p66Shc mediates high-glucose and
angiotensin II-induced oxidative stress renal tubular injury via mitochondrial-dependent apoptotic pathway. Am J Physiol Renal Physiol 299: F1014 F1025, 2010.
1713. Sun N, Wood NB, Hughes AD, Thom SAM, Yun Xu X. Effects of transmural pressure and wall shear stress on LDL accumulation in the arterial wall: a numerical study
using a multilayered model. Am J Physiol Heart Circ Physiol 292: H3148 H3157, 2007.
1714. Sun W, Ge N, Yu Y, Burlingame S, Li X, Zhang M, Ye S, Fu S, Yang J. Phosphorylation
of Thr-516 and Ser-520 in the kinase activation loop of MEKK3 is required for
1729. Tabas I. The role of endoplasmic reticulum stress in the progression of atherosclerosis. Circ Res 107: 839 850, 2010.
1731. Tabas I, Tall A, Accili D. The impact of macrophage insulin resistance on advanced
atherosclerotic plaque progression. Circ Res 106: 58 67, 2010.
1732. Tabas I, Williams KJ, Boren J. Subendothelial lipoprotein retention as the initiating
process in atherosclerosis: update and therapeutic implications. Circulation 116:
18321844, 2007.
1733. Tabata M, Kadomatsu T, Fukuhara S, Miyata K, Ito Y, Endo M, Urano T, Zhu HJ,
Tsukano H, Tazume H, Kaikita K, Miyashita K, Iwawaki T, Shimabukuro M, Sakaguchi K, Ito T, Nakagata N, Yamada T, Katagiri H, Kasuga M, Ando Y, Ogawa H,
1531
1703. Sullivan PM, Mezdour H, Quarfordt SH, Maeda N. Type III hyperlipoproteinemia
and spontaneous atherosclerosis in mice resulting from gene replacement of mouse
Apoe with human Apoe*2. J Clin Invest 102: 130 135, 1998.
lysophosphatidic acid-mediated optimal IkappaB kinase beta (IKKbeta)/nuclear factor-kappaB (NF-kappaB) activation. J Biol Chem 285: 79117918, 2010.
PAUL N. HOPKINS
Mochizuki N, Itoh H, Suda T, Oike Y. Angiopoietin-like protein 2 promotes chronic
adipose tissue inflammation and obesity-related systemic insulin resistance. Cell
Metab 10: 178 188, 2009.
1751. Tan MH, Sun Z, Opitz SL, Schmidt TE, Peters JH, George EL. Deletion of the
alternatively spliced fibronectin EIIIA domain in mice reduces atherosclerosis. Blood
104: 1118, 2004.
1734. Tacken PJ, Delsing DJ, Gijbels MJ, Quax PH, Havekes LM, Hofker MH, van Dijk KW.
VLDL receptor deficiency enhances intimal thickening after vascular injury but does
not affect atherosclerotic lesion area. Atherosclerosis 162: 103110, 2002.
1735. Tafesse FG, Ternes P, Holthuis JC. The multigenic sphingomyelin synthase family. J
Biol Chem 281: 2942129425, 2006.
1736. Tai LK, Okuda M, Abe J, Yan C, Berk BC. Fluid shear stress activates proline-rich
tyrosine kinase via reactive oxygen species-dependent pathway. Arterioscler Thromb
Vasc Biol 22: 1790 1796, 2002.
1737. Tai LK, Zheng Q, Pan S, Jin ZG, Berk BC. Flow activates ERK1/2 and endothelial
nitric oxide synthase via a pathway involving PECAM1, SHP2, and Tie2. J Biol Chem
280: 29620 29624, 2005.
1738. Takabe W, Alberts-Grill N, Jo H. Disturbed flow: p53 SUMOylation in the turnover
of endothelial cells. J Cell Biol 193: 805 807, 2011.
1739. Takabe W, Li R, Ai L, Yu F, Berliner JA, Hsiai TK. Oxidized low-density lipoproteinactivated c-Jun NH2-terminal kinase regulates manganese superoxide dismutase
ubiquitination: implication for mitochondrial redox status and apoptosis. Arterioscler
Thromb Vasc Biol 30: 436 441, 2010.
1741. Takada Y, Mukhopadhyay A, Kundu GC, Mahabeleshwar GH, Singh S, Aggarwal BB.
Hydrogen peroxide activates NF-kappa B through tyrosine phosphorylation of I
kappa B alpha and serine phosphorylation of p65: evidence for the involvement of I
kappa B alpha kinase and Syk protein-tyrosine kinase. J Biol Chem 278: 2423324241,
2003.
1742. Takahashi K, Tanase-Nicola S, ten Wolde PR. Spatio-temporal correlations can drastically change the response of a MAPK pathway. Proc Natl Acad Sci USA 107: 2473
2478, 2010.
1743. Takata Y, Liu J, Yin F, Collins AR, Lyon CJ, Lee CH, Atkins AR, Downes M, Barish GD,
Evans RM, Hsueh WA, Tangirala RK. PPARdelta-mediated antiinflammatory mechanisms inhibit angiotensin II-accelerated atherosclerosis. Proc Natl Acad Sci USA 105:
4277 4282, 2008.
1744. Takayama Y, May P, Anderson RG, Herz J. Low density lipoprotein receptor-related
protein 1 (LRP1) controls endocytosis and c-CBL-mediated ubiquitination of the
platelet-derived growth factor receptor beta (PDGFR beta). J Biol Chem 280:
18504 18510, 2005.
1745. Takeda M, Yamashita T, Sasaki N, Nakajima K, Kita T, Shinohara M, Ishida T, Hirata
K. Oral administration of an active form of vitamin D3 (calcitriol) decreases atherosclerosis in mice by inducing regulatory T cells and immature dendritic cells with
tolerogenic functions. Arterioscler Thromb Vasc Biol 30: 24952503, 2010.
1746. Takemura Y, Ouchi N, Shibata R, Aprahamian T, Kirber MT, Summer RS, Kihara S,
Walsh K. Adiponectin modulates inflammatory reactions via calreticulin receptordependent clearance of early apoptotic bodies. J Clin Invest 117: 375386, 2007.
1747. Taleb S, Romain M, Ramkhelawon B, Uyttenhove C, Pasterkamp G, Herbin O,
Esposito B, Perez N, Yasukawa H, Van Snick J, Yoshimura A, Tedgui A, Mallat Z. Loss
of SOCS3 expression in T cells reveals a regulatory role for interleukin-17 in atherosclerosis. J Exp Med 206: 20672077, 2009.
1748. Taleb S, Tedgui A, Mallat Z. Interleukin-17: friend or foe in atherosclerosis? Curr Opin
Lipidol 21: 404 408, 2010.
1749. Tall AR. Cholesterol efflux pathways and other potential mechanisms involved in the
athero-protective effect of high density lipoproteins. J Intern Med 263: 256 273,
2008.
1750. Tamarappoo B, Dey D, Shmilovich H, Nakazato R, Gransar H, Cheng VY, Friedman
JD, Hayes SW, Thomson LEJ, Slomka PJ, Rozanski A, Berman DS. Increased pericardial fat volume measured from noncontrast CT predicts myocardial ischemia by
SPECT. JACC: Cardiovascular Imaging 3: 1104 1112, 2010.
1532
1754. Tang EH, Shvartz E, Shimizu K, Rocha VZ, Zheng C, Fukuda D, Shi GP, Sukhova G,
Libby P. Deletion of EP4 on bone marrow-derived cells enhances inflammation and
angiotensin II-induced abdominal aortic aneurysm formation. Arterioscler Thromb
Vasc Biol 31: 261269, 2011.
1755. Tang Z, Wang A, Yuan F, Yan Z, Liu B, Chu JS, Helms JA, Li S. Differentiation of
multipotent vascular stem cells contributes to vascular diseases. Nature Commun 3:
875, 2012.
1756. Tangirala RK, Bischoff ED, Joseph SB, Wagner BL, Walczak R, Laffitte BA, Daige CL,
Thomas D, Heyman RA, Mangelsdorf DJ, Wang X, Lusis AJ, Tontonoz P, Schulman
IG. Identification of macrophage liver X receptors as inhibitors of atherosclerosis.
Proc Natl Acad Sci USA 99: 11896 11901, 2002.
1757. Tanigawa H, Billheimer JT, Tohyama J, Zhang Y, Rothblat G, Rader DJ. Expression of
cholesteryl ester transfer protein in mice promotes macrophage reverse cholesterol
transport. Circulation 116: 12671273, 2007.
1758. Taniyama Y, Fuse H, Satomi T, Tozawa R, Yasuhara Y, Shimakawa K, Shibata S,
Hattori M, Nakata M, Taketomi S. Loss of lysophospholipase 3 increases atherosclerosis in apolipoprotein E-deficient mice. Biochem Biophys Res Commun 330: 104
110, 2005.
1759. Tanizawa K. Production of H2S by 3-mercaptopyruvate sulfurtransferase. J Biochem
2011.
1760. Tarbell JM, Pahakis MY. Mechanotransduction and the glycocalyx. J Intern Med 259:
339 350, 2006.
1761. Tardif JC, LAllier PL, Ibrahim R, Gregoire JC, Nozza A, Cossette M, Kouz S, Lavoie
MA, Paquin J, Brotz TM, Taub R, Pressacco J. Treatment with 5-lipoxygenase inhibitor VIA-2291 (Atreleuton) in patients with recent acute coronary syndrome. Circ
Cardiovasc Imaging 3: 298 307, 2010.
1762. Tarling EJ, Edwards PA. ATP binding cassette transporter G1 (ABCG1) is an intracellular sterol transporter. Proc Natl Acad Sci USA 108: 19719 19724, 2011.
1763. Tarling EJ, Edwards PA. Dancing with the sterols: critical roles for ABCG1, ABCA1,
miRNAs, and nuclear and cell surface receptors in controlling cellular sterol homeostasis. Biochim Biophys Acta 2011.
1764. Taub DD, Oppenheim JJ. Review of the chemokine meeting the Third International
Symposium of Chemotactic Cytokines. Cytokine 5: 175179, 1993.
1765. Tchikov V, Bertsch U, Fritsch J, Edelmann B, Schutze S. Subcellular compartmentalization of TNF receptor-1 and CD95 signaling pathways. Eur J Cell Biol 2010.
1766. Tedgui A, Mallat Z. Cytokines in atherosclerosis: pathogenic and regulatory pathways. Physiol Rev 86: 515581, 2006.
1767. ten Klooster JP, Jaffer ZM, Chernoff J, Hordijk PL. Targeting and activation of Rac1
are mediated by the exchange factor beta-Pix. J Cell Biol 172: 759 769, 2006.
1768. Tennant GM, Wadsworth RM, Kennedy S. PAR-2 mediates increased inflammatory
cell adhesion and neointima formation following vascular injury in the mouse. Atherosclerosis 198: 57 64, 2008.
1769. Terasaka N, Wang N, Yvan-Charvet L, Tall AR. High-density lipoprotein protects
macrophages from oxidized low-density lipoprotein-induced apoptosis by promoting efflux of 7-ketocholesterol via ABCG1. Proc Natl Acad Sci USA 104: 1509315098,
2007.
1770. Terasaka N, Westerterp M, Koetsveld J, Fernandez-Hernando C, Yvan-Charvet L,
Wang N, Sessa WC, Tall AR. ATP-binding cassette transporter G1 and high-density
lipoprotein promote endothelial NO synthesis through a decrease in the interaction
of caveolin-1 and endothelial NO synthase. Arterioscler Thromb Vasc Biol 30: 2219
2225, 2010.
1740. Takada Y, Khuri FR, Aggarwal BB. Protein farnesyltransferase inhibitor (SCH 66336)
abolishes NF-kappaB activation induced by various carcinogens and inflammatory
stimuli leading to suppression of NF-kappaB-regulated gene expression and upregulation of apoptosis. J Biol Chem 279: 2628726299, 2004.
1753. Tang EH, Shimizu K, Christen T, Rocha VZ, Shvartz E, Tesmenitsky Y, Sukhova G,
Shi GP, Libby P. Lack of EP4 receptors on bone marrow-derived cells enhances
inflammation in atherosclerotic lesions. Cardiovasc Res 89: 234 243, 2011.
1783. Thelen M, Stein JV. How chemokines invite leukocytes to dance. Nat Immunol 9:
953959, 2008.
1784. Thi MM, Tarbell JM, Weinbaum S, Spray DC. The role of the glycocalyx in reorganization of the actin cytoskeleton under fluid shear stress: a bumper-car model. Proc
Natl Acad Sci USA 101: 1648316488, 2004.
1785. Thiery J, Nebendahl K, Rapp K, Kluge R, Teupser D, Seidel D. Low atherosclerotic
response of a strain of rabbits to diet-induced hypercholesterolemia. Arterioscler
Thromb Vasc Biol 15: 11811188, 1995.
1786. Thomas MC, Pickering RJ, Tsorotes D, Koitka A, Sheehy K, Bernardi S, Toffoli B,
Nguyen-Huu TP, Head GA, Fu Y, Chin-Dusting J, Cooper ME, Tikellis C. Genetic
Ace2 deficiency accentuates vascular inflammation and atherosclerosis in the ApoE
knockout mouse. Circ Res 107: 888 897, 2010.
1787. Thompson RC, Allam AH, Lombardi GP, Wann LS, Sutherland ML, Sutherland JD,
Soliman MA, Frohlich B, Mininberg DT, Monge JM, Vallodolid CM, Cox SL, Abd
El-Maksoud G, Badr I, Miyamoto MI, El-Halim Nur El-Din A, Narula J, Finch CE,
Thomas GS. Atherosclerosis across 4000 years of human history: the Horus study of
four ancient populations. Lancet 2013.
1788. Thorp E, Cui D, Schrijvers DM, Kuriakose G, Tabas I. Mertk receptor mutation
reduces efferocytosis efficiency and promotes apoptotic cell accumulation and
plaque necrosis in atherosclerotic lesions of apoe/ mice. Arterioscler Thromb Vasc
Biol 28: 14211428, 2008.
1789. Thorp E, Li G, Seimon TA, Kuriakose G, Ron D, Tabas I. Reduced apoptosis and
plaque necrosis in advanced atherosclerotic lesions of Apoe/ and Ldlr/ mice
lacking CHOP. Cell Metab 9: 474 481, 2009.
1790. Thorp E, Li Y, Bao L, Yao PM, Kuriakose G, Rong J, Fisher EA, Tabas I. Brief report:
increased apoptosis in advanced atherosclerotic lesions of Apoe/ mice lacking
macrophage Bcl-2. Arterioscler Thromb Vasc Biol 29: 169 172, 2009.
1791. Thorp E, Subramanian M, Tabas I. The role of macrophages and dendritic cells in the
clearance of apoptotic cells in advanced atherosclerosis. Eur J Immunol 41: 2515
2518, 2011.
1792. Thum T, Borlak J. LOX-1 receptor blockade abrogates oxLDL-induced oxidative
DNA damage and prevents activation of the transcriptional repressor Oct-1 in human coronary arterial endothelium. J Biol Chem 283: 19456 19464, 2008.
1793. Tietge UJ, Pratico D, Ding T, Funk CD, Hildebrand RB, Van Berkel T, Van Eck M.
Macrophage-specific expression of group IIA sPLA2 results in accelerated atherogenesis by increasing oxidative stress. J Lipid Res 46: 1604 1614, 2005.
1794. Tilley RE, Pedersen B, Pawlinski R, Sato Y, Erlich JH, Shen Y, Day S, Huang Y, Eitzman
DT, Boisvert WA, Curtiss LK, Fay WP, Mackman N. Atherosclerosis in mice is not
affected by a reduction in tissue factor expression. Arterioscler Thromb Vasc Biol 26:
555562, 2006.
1795. Timmins JM, Lee JY, Boudyguina E, Kluckman KD, Brunham LR, Mulya A, Gebre AK,
Coutinho JM, Colvin PL, Smith TL, Hayden MR, Maeda N, Parks JS. Targeted inactivation of hepatic Abca1 causes profound hypoalphalipoproteinemia and kidney
hypercatabolism of apoA-I. J Clin Invest 115: 13331342, 2005.
1796. Ting LH, Jahn JR, Jung JI, Shuman BR, Feghhi S, Han SJ, Rodriguez ML, Sniadecki NJ.
Flow mechanotransduction regulates traction forces, intercellular forces, and adherens junctions. Am J Physiol Heart Circ Physiol 302: H2220 H2229, 2012.
1797. To K, Agrotis A, Besra G, Bobik A, Toh BH. NKT cell subsets mediate differential
proatherogenic effects in ApoE/ mice. Arterioscler Thromb Vasc Biol 29: 671 677,
2009.
1798. Tole S, Durkan AM, Huang YW, Liu GY, Leung A, Jones LL, Taylor JA, Robinson LA.
Thromboxane prostanoid receptor stimulation induces shedding of the transmembrane chemokine CX3CL1 yet enhances CX3CL1-dependent leukocyte adhesion.
Am J Physiol Cell Physiol 298: C1469 C1480, 2010.
1799. Tom B, Garrelds IM, Scalbert E, Stegmann AP, Boomsma F, Saxena PR, Danser AH.
ACE-versus chymase-dependent angiotensin II generation in human coronary arteries: a matter of efficiency? Arterioscler Thromb Vasc Biol 23: 251256, 2003.
1800. Tomar A, Schlaepfer DD. Focal adhesion kinase: switching between GAPs and GEFs
in the regulation of cell motility. Curr Opin Cell Biol 21: 676 683, 2009.
1533
1774. Teslovich TM, Musunuru K, Smith AV, Edmondson AC, Stylianou IM, Koseki M,
Pirruccello JP, Ripatti S, Chasman DI, Willer CJ, Johansen CT, Fouchier SW, Isaacs A,
Peloso GM, Barbalic M, Ricketts SL, Bis JC, Aulchenko YS, Thorleifsson G, Feitosa
MF, Chambers J, Orho-Melander M, Melander O, Johnson T, Li X, Guo X, Li M, Shin
Cho Y, Jin Go M, Jin Kim Y, Lee JY, Park T, Kim K, Sim X, Twee-Hee Ong R,
Croteau-Chonka DC, Lange LA, Smith JD, Song K, Hua Zhao J, Yuan X, Luan J,
Lamina C, Ziegler A, Zhang W, Zee RY, Wright AF, Witteman JC, Wilson JF, Willemsen G, Wichmann HE, Whitfield JB, Waterworth DM, Wareham NJ, Waeber G,
Vollenweider P, Voight BF, Vitart V, Uitterlinden AG, Uda M, Tuomilehto J, Thompson JR, Tanaka T, Surakka I, Stringham HM, Spector TD, Soranzo N, Smit JH, Sinisalo
J, Silander K, Sijbrands EJ, Scuteri A, Scott J, Schlessinger D, Sanna S, Salomaa V,
Saharinen J, Sabatti C, Ruokonen A, Rudan I, Rose LM, Roberts R, Rieder M, Psaty
BM, Pramstaller PP, Pichler I, Perola M, Penninx BW, Pedersen NL, Pattaro C,
Parker AN, Pare G, Oostra BA, ODonnell CJ, Nieminen MS, Nickerson DA, Montgomery GW, Meitinger T, McPherson R, McCarthy MI, McArdle W, Masson D,
Martin NG, Marroni F, Mangino M, Magnusson PK, Lucas G, Luben R, Loos RJ, Lokki
ML, Lettre G, Langenberg C, Launer LJ, Lakatta EG, Laaksonen R, Kyvik KO, Kronenberg F, Konig IR, Khaw KT, Kaprio J, Kaplan LM, Johansson A, Jarvelin MR, Cecile
JWJA, Ingelsson E, Igl W, Kees Hovingh G, Hottenga JJ, Hofman A, Hicks AA, Hengstenberg C, Heid IM, Hayward C, Havulinna AS, Hastie ND, Harris TB, Haritunians
T, Hall AS, Gyllensten U, Guiducci C, Groop LC, Gonzalez E, Gieger C, Freimer NB,
Ferrucci L, Erdmann J, Elliott P, Ejebe KG, Doring A, Dominiczak AF, Demissie S,
Deloukas P, de Geus EJ, de Faire U, Crawford G, Collins FS, Chen YD, Caulfield MJ,
Campbell H, Burtt NP, Bonnycastle LL, Boomsma DI, Boekholdt SM, Bergman RN,
Barroso I, Bandinelli S, Ballantyne CM, Assimes TL, Quertermous T, Altshuler D,
Seielstad M, Wong TY, Tai ES, Feranil AB, Kuzawa CW, Adair LS, Taylor HA, Jr,
Borecki IB, Gabriel SB, Wilson JG, Holm H, Thorsteinsdottir U, Gudnason V, Krauss
RM, Mohlke KL, Ordovas JM, Munroe PB, Kooner JS, Tall AR, Hegele RA, Kastelein
JJ, Schadt EE, Rotter JI, Boerwinkle E, Strachan DP, Mooser V, Stefansson K, Reilly
MP, Samani NJ, Schunkert H, Cupples LA, Sandhu MS, Ridker PM, Rader DJ, van
Duijn CM, Peltonen L, Abecasis GR, Boehnke M, Kathiresan S. Biological, clinical and
population relevance of 95 loci for blood lipids. Nature 466: 707713, 2010.
1782. The Homocysteine Studies Collaboration. Homocysteine and risk of ischemic heart
disease and stroke: a meta-analysis. JAMA 288: 20152022, 2002.
PAUL N. HOPKINS
1801. Tomilov AA, Bicocca V, Schoenfeld RA, Giorgio M, Migliaccio E, Ramsey JJ, Hagopian
K, Pelicci PG, Cortopassi GA. Decreased superoxide production in macrophages of
long-lived p66Shc knock-out mice. J Biol Chem 285: 11531165, 2010.
1802. Tordjman K, Bernal-Mizrachi C, Zemany L, Weng S, Feng C, Zhang F, Leone TC,
Coleman T, Kelly DP, Semenkovich CF. PPARalpha deficiency reduces insulin resistance and atherosclerosis in apoE-null mice. J Clin Invest 107: 10251034, 2001.
1803. Torreggiani M, Liu H, Wu J, Zheng F, Cai W, Striker G, Vlassara H. Advanced
glycation end product receptor-1 transgenic mice are resistant to inflammation,
oxidative stress, and postinjury intimal hyperplasia. Am J Pathol 175: 17221732,
2009.
1804. Townsend DM, Manevich Y, He L, Hutchens S, Pazoles CJ, Tew KD. Novel role for
glutathione S-transferase Pi. Regulator of protein S-glutathionylation following oxidative and nitrosative stress. J Biol Chem 284: 436 445, 2009.
1805. Tran-Lundmark K, Tran PK, Paulsson-Berne G, Friden V, Soininen R, Tryggvason K,
Wight TN, Kinsella MG, Boren J, Hedin U. Heparan sulfate in perlecan promotes
mouse atherosclerosis: roles in lipid permeability, lipid retention, and smooth muscle cell proliferation. Circ Res 103: 4352, 2008.
1806. Trelle S, Reichenbach S, Wandel S, Hildebrand P, Tschannen B, Villiger PM, Egger M,
Juni P. Cardiovascular safety of non-steroidal anti-inflammatory drugs: network
meta-analysis. BMJ 342: c7086, 2011.
1807. Tribble DL, Gong EL, Leeuwenburgh C, Heinecke JW, Carlson EL, Verstuyft JG,
Epstein CJ. Fatty streak formation in fat-fed mice expressing human copper-zinc
superoxide dismutase. Arterioscler Thromb Vasc Biol 17: 1734 1740, 1997.
1820. Tuinenburg A, Mauser-Bunschoten EP, Verhaar MC, Biesma DH, Schutgens RE.
Cardiovascular disease in patients with hemophilia. J Thromb Haemost 7: 247254,
2009.
1821. Tupin E, Nicoletti A, Elhage R, Rudling M, Ljunggren HG, Hansson GK, Berne GP.
CD1d-dependent activation of NKT cells aggravates atherosclerosis. J Exp Med 199:
417 422, 2004.
1822. Turnbull F, Neal B, Ninomiya T, Algert C, Arima H, Barzi F, Bulpitt C, Chalmers J,
Fagard R, Gleason A, Heritier S, Li N, Perkovic V, Woodward M, MacMahon S.
Effects of different regimens to lower blood pressure on major cardiovascular events
in older and younger adults: meta-analysis of randomised trials. BMJ 336: 11211123,
2008.
1823. Turnbull F, Neal B, Pfeffer M, Kostis J, Algert C, Woodward M, Chalmers J, Zanchetti
A, MacMahon S. Blood pressure-dependent and independent effects of agents that
inhibit the renin-angiotensin system. J Hypertens 25: 951958, 2007.
1824. Twu YC, Gold MR, Teh HS. TNFR1 delivers pro-survival signals that are required for
limiting TNFR2-dependent activation-induced cell death (AICD) in CD8() T cells.
Eur J Immunol 2010.
1825. Tybulewicz VL, Henderson RB. Rho family GTPases and their regulators in lymphocytes. Nat Rev Immunol 9: 630 644, 2009.
1826. Tziakas DN, Chalikias GK, Stakos D, Boudoulas H. The role of red blood cells in the
progression and instability of atherosclerotic plaque. Int J Cardiol. In press.
1827. Tzima E. Role of small GTPases in endothelial cytoskeletal dynamics and the shear
stress response. Circ Res 98: 176 185, 2006.
1828. Tzima E, Irani-Tehrani M, Kiosses WB, Dejana E, Schultz DA, Engelhardt B, Cao G,
DeLisser H, Schwartz MA. A mechanosensory complex that mediates the endothelial cell response to fluid shear stress. Nature 437: 426 431, 2005.
1829. Uchiyama T, Atsuta H, Utsugi T, Oguri M, Hasegawa A, Nakamura T, Nakai A,
Nakata M, Maruyama I, Tomura H, Okajima F, Tomono S, Kawazu S, Nagai R,
Kurabayashi M. HSF1 and constitutively active HSF1 improve vascular endothelial
function (heat shock proteins improve vascular endothelial function). Atherosclerosis
190: 321329, 2007.
1830. Uehara Y, Urata H, Ideishi M, Arakawa K, Saku K. Chymase inhibition suppresses
high-cholesterol diet-induced lipid accumulation in the hamster aorta. Cardiovasc Res
55: 870 876, 2002.
1812. Tsai YC, Hsieh HJ, Liao F, Ni CW, Chao YJ, Hsieh CY, Wang DL. Laminar flow
attenuates interferon-induced inflammatory responses in endothelial cells. Cardiovasc Res 74: 497505, 2007.
1813. Tsao CW, Lin CF, Wu HT, Ma CT, Huang WC, Hsieh CY, Choi PC, Young KC.
Glycogen synthase kinase-3beta is critical for interferon-alpha-induced serotonin
uptake in human Jurkat T cells. J Cell Physiol 227: 2556 2566, 2012.
1832. Umetani M, Mangelsdorf DJ, Shaul PW. Abstract 18427: 27-hydroxycholesterol, the
first identified endogenous SERM, promotes atherogenesis in mice. Circulation 122:
A18427, 2010.
1814. Tsiokas L. Function and regulation of TRPP2 at the plasma membrane. Am J Physiol
Renal Physiol 297: F1F9, 2009.
1815. Tsou CL, Peters W, Si Y, Slaymaker S, Aslanian AM, Weisberg SP, Mack M, Charo IF.
Critical roles for CCR2 and MCP-3 in monocyte mobilization from bone marrow
and recruitment to inflammatory sites. J Clin Invest 117: 902909, 2007.
1816. Tsoyi K, Kim WS, Kim YM, Kim HJ, Seo HG, Lee JH, Yun-Choi HS, Chang KC.
Upregulation of PTEN by CKD712, a synthetic tetrahydroisoquinoline alkaloid, selectively inhibits lipopolysaccharide-induced VCAM-1 but not ICAM-1 expression in
human endothelial cells. Atherosclerosis 207: 412 419, 2009.
1817. Tsuchiya K, Tanaka J, Shuiqing Y, Welch CL, DePinho RA, Tabas I, Tall AR, Goldberg
IJ, Accili D. FoxOs integrate pleiotropic actions of insulin in vascular endothelium to
protect mice from atherosclerosis. Cell Metab 15: 372381, 2012.
1818. Tsuchiya K, Westerterp M, Murphy AJ, Subramanian V, Ferrante A, Tall AR, Accili D.
Expanded granulocyte/monocyte compartment in myeloid-specific triple Foxo
knockout increases oxidative stress and accelerates atherosclerosis in mice. Circ Res
112: 9921003, 2013.
1534
1835. Upadhya S, Mooteri S, Peckham N, Pai RG. Atherogenic effect of interleukin-2 and
antiatherogenic effect of interleukin-2 antibody in apo-E-deficient mice. Angiology 55:
289 294, 2004.
1836. Urbich C, Mallat Z, Tedgui A, Clauss M, Zeiher AM, Dimmeler S. Upregulation of
TRAF-3 by shear stress blocks CD40-mediated endothelial activation. J Clin Invest
108: 14511458, 2001.
1837. Vaisman BL, Andrews KL, Khong SM, Wood KC, Moore XL, Fu Y, Kepka-Lenhart
DM, Morris SM Jr, Remaley AT, Chin-Dusting JP. Selective endothelial overexpression of arginase II induces endothelial dysfunction and hypertension and enhances
atherosclerosis in mice. PLoS ONE 7: e39487, 2012.
1838. Vaisman BL, Demosky SJ, Stonik JA, Ghias M, Knapper CL, Sampson ML, Dai C,
Levine SJ, Remaley AT. Endothelial expression of human ABCA1 in mice increases
1808. Trigatti B, Rayburn H, Vinals M, Braun A, Miettinen H, Penman M, Hertz M, Schrenzel M, Amigo L, Rigotti A, Krieger M. Influence of the high density lipoprotein
receptor SR-BI on reproductive and cardiovascular pathophysiology. Proc Natl Acad
Sci USA 96: 93229327, 1999.
1847. Van der Heiden K, Hierck BP, Krams R, de Crom R, Cheng C, Baiker M, Pourquie
MJ, Alkemade FE, DeRuiter MC, Gittenberger-de Groot AC, Poelmann RE. Endothelial primary cilia in areas of disturbed flow are at the base of atherosclerosis.
Atherosclerosis 196: 542550, 2008.
1848. Van Eck M, Bos IS, Hildebrand RB, Van Rij BT, Van Berkel TJ. Dual role for scavenger
receptor class B, type I on bone marrow-derived cells in atherosclerotic lesion
development. Am J Pathol 165: 785794, 2004.
1849. Van Eck M, Bos IS, Kaminski WE, Orso E, Rothe G, Twisk J, Bottcher A, Van
Amersfoort ES, Christiansen-Weber TA, Fung-Leung WP, Van Berkel TJ, Schmitz G.
Leukocyte ABCA1 controls susceptibility to atherosclerosis and macrophage recruitment into tissues. Proc Natl Acad Sci USA 99: 6298 6303, 2002.
1850. Van Eck M, Zimmermann R, Groot PH, Zechner R, Van Berkel TJ. Role of macrophage-derived lipoprotein lipase in lipoprotein metabolism and atherosclerosis. Arterioscler Thromb Vasc Biol 20: E53 62, 2000.
1851. Van Eden W, Wick G, Albani S, Cohen I. Stress, heat shock proteins, and autoimmunity: how immune responses to heat shock proteins are to be used for the control
of chronic inflammatory diseases. Ann NY Acad Sci 1113: 217237, 2007.
1852. Van Es T, van Puijvelde GH, Foks AC, Habets KL, Bot I, Gilboa E, Van Berkel TJ,
Kuiper J. Vaccination against Foxp3() regulatory T cells aggravates atherosclerosis.
Atherosclerosis 209: 74 80, 2010.
1853. Van Es T, van Puijvelde GH, Ramos OH, Segers FM, Joosten LA, van den Berg WB,
Michon IM, de Vos P, van Berkel TJ, Kuiper J. Attenuated atherosclerosis upon
IL-17R signaling disruption in LDLr deficient mice. Biochem Biophys Res Commun 388:
261265, 2009.
1854. Van Gils JM, Derby MC, Fernandes LR, Ramkhelawon B, Ray TD, Rayner KJ,
Parathath S, Distel E, Feig JL, Alvarez-Leite JI, Rayner AJ, McDonald TO, OBrien KD,
Stuart LM, Fisher EA, Lacy-Hulbert A, Moore KJ. The neuroimmune guidance cue
netrin-1 promotes atherosclerosis by inhibiting the emigration of macrophages from
plaques. Nat Immunol 13: 136 143, 2012.
1855. Van Haperen R, de Waard M, van Deel E, Mees B, Kutryk M, van Aken T, Hamming
J, Grosveld F, Duncker DJ, de Crom R. Reduction of blood pressure, plasma cholesterol, and atherosclerosis by elevated endothelial nitric oxide. J Biol Chem 277:
48803 48807, 2002.
1857. Van Herck JL, De Meyer GR, Martinet W, Vanhoutte G, Verhoye M, Van Der Linden
A, Bult H, Vrints CJ, Herman AG. Abstract 12751: impaired elastin function promotes acute plaque rupture, cerebral infarction and sudden death in ApoE-deficient
mice. Circulation 122: A12751, 2010.
1858. Van Leeuwen M, Gijbels MJ, Duijvestijn A, Smook M, van de Gaar MJ, Heeringa P, de
Winther MP, Tervaert JW. Accumulation of myeloperoxidase-positive neutrophils in
atherosclerotic lesions in LDLR/ mice. Arterioscler Thromb Vasc Biol 28: 84 89,
2008.
1859. Van Puijvelde GH, Hauer AD, de Vos P, van den Heuvel R, van Herwijnen MJ, van der
Zee R, van Eden W, van Berkel TJ, Kuiper J. Induction of oral tolerance to oxidized
low-density lipoprotein ameliorates atherosclerosis. Circulation 114: 1968 1976,
2006.
1860. Van Puijvelde GH, van Es T, van Wanrooij EJ, Habets KL, de Vos P, van der Zee R,
van Eden W, van Berkel TJ, Kuiper J. Induction of oral tolerance to HSP60 or an
HSP60-peptide activates T cell regulation and reduces atherosclerosis. Arterioscler
Thromb Vasc Biol 27: 26772683, 2007.
1861. Van Puijvelde GHM, van Es T, van Wanrooij EJA, Habets KLL, de Vos P, van der Zee
R, van Eden W, van Berkel TJC, Kuiper J. Induction of oral tolerance to HSP60 or an
HSP60-peptide activates T Cell regulation andreduces atherosclerosis. Arterioscler
Thromb Vasc Biol 27: 26772683, 2007.
1862. Van Vlijmen BJ, Gerritsen G, Franken AL, Boesten LS, Kockx MM, Gijbels MJ, Vierboom MP, van Eck M, van De Water B, van Berkel TJ, Havekes LM. Macrophage p53
deficiency leads to enhanced atherosclerosis in APOE*3-Leiden transgenic mice.
Circ Res 88: 780 786, 2001.
1863. Van Vre EA, Ait-Oufella H, Tedgui A, Mallat Z. Apoptotic cell death and efferocytosis
in atherosclerosis. Arterioscler Thromb Vasc Biol 32: 887 893, 2012.
1864. Van Wanrooij EJ, van Puijvelde GH, de Vos P, Yagita H, van Berkel TJ, Kuiper J.
Interruption of the Tnfrsf4/Tnfsf4 (OX40/OX40L) pathway attenuates atherogenesis in low-density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc Biol
27: 204 210, 2007.
1865. Van Wanrooij EJA, de Vos P, Bixel MG, Vestweber D, van Berkel TJC, Kuiper J.
Vaccination against CD99 inhibits atherogenesis in low-density lipoprotein receptordeficient mice. Cardiovasc Res 78: 590 596, 2008.
1866. Vanhoutte PM. Endothelium-dependent contractions in hypertension: when prostacyclin becomes ugly. Hypertension 57: 526 531, 2011.
1867. Vazquez-Figueroa JG, Rinehart S, McCree A, Teramoto T, Matsushima T, Kinoshita
M, Pryor A, Alman B, Yi H, Joshi P, Voros S. Abstract 16886: first quantification of
intestinal-apoB48 and hepatic-apoB100 particles in human atherosclerotic plaque by
dual immunofluorescence staining. Circulation 122: A16886, 2010.
1868. Vecchione C, Carnevale D, Di Pardo A, Gentile MT, Damato A, Cocozza G, Antenucci G, Mascio G, Bettarini U, Landolfi A, Iorio L, Maffei A, Lembo G. Pressureinduced vascular oxidative stress is mediated through activation of integrin-linked
kinase 1/betaPIX/Rac-1 pathway. Hypertension 54: 1028 1034, 2009.
1869. Vedhachalam C, Duong PT, Nickel M, Nguyen D, Dhanasekaran P, Saito H, Rothblat
GH, Lund-Katz S, Phillips MC. Mechanism of ATP-binding cassette transporter A1mediated cellular lipid efflux to apolipoprotein A-I and formation of high density
lipoprotein particles. J Biol Chem 282: 2512325130, 2007.
1870. Vedre A, Pathak DR, Crimp M, Lum C, Koochesfahani M, Abela GS. Physical factors
that trigger cholesterol crystallization leading to plaque rupture. Atherosclerosis 203:
89 96, 2009.
1871. Veillard NR, Steffens S, Pelli G, Lu B, Kwak BR, Gerard C, Charo IF, Mach F.
Differential influence of chemokine receptors CCR2 and CXCR3 in development of
atherosclerosis in vivo. Circulation 112: 870 878, 2005.
1872. Vendrov AE, Hakim ZS, Madamanchi NR, Rojas M, Madamanchi C, Runge MS.
Atherosclerosis is attenuated by limiting superoxide generation in both macrophages
and vessel wall cells. Arterioscler Thromb Vasc Biol 27: 2714 2721, 2007.
1873. Veniant MM, Withycombe S, Young SG. Lipoprotein size and atherosclerosis susceptibility in APOE(/) and LDLR(/) mice. Arterioscler Thromb Vasc Biol 21:
15671570, 2001.
1535
1846. Van der Does AM, Beekhuizen H, Ravensbergen B, Vos T, Ottenhoff THM, van
Dissel JT, Drijfhout JW, Hiemstra PS, Nibbering PH. LL-37 directs macrophage
differentiation toward macrophages with a proinflammatory signature. J Immunol
185: 14421449, 2010.
1856. Van Haperen R, Samyn H, Moerland M, van Gent T, Peeters M, Grosveld F, van Tol
A, de Crom R. Elevated expression of phospholipid transfer protein in bone marrow
derived cells causes atherosclerosis. PLoS ONE 3: e2255, 2008.
PAUL N. HOPKINS
1874. Vergeer M, Korporaal SJ, Franssen R, Meurs I, Out R, Hovingh GK, Hoekstra M,
Sierts JA, Dallinga-Thie GM, Motazacker MM, Holleboom AG, Van Berkel TJ, Kastelein JJ, Van Eck M, Kuivenhoven JA. Genetic variant of the scavenger receptor BI in
humans. N Engl J Med 364: 136 145, 2011.
1875. Verstrepen L, Verhelst K, van Loo G, Carpentier I, Ley SC, Beyaert R. Expression,
biological activities and mechanisms of action of A20 (TNFAIP3). Biochem Pharmacol
80: 2009 2020, 2010.
1876. Vicente CP, He L, Tollefsen DM. Accelerated atherogenesis and neointima formation in heparin cofactor II deficient mice. Blood 110: 4261 4267, 2007.
1877. Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT. MicroRNAs
are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol 13: 423 433, 2011.
1878. Viji RI, Sameer Kumar VB, Kiran MS, Sudhakaran PR. Modulation of endothelial nitric
oxide synthase by fibronectin. Mol Cell Biochem 323: 91100, 2009.
1879. Vikramadithyan RK, Hu Y, Noh HL, Liang CP, Hallam K, Tall AR, Ramasamy R,
Goldberg IJ. Human aldose reductase expression accelerates diabetic atherosclerosis in transgenic mice. J Clin Invest 115: 2434 2443, 2005.
1880. Vikramadithyan RK, Kako Y, Chen G, Hu Y, Arikawa-Hirasawa E, Yamada Y, Goldberg IJ. Atherosclerosis in perlecan heterozygous mice. J Lipid Res 45: 1806 1812,
2004.
1881. Villarreal G Jr, Zhang Y, Larman HB, Gracia-Sancho J, Koo A, Garcia-Cardena G.
Defining the regulation of KLF4 expression and its downstream transcriptional targets in vascular endothelial cells. Biochem Biophys Res Commun 391: 984 989, 2010.
1883. Voight BF, Peloso GM, Orho-Melander M, Frikke-Schmidt R, Barbalic M, Jensen MK,
Hindy G, Holm H, Ding EL, Johnson T, Schunkert H, Samani NJ, Clarke R, Hopewell
JC, Thompson JF, Li M, Thorleifsson G, Newton-Cheh C, Musunuru K, Pirruccello
JP, Saleheen D, Chen L, Stewart A, Schillert A, Thorsteinsdottir U, Thorgeirsson G,
Anand S, Engert JC, Morgan T, Spertus J, Stoll M, Berger K, Martinelli N, Girelli D,
McKeown PP, Patterson CC, Epstein SE, Devaney J, Burnett MS, Mooser V, Ripatti
S, Surakka I, Nieminen MS, Sinisalo J, Lokki ML, Perola M, Havulinna A, de Faire U,
Gigante B, Ingelsson E, Zeller T, Wild P, de Bakker PI, Klungel OH, Maitland-van der
Zee AH, Peters BJ, de Boer A, Grobbee DE, Kamphuisen PW, Deneer VH, Elbers
CC, Onland-Moret NC, Hofker MH, Wijmenga C, Verschuren WM, Boer JM, van
der Schouw YT, Rasheed A, Frossard P, Demissie S, Willer C, Do R, Ordovas JM,
Abecasis GR, Boehnke M, Mohlke KL, Daly MJ, Guiducci C, Burtt NP, Surti A,
Gonzalez E, Purcell S, Gabriel S, Marrugat J, Peden J, Erdmann J, Diemert P, Willenborg C, Konig IR, Fischer M, Hengstenberg C, Ziegler A, Buysschaert I, Lambrechts
D, Van de Werf F, Fox KA, El Mokhtari NE, Rubin D, Schrezenmeir J, Schreiber S,
Schafer A, Danesh J, Blankenberg S, Roberts R, McPherson R, Watkins H, Hall AS,
Overvad K, Rimm E, Boerwinkle E, Tybjaerg-Hansen A, Cupples LA, Reilly MP,
Melander O, Mannucci PM, Ardissino D, Siscovick D, Elosua R, Stefansson K,
ODonnell CJ, Salomaa V, Rader DJ, Peltonen L, Schwartz SM, Altshuler D, Kathiresan S. Plasma HDL cholesterol and risk of myocardial infarction: a mendelian randomisation study. Lancet 380: 572580, 2012.
1891. Vuletic S, Dong W, Wolfbauer G, Tang C, Albers JJ. PLTP regulates STAT3 and
NFkappaB in differentiated THP1 cells and human monocyte-derived macrophages.
Biochim Biophys Acta 1813: 19171924, 2011.
1892. Walton KA, Hsieh X, Gharavi N, Wang S, Wang G, Yeh M, Cole AL, Berliner JA.
Receptors involved in the oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine-mediated synthesis of interleukin-8. A role for Toll-like receptor 4 and
a glycosylphosphatidylinositol-anchored protein. J Biol Chem 278: 2966129666,
2003.
1893. Wan JB, Huang LL, Rong R, Tan R, Wang J, Kang JX. Endogenously decreasing tissue
n-6/n-3 fatty acid ratio reduces atherosclerotic lesions in apolipoprotein E-deficient
mice by inhibiting systemic and vascular inflammation. Arterioscler Thromb Vasc Biol
30: 24872494, 2010.
1894. Wan W, Lim JK, Lionakis MS, Rivollier A, McDermott DH, Kelsall BL, Farber JM,
Murphy PM. Genetic deletion of chemokine receptor Ccr6 decreases atherogenesis
in ApoE-deficient mice. Circ Res 109: 374 381, 2011.
1895. Wang HW, Liu PY, Oyama N, Rikitake Y, Kitamoto S, Gitlin J, Liao JK, Boisvert WA.
Deficiency of ROCK1 in bone marrow-derived cells protects against atherosclerosis
in LDLR/ mice. FASEB J 22: 35613570, 2008.
1896. Wang H, Jiang X, Yang F, Gaubatz JW, Ma L, Magera MJ, Yang X, Berger PB, Durante
W, Pownall HJ, Schafer AI. Hyperhomocysteinemia accelerates atherosclerosis in
cystathionine beta-synthase and apolipoprotein E double knock-out mice with and
without dietary perturbation. Blood 101: 39013907, 2003.
1897. Wang H, Tang R, Zhang W, Amirikian K, Geng Z, Geng J, Hebbel RP, Xia L, Marth JD,
Fukuda M, Katoh S, Huo Y. Core2 1 6-N-glucosaminyltransferase-I is crucial for the
formation of atherosclerotic lesions in apolipoprotein E-deficient mice. Arterioscler
Thromb Vasc Biol 29: 180 187, 2009.
1898. Wang H, Zhang W, Zhu C, Bucher C, Blazar BR, Zhang C, Chen JF, Linden J, Wu C,
Huo Y. Inactivation of the adenosine A2A receptor protects apolipoprotein E-deficient mice from atherosclerosis. Arterioscler Thromb Vasc Biol 29: 1046 1052, 2009.
1899. Wang J, Pan S, Berk BC. Glutaredoxin mediates Akt and eNOS activation by flow in
a glutathione reductase-dependent manner. Arterioscler Thromb Vasc Biol 27: 1283
1288, 2007.
1900. Wang KY, Tanimoto A, Guo X, Yamada S, Shimajiri S, Murata Y, Ding Y, Tsutsui M,
Kato S, Watanabe T, Ohtsu H, Hirano K, Kohno K, Sasaguri Y. Histamine deficiency
decreases atherosclerosis and inflammatory response in apolipoprotein E knockout
mice independently of serum cholesterol level. Arterioscler Thromb Vasc Biol 31:
800 807, 2011.
1901. Wang L, Johnson EE, Shi HN, Walker WA, Wessling-Resnick M, Cherayil BJ. Attenuated inflammatory responses in hemochromatosis reveal a role for iron in the
regulation of macrophage cytokine translation. J Immunol 181: 27232731, 2008.
1884. Voloshyna I, Reiss AB. The ABC transporters in lipid flux and atherosclerosis. Prog
Lipid Res 2011.
1902. Wang L, Miller C, Swarthout RF, Rao M, Mackman N, Taubman MB. Vascular
smooth muscle-derived tissue factor is critical for arterial thrombosis after ferric
chloride-induced injury. Blood 113: 705713, 2009.
1885. Von Der Thusen JH, Kuiper J, Fekkes ML, De Vos P, Van Berkel TJ, Biessen EA.
Attenuation of atherogenesis by systemic and local adenovirus-mediated gene transfer of interleukin-10 in LDLr/ mice. FASEB J 15: 2730 2732, 2001.
1903. Wang M, Monticone RE, Lakatta EG. Arterial aging: a journey into subclinical arterial
disease. Curr Opin Nephrol Hypertens 19: 201207, 2010.
1886. Von der Thusen JH, van Berkel TJ, Biessen EA. Induction of rapid atherogenesis by
perivascular carotid collar placement in apolipoprotein E-deficient and low-density
lipoprotein receptor-deficient mice. Circulation 103: 1164 1170, 2001.
1904. Wang M, Takagi G, Asai K, Resuello RG, Natividad FF, Vatner DE, Vatner SF, Lakatta
EG. Aging increases aortic MMP-2 activity and angiotensin II in nonhuman primates.
Hypertension 41: 1308 1316, 2003.
1887. Von der Thusen JH, van Vlijmen BJ, Hoeben RC, Kockx MM, Havekes LM, van Berkel
TJ, Biessen EA. Induction of atherosclerotic plaque rupture in apolipoprotein E/
mice after adenovirus-mediated transfer of p53. Circulation 105: 2064 2070, 2002.
1905. Wang M, Zhao D, Spinetti G, Zhang J, Jiang LQ, Pintus G, Monticone R, Lakatta EG.
Matrix metalloproteinase 2 activation of transforming growth factor-beta1 (TGFbeta1) and TGF-beta1-type II receptor signaling within the aged arterial wall. Arterioscler Thromb Vasc Biol 26: 15031509, 2006.
1536
1906. Wang M, Zukas AM, Hui Y, Ricciotti E, Pure E, FitzGerald GA. Deletion of microsomal prostaglandin E synthase-1 augments prostacyclin and retards atherogenesis.
Proc Natl Acad Sci USA 103: 1450714512, 2006.
1907. Wang N, Miao H, Li YS, Zhang P, Haga JH, Hu Y, Young A, Yuan S, Nguyen P, Wu
CC, Chien S. Shear stress regulation of Kruppel-like factor 2 expression is flow
pattern-specific. Biochem Biophys Res Commun 341: 1244 1251, 2006.
1882. Virmani R, Burke AP, Farb A, Kolodgie FD. Pathology of the vulnerable plaque. J Am
Coll Cardiol 47: C1318, 2006.
1926. Wang Z, Nicholls SJ, Rodriguez ER, Kummu O, Horkko S, Barnard J, Reynolds WF,
Topol EJ, DiDonato JA, Hazen SL. Protein carbamylation links inflammation, smoking, uremia and atherogenesis. Nat Med 13: 1176 1184, 2007.
1909. Wang R. Signaling pathways for the vascular effects of hydrogen sulfide. Curr Opin
Nephrol Hypertens 20: 107112, 2011.
1927. Wannamethee SG, Lowe GDO, Shaper AG, Rumley A, Lennon L, Whincup PH.
Associations between cigarette smoking, pipe/cigar smoking, and smoking cessation,
and haemostatic and inflammatory markers for cardiovascular disease. Eur Heart J
26: 17651773, 2005.
1910. Wang S, Subramanian V, Lu H, Howatt DA, Moorleghen JJ, Charnigo R, Cassis LA,
Daugherty A. Deficiency of receptor-associated protein attenuates angiotensin IIinduced atherosclerosis in hypercholesterolemic mice without influencing abdominal aortic aneurysms. Atherosclerosis 220: 375380, 2012.
1911. Wang S, Zhang M, Liang B, Xu J, Xie Z, Liu C, Viollet B, Yan D, Zou MH. AMPKalpha2 deletion causes aberrant expression and activation of NAD(P)H oxidase and
consequent endothelial dysfunction in vivo: role of 26S proteasomes. Circ Res 106:
11171128, 2010.
1912. Wang W, Ha CH, Jhun BS, Wong C, Jain MK, Jin ZG. Fluid shear stress stimulates
phosphorylation-dependent nuclear export of HDAC5 and mediates expression of
KLF2 and eNOS. Blood 115: 29712979, 2010.
1913. Wang X, Collins HL, Ranalletta M, Fuki IV, Billheimer JT, Rothblat GH, Tall AR, Rader
DJ. Macrophage ABCA1 and ABCG1, but not SR-BI, promote macrophage reverse
cholesterol transport in vivo. J Clin Invest 117: 2216 2224, 2007.
1914. Wang X, Dong J, Zhao Y, Li Y, Wu M. Adenovirus-mediated sphingomyelin synthase
2 increases atherosclerotic lesions in ApoE KO mice. Lipids Health Dis 10: 7, 2011.
1916. Wang X, Phelan SA, Petros C, Taylor EF, Ledinski G, Jurgens G, Forsman-Semb K,
Paigen B. Peroxiredoxin 6 deficiency and atherosclerosis susceptibility in mice: significance of genetic background for assessing atherosclerosis. Atherosclerosis 177:
6170, 2004.
1917. Wang X, Ria M, Kelmenson PM, Eriksson P, Higgins DC, Samnegard A, Petros C,
Rollins J, Bennet AM, Wiman B, de Faire U, Wennberg C, Olsson PG, Ishii N,
Sugamura K, Hamsten A, Forsman-Semb K, Lagercrantz J, Paigen B. Positional identification of TNFSF4, encoding OX40 ligand, as a gene that influences atherosclerosis
susceptibility. Nat Genet 37: 365372, 2005.
1918. Wang XQ, Nigro P, World C, Fujiwara K, Yan C, Berk BC. Thioredoxin interacting
protein promotes endothelial cell inflammation in response to disturbed flow by
increasing leukocyte adhesion and repressing Kruppel-like factor 2. Circ Res 110:
560 568, 2012.
1919. Wang Y, Li J, Tang L, Charnigo R, de Villiers W, Eckhardt E. T-lymphocyte responses
to intestinally absorbed antigens can contribute to adipose tissue inflammation and
glucose intolerance during high fat feeding. PLoS ONE 5: e13951, 2010.
1920. Wang Y, Niimi M, Nishijima K, Waqar AB, Yu Y, Koike T, Kitajima S, Liu E, Inoue T,
Kohashi M, Keyamura Y, Yoshikawa T, Zhang J, Ma L, Zha X, Watanabe T, Asada Y,
Chen YE, Fan J. Human apolipoprotein A-II protects against diet-induced atherosclerosis in transgenic rabbits. Arterioscler Thromb Vasc Biol 33: 224 231, 2013.
1940. Welch CL, Sun Y, Arey BJ, Lemaitre V, Sharma N, Ishibashi M, Sayers S, Li R, Gorelik
A, Pleskac N, Collins-Fletcher K, Yasuda Y, Bromme D, DArmiento JM, Ogletree
ML, Tall AR. Spontaneous atherothrombosis and medial degradation in Apoe/,
Npc1/ mice. Circulation 116: 2444 2452, 2007.
1922. Wang YH, Wu MM, Hong CT, Lien LM, Hsieh YC, Tseng HP, Chang SF, Su CL,
Chiou HY, Chen CJ. Effects of arsenic exposure and genetic polymorphisms of p53,
glutathione S-transferase M1, T1, and P1 on the risk of carotid atherosclerosis in
Taiwan. Atherosclerosis 192: 305312, 2007.
1923. Wang YX, Ulu A, Zhang LN, Hammock B. Soluble epoxide hydrolase in atherosclerosis. Curr Atheroscler Rep 12: 174 183, 2010.
1942. Wentzel JJ, Corti R, Fayad ZA, Wisdom P, Macaluso F, Winkelman MO, Fuster V,
Badimon JJ. Does shear stress modulate both plaque progression and regression in
the thoracic aorta? Human study using serial magnetic resonance imaging. J Am Coll
Cardiol 45: 846 854, 2005.
1924. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB,
Fu X, Chung YM, Wu Y, Schauer P, Smith JD, Allayee H, Tang WH, DiDonato JA,
Lusis AJ, Hazen SL. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 472: 57 63, 2011.
1925. Wang Z, Liu B, Wang P, Dong X, Fernandez-Hernando C, Li Z, Hla T, Li Z, Claffey
K, Smith JD, Wu D. Phospholipase C 3 deficiency leads to macrophage hypersensitivity to apoptotic induction and reduction of atherosclerosis in mice. J Clin Invest
118: 195204, 2008.
1537
1928. Warden CH, Hedrick CC, Qiao JH, Castellani LW, Lusis AJ. Atherosclerosis in
transgenic mice overexpressing apolipoprotein A-II. Science 261: 469 472, 1993.
PAUL N. HOPKINS
1945. West AP, Brodsky IE, Rahner C, Woo DK, Erdjument-Bromage H, Tempst P, Walsh
MC, Choi Y, Shadel GS, Ghosh S. TLR signalling augments macrophage bactericidal
activity through mitochondrial ROS. Nature 472: 476 480, 2011.
1965. Willner EL, Tow B, Buhman KK, Wilson M, Sanan DA, Rudel LL, Farese RV Jr.
Deficiency of acyl CoA:cholesterol acyltransferase 2 prevents atherosclerosis in
apolipoprotein E-deficient mice. Proc Natl Acad Sci USA 100: 12621267, 2003.
1946. Westerterp M, Koetsveld J, Yu S, Han S, Li R, Goldberg IJ, Welch CL, Tall AR.
Increased atherosclerosis in mice with vascular ATP-binding cassette transporter G1
deficiency brief report. Arterioscler Thromb Vasc Biol 30: 21032105, 2010.
1966. Wilson SJ, Cavanagh CC, Lesher AM, Frey AJ, Russell SE, Smyth EM. Activationdependent stabilization of the human thromboxane receptor: role of reactive oxygen species. J Lipid Res 50: 10471056, 2009.
1947. Westlake SL, Colebatch AN, Baird J, Kiely P, Quinn M, Choy E, Ostor AJ, Edwards
CJ. The effect of methotrexate on cardiovascular disease in patients with rheumatoid
arthritis: a systematic literature review. Rheumatology 49: 295307, 2010.
1948. Westra J, de Groot L, Plaxton SL, Brouwer E, Posthumus MD, Kallenberg CG, Bijl M.
Angiopoietin-2 is highly correlated with inflammation and disease activity in recentonset rheumatoid arthritis and could be predictive for cardiovascular disease. Rheumatology 50: 665 673, 2011.
1968. Wohrle FU, Daly RJ, Brummer T. Function, regulation and pathological roles of the
Gab/DOS docking proteins. Cell Commun Signal 7: 22, 2009.
1949. Westrick RJ, Bodary PF, Xu Z, Shen YC, Broze GJ, Eitzman DT. Deficiency of tissue
factor pathway inhibitor promotes atherosclerosis and thrombosis in mice. Circulation 103: 3044 3046, 2001.
1950. Wettschureck N, Offermanns S. Mammalian G proteins and their cell type specific
functions. Physiol Rev 85: 1159 1204, 2005.
1951. Wetzelberger K, Baba SP, Thirunavukkarasu M, Ho YS, Maulik N, Barski OA, Conklin DJ, Bhatnagar A. Postischemic deactivation of cardiac aldose reductase: role of
glutathione S-transferase P and glutaredoxin in regeneration of reduced thiols from
sulfenic acids. J Biol Chem 285: 2613526148, 2010.
1953. Whetzel AM, Bolick DT, Srinivasan S, Macdonald TL, Morris MA, Ley K, Hedrick
CC. Sphingosine-1 phosphate prevents monocyte/endothelial interactions in type 1
diabetic NOD mice through activation of the S1P1 receptor. Circ Res 99: 731739,
2006.
1954. White CR, Frangos JA. The shear stress of it all: the cell membrane and mechanochemical transduction. Philos Trans R Soc Lond B Biol Sci 362: 1459 1467, 2007.
1955. Whitman SC, Rateri DL, Szilvassy SJ, Yokoyama W, Daugherty A. Depletion of
natural killer cell function decreases atherosclerosis in low-density lipoprotein receptor null mice. Arterioscler Thromb Vasc Biol 24: 1049 1054, 2004.
1956. Whitsett J, Picklo MJ Sr, Vasquez-Vivar J. 4-Hydroxy-2-nonenal increases superoxide
anion radical in endothelial cells via stimulated GTP cyclohydrolase proteasomal
degradation. Arterioscler Thromb Vasc Biol 27: 2340 2347, 2007.
1957. Wick G, Knoflach M, Xu Q. Autoimmune and inflammatory mechanisms in atherosclerosis. Annu Rev Immunol 22: 361 403, 2004.
1958. Wicovsky A, Henkler F, Salzmann S, Scheurich P, Kneitz C, Wajant H. Tumor
necrosis factor receptor-associated factor-1 enhances proinflammatory TNF receptor-2 signaling and modifies TNFR1-TNFR2 cooperation. Oncogene 28: 1769 1781,
2009.
1959. Widlansky ME, Gutterman DD. Regulation of endothelial function by mitochondrial
reactive oxygen species. Antioxid Redox Signal 2011.
1960. Wigren M, Kolbus D, Duner P, Ljungcrantz I, Soderberg I, Bjorkbacka H, Fredrikson
GN, Nilsson J. Evidence for a role of regulatory T cells in mediating the atheroprotective effect of apolipoprotein B peptide vaccine. J Intern Med 269: 546 556, 2011.
1961. Wilbert-Lampen U, Leistner D, Greven S, Pohl T, Sper S, Volker C, Guthlin D, Plasse
A, Knez A, Kuchenhoff H, Steinbeck G. Cardiovascular events during World Cup
soccer. N Engl J Med 358: 475 483, 2008.
1962. Williams KJ, Feig JE, Fisher EA. Rapid regression of atherosclerosis: insights from the
clinical and experimental literature. Nat Clin Pract Cardiovasc Med 5: 91102, 2008.
1963. Williams MR, Sakurai Y, Zughaier SM, Eskin SG, McIntire LV. Transmigration across
activated endothelium induces transcriptional changes, inhibits apoptosis, and decreases antimicrobial protein expression in human monocytes. J Leukoc Biol 2009.
1964. Willis MS, Townley-Tilson WH, Kang EY, Homeister JW, Patterson C. Sent to destroy: the ubiquitin proteasome system regulates cell signaling and protein quality
control in cardiovascular development and disease. Circ Res 106: 463 478, 2010.
1538
1952. Weyrich AS, Denis MM, Schwertz H, Tolley ND, Foulks J, Spencer E, Kraiss LW,
Albertine KH, McIntyre TM, Zimmerman GA. mTOR-dependent synthesis of Bcl-3
controls the retraction of fibrin clots by activated human platelets. Blood 109: 1975
1983, 2007.
1969. Woldt E, Terrand J, Mlih M, Matz RL, Bruban V, Coudane F, Foppolo S, El Asmar Z,
Chollet ME, Ninio E, Bednarczyk A, Thierse D, Schaeffer C, Van Dorsselaer A,
Boudier C, Wahli W, Chambon P, Metzger D, Herz J, Boucher P. The nuclear
hormone receptor PPARgamma counteracts vascular calcification by inhibiting
Wnt5a signalling in vascular smooth muscle cells. Nature Commun 3: 1077, 2012.
1992. Xiao Q, Danton MJ, Witte DP, Kowala MC, Valentine MT, Degen JL. Fibrinogen
deficiency is compatible with the development of atherosclerosis in mice. J Clin Invest
101: 1184 1194, 1998.
1993. Xiao Q, Danton MJS, Witte DP, Kowala MC, Valentine MT, Bugge TH, Degen JL.
Plasminogen deficiency accelerates vessel wall disease in mice predisposed to?atherosclerosis. Proc Natl Acad Sci USA 94: 1033510340, 1997.
2011. Yamawaki H, Lehoux S, Berk BC. Chronic physiological shear stress inhibits tumor
necrosis factor-induced proinflammatory responses in rabbit aorta perfused ex vivo.
Circulation 108: 1619 1625, 2003.
1994. Xiao Z, Wang T, Qin H, Huang C, Feng Y, Xia Y. Endoplasmic reticulum Ca2
release modulates endothelial nitric-oxide synthase via extracellular signal-regulated
kinase (ERK1/2)-mediated serine 635 phosphorylation. J Biol Chem 286: 20100
20108, 2011.
2012. Yamawaki H, Pan S, Lee RT, Berk BC. Fluid shear stress inhibits vascular inflammation by decreasing thioredoxin-interacting protein in endothelial cells. J Clin Invest
115: 733738, 2005.
1995. Xie L, Qiao X, Wu Y, Tang J. beta-Arrestin1 mediates the endocytosis and functions
of macrophage migration inhibitory factor. PLoS ONE 6: e16428, 2011.
1996. Xiong Y, Huo Y, Chen C, Zeng H, Lu X, Wei C, Ruan C, Zhang X, Hu Z, Shibuya M,
Luo J. Vascular endothelial growth factor (VEGF) receptor-2 tyrosine 1175 signaling
controls VEGF-induced von Willebrand factor release from endothelial cells via
phospholipase C-1- and protein kinase A-dependent pathways. J Biol Chem 284:
2321723224, 2009.
1997. Xiong Y, Uys JD, Tew KD, Townsend DM. S-glutathionylation: from molecular
mechanisms to health outcomes. Antioxid Redox Signal 2011.
1998. Xu G, Tan X, Wang H, Sun W, Shi Y, Burlingame S, Gu X, Cao G, Zhang T, Qin J,
Yang J. Ubiquitin-specific peptidase 21 inhibits tumor necrosis factor alpha-induced
nuclear factor kappaB activation via binding to and deubiquitinating receptor-interacting protein 1. J Biol Chem 285: 969 978, 2010.
1999. Xu H, Zhu J, Smith S, Foldi J, Zhao B, Chung AY, Outtz H, Kitajewski J, Shi C, Weber
S, Saftig P, Li Y, Ozato K, Blobel CP, Ivashkiv LB, Hu X. Notch-RBP-J signaling
regulates the transcription factor IRF8 to promote inflammatory macrophage polarization. Nat Immunol 13: 642 650, 2012.
2000. Xu J, Zou MH. Molecular insights and therapeutic targets for diabetic endothelial
dysfunction. Circulation 120: 1266 1286, 2009.
2013. Yan C, Huang A, Kaley G, Sun D. Chronic high blood flow potentiates shear stressinduced release of NO in arteries of aged rats. Am J Physiol Heart Circ Physiol 293:
H3105H3110, 2007.
2014. Yan C, Huang A, Wu Z, Kaminski PM, Wolin MS, Hintze TH, Kaley G, Sun D.
Increased superoxide leads to decreased flow-induced dilation in resistance arteries
of Mn-SOD-deficient mice. Am J Physiol Heart Circ Physiol 288: H2225H2231, 2005.
2015. Yan D, Jauhiainen M, Hildebrand RB, Willems van Dijk K, Van Berkel TJ, Ehnholm C,
Van Eck M, Olkkonen VM. Expression of human OSBP-related protein 1L in macrophages enhances atherosclerotic lesion development in LDL receptor-deficient
mice. Arterioscler Thromb Vasc Biol 27: 1618 1624, 2007.
2016. Yancey PG, Blakemore J, Ding L, Fan D, Overton CD, Zhang Y, Linton MF, Fazio S.
Macrophage LRP-1 controls plaque cellularity by regulating efferocytosis and Akt
activation. Arterioscler Thromb Vasc Biol 30: 787795, 2010.
2017. Yancey PG, Ding Y, Fan D, Blakemore JL, Zhang Y, Ding L, Zhang J, Linton MF, Fazio
S. Low-density lipoprotein receptor-related protein 1 prevents early atherosclerosis
by limiting lesional apoptosis and inflammatory Ly-6Chigh monocytosis: evidence
that the effects are not apolipoprotein E dependent. Circulation 124: 454 464,
2011.
2018. Yang D, Chen H, Koupenova M, Carroll SH, Eliades A, Freedman JE, Toselli P, Ravid
K. A new role for the A2b adenosine receptor in regulating platelet function. J
Thromb Haemost 8: 817 827, 2010.
1539
1989. Xia M, Guerra N, Sukhova GK, Yang K, Miller CK, Shi GP, Raulet DH, Xiong N.
Immune activation resulting from NKG2D/ligand interaction promotes atherosclerosis. Circulation 124: 29332943, 2011.
2001. Xu S, Bayat H, Hou X, Jiang B. Ribosomal S6 kinase-1 modulates interleukin-1induced persistent activation of NF-B through phosphorylation of IB. Am J Physiol
Cell Physiol 291: C1336 C1345, 2006.
PAUL N. HOPKINS
2019. Yang D, Koupenova M, McCrann DJ, Kopeikina KJ, Kagan HM, Schreiber BM, Ravid
K. The A2b adenosine receptor protects against vascular injury. Proc Natl Acad Sci
USA 105: 792796, 2008.
2037. Yi X, Xu L, Kim K, Kim HS, Maeda N. Genetic reduction of lipoic acid synthase
expression modestly increases atherosclerosis in male, but not in female, apolipoprotein E-deficient mice. Atherosclerosis 211: 424 430, 2010.
2038. Yin K, Deng X, Mo ZC, Zhao GJ, Jiang J, Cui LB, Tan CZ, Wen GB, Fu Y, Tang CK.
Tristetraprolin-dependent posttranscriptional regulation of inflammatory cytokine
mRNA expression by apolipoprotein A-I: role of ATP-binding membrane cassette
transporter A1 and signal transducer and activator of transcription 3. J Biol Chem 286:
13834 13845, 2011.
2021. Yang H, Roberts LJ, Shi MJ, Zhou LC, Ballard BR, Richardson A, Guo ZM. Retardation
of atherosclerosis by overexpression of catalase or both Cu/Zn-superoxide dismutase and catalase in mice lacking apolipoprotein E. Circ Res 95: 10751081, 2004.
2022. Yang H, Zhou L, Wang Z, Roberts LJ, 2nd Lin X, Zhao Y, Guo Z. Overexpression of
antioxidant enzymes in ApoE-deficient mice suppresses benzo(a)pyrene-accelerated atherosclerosis. Atherosclerosis 207: 5158, 2009.
2023. Yang L, Guan H, He J, Zeng L, Yuan Z, Xu M, Zhang W, Wu X, Guan J. VEGF
increases the proliferative capacity and eNOS/NO levels of endothelial progenitor
cells through the calcineurin/NFAT signalling pathway. Cell Biol Int 36: 2127, 2012.
2024. Yang SK, Wang YC, Chao CC, Chuang YJ, Lan CY, Chen BS. Dynamic cross-talk
analysis among TNF-R, TLR-4 and IL-1R signalings in TNFalpha-induced inflammatory responses. BMC Med Genomics 3: 19, 2010.
2025. Yang WS, Seo JW, Han NJ, Choi J, Lee KU, Ahn H, Lee SK, Park SK. High glucoseinduced NF-kappaB activation occurs via tyrosine phosphorylation of IkappaBalpha
in human glomerular endothelial cells: involvement of Syk tyrosine kinase. Am J
Physiol Renal Physiol 294: F1065F1075, 2008.
2027. Yang YM, Huang A, Kaley G, Sun D. eNOS uncoupling and endothelial dysfunction in
aged vessels. Am J Physiol Heart Circ Physiol 297: H1829 H1836, 2009.
2028. Yao J, Kim TW, Qin J, Jiang Z, Qian Y, Xiao H, Lu Y, Qian W, Gulen MF, Sizemore N,
DiDonato J, Sato S, Akira S, Su B, Li X. Interleukin-1 (IL-1)-induced TAK1-dependent
versus MEKK3-dependent NFB activation pathways bifurcate at IL-1 receptorassociated kinase modification. J Biol Chem 282: 6075 6089, 2007.
2029. Yao Y, Bennett BJ, Wang X, Rosenfeld ME, Giachelli C, Lusis AJ, Bostrom KI. Inhibition of bone morphogenetic proteins protects against atherosclerosis and vascular
calcification. Circ Res 107: 485 494, 2010.
2030. Yatera Y, Shibata K, Furuno Y, Sabanai K, Morisada N, Nakata S, Morishita T,
Toyohira Y, Wang KY, Tanimoto A, Sasaguri Y, Tasaki H, Nakashima Y, Shimokawa
H, Yanagihara N, Otsuji Y, Tsutsui M. Severe dyslipidaemia, atherosclerosis, and
sudden cardiac death in mice lacking all NO synthases fed a high-fat diet. Cardiovasc
Res 87: 675 682, 2010.
2040. Yin MJ, Yamamoto Y, Gaynor RB. The anti-inflammatory agents aspirin and salicylate
inhibit the activity of IB kinase-beta. Nature 396: 77 80, 1998.
2041. Yin W, Romeo S, Chang S, Grishin NV, Hobbs HH, Cohen JC. Genetic variation in
ANGPTL4 provides insights into protein processing and function. J Biol Chem 284:
1321313222, 2009.
2042. Ying Z, Giachini FR, Tostes RC, Webb RC. PYK2/PDZ-RhoGEF links Ca2 signaling
to RhoA. Arterioscler Thromb Vasc Biol 29: 16571663, 2009.
2043. Yoon SI, Hong M, Wilson IA. An unusual dimeric structure and assembly for TLR4
regulator RP105-MD-1. Nat Struct Mol Biol 18: 1028 1035, 2011.
2044. Yoon Y, Galloway CA, Jhun BS, Yu T. Mitochondrial dynamics in diabetes. Antioxid
Redox Signal 14: 439 457, 2011.
2045. Yoshisue H, Suzuki K, Kawabata A, Ohya T, Zhao H, Sakurada K, Taba Y, Sasaguri T,
Sakai N, Yamashita S, Matsuzawa Y, Nojima H. Large scale isolation of nonuniform
shear stress-responsive genes from cultured human endothelial cells through the
preparation of a subtracted cDNA library. Atherosclerosis 162: 323334, 2002.
2046. Young A, Wu W, Sun W, Benjamin Larman H, Wang N, Li YS, Shyy JY, Chien S,
Garcia-Cardena G. Flow activation of AMP-activated protein kinase in vascular endothelium leads to Kruppel-like factor 2 expression. Arterioscler Thromb Vasc Biol 29:
19021908, 2009.
2047. Yu T, Jhun BS, Yoon Y. High-glucose stimulation increases reactive oxygen species
production through the calcium and mitogen-activated protein kinase-mediated activation of mitochondrial fission. Antioxid Redox Signal 14: 425 437, 2011.
2048. Yu T, Robotham JL, Yoon Y. Increased production of reactive oxygen species in
hyperglycemic conditions requires dynamic change of mitochondrial morphology.
Proc Natl Acad Sci USA 103: 26532658, 2006.
2049. Yu Y, Lucitt MB, Stubbe J, Cheng Y, Friis UG, Hansen PB, Jensen BL, Smyth EM,
FitzGerald GA. Prostaglandin F2alpha elevates blood pressure and promotes atherosclerosis. Proc Natl Acad Sci USA 106: 79857990, 2009.
2050. Yu Z, Crichton I, Tang SY, Hui Y, Ricciotti E, Levin MD, Lawson JA, Pure E, FitzGerald GA. Disruption of the 5-lipoxygenase pathway attenuates atherogenesis consequent to COX-2 deletion in mice. Proc Natl Acad Sci USA 109: 6727 6732, 2012.
2051. Yuan M, Konstantopoulos N, Lee J, Hansen L, Li ZW, Karin M, Shoelson SE. Reversal
of obesity- and diet-induced insulin resistance with salicylates or targeted disruption
of Ikkbeta. Science 293: 16731677, 2001.
2032. Ye Z, Liu EH, Higgins JP, Keavney BD, Lowe GD, Collins R, Danesh J. Seven haemostatic gene polymorphisms in coronary disease: meta-analysis of 66,155 cases and
91,307 controls. Lancet 367: 651 658, 2006.
2052. Yun S, Leung VW, Botto M, Boyle JJ, Haskard DO. Brief report: accelerated atherosclerosis in low-density lipoprotein receptor-deficient mice lacking the membranebound complement regulator CD59. Arterioscler Thromb Vasc Biol 28: 1714 1716,
2008.
2033. Yeh JC, Otte LA, Frangos JA. Regulation of G protein-coupled receptor activities by
the platelet-endothelial cell adhesion molecule, PECAM-1. Biochemistry 47: 9029
9039, 2008.
2053. Yvan-Charvet L, Pagler T, Gautier EL, Avagyan S, Siry RL, Han S, Welch CL, Wang N,
Randolph GJ, Snoeck HW, Tall AR. ATP-binding cassette transporters and HDL
suppress hematopoietic stem cell proliferation. Science 328: 1689 1693, 2010.
2034. Yesilaltay A, Daniels K, Pal R, Krieger M, Kocher O. Loss of PDZK1 causes coronary
artery occlusion and myocardial infarction in Paigen diet-fed apolipoprotein E deficient mice. PLoS ONE 4: e8103, 2009.
2035. Yet SF, Layne MD, Liu X, Chen YH, Ith B, Sibinga NE, Perrella MA. Absence of heme
oxygenase-1 exacerbates atherosclerotic lesion formation and vascular remodeling.
FASEB J 17: 1759 1761, 2003.
2055. Zaidel-Bar R, Geiger B. The switchable integrin adhesome. J Cell Sci 123: 13851388,
2010.
1540
2026. Yang X, Peterson L, Thieringer R, Deignan JL, Wang X, Zhu J, Wang S, Zhong H,
Stepaniants S, Beaulaurier J, Wang IM, Rosa R, Cumiskey AM, Luo JM, Luo Q,
Shah K, Xiao J, Nickle D, Plump A, Schadt EE, Lusis AJ, Lum PY. Identification and
validation of genes affecting aortic lesions in mice. J Clin Invest 120: 2414 2422,
2010.
2039. Yin K, Liao DF, Tang CK. ATP-binding membrane cassette transporter A1 (ABCA1):
a possible link between inflammation and reverse cholesterol transport. Mol Med 16:
438 449, 2010.
2065. Zernecke A, Weber C. Chemokines in the vascular inflammatory response of atherosclerosis. Cardiovasc Res 86: 192201, 2010.
2066. Zhang C, Zhao YX, Zhang YH, Zhu L, Deng BP, Zhou ZL, Li SY, Lu XT, Song LL, Lei
XM, Tang WB, Wang N, Pan CM, Song HD, Liu CX, Dong B, Zhang Y, Cao Y.
Angiotensin-converting enzyme 2 attenuates atherosclerotic lesions by targeting
vascular cells. Proc Natl Acad Sci USA 107: 15886 15891, 2010.
2067. Zhang D, Jiang X, Fang P, Yan Y, Song J, Gupta S, Schafer AI, Durante W, Kruger WD,
Yang X, Wang H. Hyperhomocysteinemia promotes inflammatory monocyte generation and accelerates atherosclerosis in transgenic cystathionine beta-synthasedeficient mice. Circulation 120: 18931902, 2009.
2068. Zhang DX, Gutterman DD. Mitochondrial reactive oxygen species-mediated signaling in endothelial cells. Am J Physiol Heart Circ Physiol 292: H2023H2031, 2007.
2069. Zhang G, Marshall AL, Thomas AL, Kernan KA, Su Y, Leboeuf RC, Dong XR, Tchao
BN. In vivo knockdown of nicotinic acetylcholine receptor alpha1 diminishes aortic
atherosclerosis. Atherosclerosis 215: 34 42, 2011.
2070. Zhang H, Luo Y, Zhang W, He Y, Dai S, Zhang R, Huang Y, Bernatchez P, Giordano
FJ, Shadel G, Sessa WC, Min W. Endothelial-specific expression of mitochondrial
thioredoxin improves endothelial cell function and reduces atherosclerotic lesions.
Am J Pathol 170: 1108 1120, 2007.
2071. Zhang H, Park Y, Wu J, Chen X, Lee S, Yang J, Dellsperger KC, Zhang C. Role of
TNF-alpha in vascular dysfunction. Clin Sci 116: 219 230, 2009.
2072. Zhang H, Sunnarborg SW, McNaughton KK, Johns TG, Lee DC, Faber JE. Heparinbinding epidermal growth factor-like growth factor signaling in flow-induced arterial
remodeling. Circ Res 102: 12751285, 2008.
2073. Zhang J, Fukuhara S, Sako K, Takenouchi T, Kitani H, Kume T, Koh GY, Mochizuki N.
Angiopoietin-1/Tie2 signal augments basal Notch signal controlling vascular quiescence by inducing delta-like 4 expression through AKT-mediated activation of betacatenin. J Biol Chem 286: 8055 8066, 2011.
2074. Zhang J, Xie Z, Dong Y, Wang S, Liu C, Zou MH. Identification of nitric oxide as an
endogenous activator of the AMP-activated proteinkinase in vascular endothelial
cells. J Biol Chem 283: 2745227461, 2008.
2075. Zhang L, Blackwell K, Thomas GS, Sun S, Yeh WC, Habelhah H. TRAF2 suppresses
basal IKK activity in resting cells and TNF can activate IKK in TRAF2 and TRAF5
double knockout cells. J Mol Biol 389: 495510, 2009.
1541
2076. Zhang L, Connelly JJ, Peppel K, Brian L, Shah SH, Nelson S, Crosslin DR, Wang T,
Allen A, Kraus WE, Gregory SG, Hauser ER, Freedman NJ. Aging-related atherosclerosis is exacerbated by arterial expression of tumor necrosis factor receptor-1:
evidence from mouse models and human association studies. Hum Mol Genet 19:
2754 2766, 2010.
PAUL N. HOPKINS
2093. Zhao M, Wigren M, Duner P, Kolbus D, Olofsson KE, Bjorkbacka H, Nilsson J,
Fredrikson GN. FcgammaRIIB inhibits the development of atherosclerosis in lowdensity lipoprotein receptor-deficient mice. J Immunol 184: 22532260, 2010.
2094. Zhao R, Ma X, Xie X, Shen GX. Involvement of NADPH oxidase in oxidized LDLinduced upregulation of heat shock factor-1 and plasminogen activator inhibitor-1 in
vascular endothelial cells. Am J Physiol Endocrinol Metab 297: E104 E111, 2009.
2095. Zhao Y, Chen X, Yang H, Zhou L, Okoro EU, Guo Z. A novel function of apolipoprotein E: upregulation of ATP-binding cassette transporter A1 expression. PLoS
ONE 6: e21453, 2011.
2096. Zhao Y, Howatt DA, Gizard F, Nomiyama T, Findeisen HM, Heywood EB, Jones KL,
Conneely OM, Daugherty A, Bruemmer D. Deficiency of the NR4A orphan nuclear
receptor NOR1 decreases monocyte adhesion and atherosclerosis. Circ Res 107:
501511, 2010.
2097. Zheng C, Kabaleeswaran V, Wang Y, Cheng G, Wu H. Crystal structures of the
TRAF2: cIAP2 and the TRAF1: TRAF2: cIAP2 complexes: affinity, specificity, and
regulation. Mol Cell 38: 101113, 2010.
2098. Zheng Z, Chen H, Li J, Li T, Zheng B, Zheng Y, Jin H, He Y, Gu Q, Xu X. Sirtuin
1-mediated cellular metabolic memory of high glucose via the LKB1/AMPK/ROS
pathway and therapeutic effects of metformin. Diabetes 61: 217228, 2012.
2099. Zhong J, Gavrilescu LC, Molnar A, Murray L, Garafalo S, Kehrl JH, Simon AR, Van Etten
RA, Kyriakis JM. GCK is essential to systemic inflammation and pattern recognition
receptor signaling to JNK and p38. Proc Natl Acad Sci USA 106: 4372 4377, 2009.
2100. Zhou B, Margariti A, Zeng L, Xu Q. Role of histone deacetylases in vascular cell
homeostasis and arteriosclerosis. Cardiovasc Res 2011.
1542
2111. Zhu M, Zhang QJ, Wang L, Li H, Liu ZP. FoxO4 inhibits atherosclerosis through its
function in bone marrow derived cells. Atherosclerosis 219: 492 498, 2011.
2112. Zhu SN, Chen M, Jongstra-Bilen J, Cybulsky MI. GM-CSF regulates intimal cell
proliferation in nascent atherosclerotic lesions. J Exp Med 206: 21412149, 2009.
2113. Zhu W, Saddar S, Seetharam D, Chambliss KL, Longoria C, Silver DL, Yuhanna IS,
Shaul PW, Mineo C. The scavenger receptor class B type I adaptor protein PDZK1
maintains endothelial monolayer integrity. Circ Res 102: 480 487, 2008.
2114. Zhu Y, Lin JHC, Liao HL, Friedli O, Verna L, Marten NW, Straus DS, Stemerman MB.
LDL induces transcription factor activator protein-1 in human endothelial cells.
Arterioscler Thromb Vasc Biol 18: 473 480, 1998.
2115. Zhuang J, Peng W, Li H, Wang W, Wei Y, Li W, Xu Y. Methylation of p15INK4b and
expression of ANRIL on chromosome 9p21 are associated with coronary artery
disease. PLoS ONE 7: e47193, 2012.
2116. Zia AA, Kamalov G, Newman KP, McGee JE, Bhattacharya SK, Ahokas RA, Sun Y,
Gerling IC, Weber KT. From aldosteronism to oxidative stress: the role of excessive
intracellular calcium accumulation. Hypertens Res 33: 10911101, 2010.
2117. Zilversmit DB. Atherogenesis: a postprandial phenomenon. Circulation 60: 473
485, 1979.
2118. Zilversmit DB. A proposal linking atherogenesis to the interaction of endothelial
lipoprotein lipase with triglyceride-rich lipoproteins. Annu Rev Nutr 1: 95121, 1981.
2119. Zirlik A, Maier C, Gerdes N, MacFarlane L, Soosairajah J, Bavendiek U, Ahrens I,
Ernst S, Bassler N, Missiou A, Patko Z, Aikawa M, Schonbeck U, Bode C, Libby P,
Peter K. CD40 ligand mediates inflammation independently of CD40 by interaction
with Mac-1. Circulation 115: 15711580, 2007.
2120. Zitvogel L, Kepp O, Kroemer G. Decoding cell death signals in inflammation and
immunity. Cell 140: 798 804, 2010.
2121. Zoll J, Fontaine V, Gourdy P, Barateau V, Vilar J, Leroyer A, Lopes-Kam I, Mallat Z,
Arnal JF, Henry P, Tobelem G, Tedgui A. Role of human smooth muscle cell progenitors in atherosclerotic plaque development and composition. Cardiovasc Res 77:
471 480, 2008.
2122. Zonneveld-Huijssoon E, van Wijk F, Roord S, Delemarre E, Meerding J, de Jager W,
Klein M, Raz E, Albani S, Kuis W, Boes M, Prakken BJ. TLR9 agonist CpG enhances
protective nasal HSP60 peptide vaccine efficacy in experimental autoimmune arthritis. Ann Rheum Dis 2012.
2123. Zou MH, Shi C, Cohen RA. High glucose via peroxynitrite causes tyrosine nitration
and inactivation of prostacyclin synthase that is associated with thromboxane/prostaglandin H(2) receptor-mediated apoptosis and adhesion molecule expression in
cultured human aortic endothelial cells. Diabetes 51: 198 203, 2002.
2124. Zschenker O, Illies T, Ameis D. Overexpression of lysosomal acid lipase and other
proteins in atherosclerosis. J Biochem 140: 2338, 2006.
2125. Zuo S, Xue Y, Tang S, Yao J, Du R, Yang P, Chen X. 14-3-3 epsilon dynamically
interacts with key components of mitogen-activated protein kinase signal module for
selective modulation of the TNF-alpha-induced time course-dependent NF-kappaB
activity. J Proteome Res 9: 34653478, 2010.
2101. Zhou C, King N, Chen KY, Breslow JL. Activation of PXR induces hypercholesterolemia in wild-type and accelerates atherosclerosis in apoE deficient mice. J Lipid Res
50: 2004 2013, 2009.
Physiological Reviews provides state of the art coverage of timely issues in the physiological and biomedical sciences. It is
published quarterly in January, April, July, and October by the American Physiological Society, 9650 Rockville Pike, Bethesda
MD 20814-3991. Copyright 2013 by the American Physiological Society. ISSN: 0031-9333, ESSN: 1522-1210. Visit our
website at http://www.the-aps.org/.
Physiological Reviews provides state of the art coverage of timely issues in the physiological and biomedical sciences. It is
published quarterly in January, April, July, and October by the American Physiological Society, 9650 Rockville Pike, Bethesda
MD 20814-3991. Copyright 2013 by the American Physiological Society. ISSN: 0031-9333, ESSN: 1522-1210. Visit our
website at http://www.the-aps.org/.