Hypercholesterolemia is a risk factor for atherothrombotic disease, largely attributed to its impact on atherosclerotic lesional cells such as macrophages. Platelets are involved in immunity and inflammation and impact atherogenesis, primarily by modulating immune and inflammatory effector cells. There is evidence that hypercholesterolemia increases the risk of atherosclerosis and thrombosis by modulating platelet biogenesis and activity. This review highlights recent findings on the impact of aberrant cholesterol metabolism on platelet biogenesis and activity and their relevance in atherosclerosis and thrombosis.

Platelets are best known for their roles in hemostasis and thrombosis. However, there is an increasing appreciation of the critical roles of platelets in immunity and inflammation in both health and disease.1-3  Atherosclerosis is a lipid-driven chronic inflammatory disease that involves localized recruitment of myeloid and immune cells (including neutrophils, monocytes, and lymphocytes) to large and medium-sized arteries.4,5  Platelets appear to play a key role in recruitment of these inflammatory effector cells.6  Activated platelets interact with endothelial cells of inflamed or atherosclerotic arteries and deposit platelet-derived cytokines such as chemokine (C-C motif) ligand 5 (CCL5) or chemokine (C-X-C motif) ligand 4 (CXCL4) onto the surface of endothelial cells, facilitating recruitment of leukocytes into the lesions.7  Genetic deficiency of CCL5 and CXCL4 or pharmacologic disruption of the functional heteromerization between CCL5 and CXCL4 decreases atherogenesis in animal models.7  Activated platelets form aggregates with neutrophils and monocytes, and the ensuing cross talk between platelets and leukocytes also plays a key role in inflammatory cytokine production, leukotriene biosynthesis, and reactive oxygen species production,8,9  with proinflammatory consequences in the vasculature. Platelet-leukocyte aggregates (PLAs) are an independent risk factor for atherothrombotic disease10-12  and promote atherogenesis in mouse models6,13  (Figure 1). An elegant recent study from Sreeramkumar et al demonstrated that neutrophils bound to endothelium scan the bloodstream for activated platelets, leading to neutrophil polarization and formation of PLAs and facilitating neutrophil migration into inflamed blood vessels.8  This interaction of platelets with leukocytes is initiated by the binding of platelet P-selectin to P-selectin ligand, which localizes to the uropod (tail) of neutrophils as they bind to endothelium. Disruption of platelet/leukocyte interactions via genetic deficiency of P-selectin13  or by anti-P-selectin–blocking antibodies decreases leukocyte recruitment and atherogenesis.14  Consistently, infusion of activated wild-type, but not P-selectin–deficient platelets, into Apoe−/− mice increases atherosclerosis.6  In addition to modulating migration and recruitment of leukocytes, platelet/leukocyte interactions could potentially impact atherosclerosis and atherothrombosis by modulating other activities of leukocytes. Neutrophil extracellular traps (NETs) generated during NETosis promote venous and arterial thrombosis in animal models15,16  and recent evidence suggests a role of NETs in atherogenesis and atherothrombotic disorders as well.17-21  Interestingly, activated platelets interact with neutrophils and promote NETosis, which appears to require P-selectin/P-selectin glycoprotein ligand 1 interactions.22  Thus, activated platelets could potentially promote atherosclerosis and thrombogenesis by facilitating NET generation. Other roles of platelets in atherosclerosis have been reviewed and discussed elsewhere.2,23 

Figure 1

Hypercholesterolemia is a risk factor for atherothrombotic disease by promoting platelet production and activation. Hypercholesterolemia promotes megakaryopoiesis, platelet (PLT) biogenesis, and myelopoiesis, leading to leukocytosis. Hypercholesterolemia also increases platelet activation, likely by elevating platelet production and by direct impact on platelets. Activated platelets form PLAs, which are further increased in leukocytosis. PLA is an independent risk factor for atherothrombotic disease. GMP, guanosine monophosphate; HSC, hematopoietic stem cell; MkP, megakaryocyte progenitor cell.

Figure 1

Hypercholesterolemia is a risk factor for atherothrombotic disease by promoting platelet production and activation. Hypercholesterolemia promotes megakaryopoiesis, platelet (PLT) biogenesis, and myelopoiesis, leading to leukocytosis. Hypercholesterolemia also increases platelet activation, likely by elevating platelet production and by direct impact on platelets. Activated platelets form PLAs, which are further increased in leukocytosis. PLA is an independent risk factor for atherothrombotic disease. GMP, guanosine monophosphate; HSC, hematopoietic stem cell; MkP, megakaryocyte progenitor cell.

Close modal

Hyperlipidemia as exemplified by familial hypercholesterolemia is associated with increased platelet activation and an underlying procoagulant state.24-26  Hyperlipidemia primes platelets and increases platelet activation in response to various agonists.24,27  Plasma cholesterol levels appear to have a critical role in modulating platelet activity because hypercholesterolemia increases platelet activation more potently than hypertriglyceridemia.24,27  Hyperlipidemia increases platelet activation likely via multiple mechanisms.28,29  Oxidized low-density lipoprotein or oxidized phospholipids, which are increased in hyperlipidemia,28,30  serve as ligands of platelet CD36 and activate platelets.28,31  Oxidized lipids also promote formation of procoagulant tissue factor microparticles derived from monocytes.26  In vitro cholesterol loading also increases human platelet activation.32  High-density lipoprotein (HDL) is a cholesterol acceptor and promotes cholesterol efflux.33  HDL has been shown to mediate various antithrombotic effects, although some of the effects are via the impact on the vasculature, such as reducing endothelial cell surface expression of adhesion molecules.34  Infusions of a reconstituted HDL (rHDL) preparation reduced ex vivo platelet activation in diabetic subjects, likely by promoting cholesterol efflux from platelets.35  These findings in humans are consistent with observations in animal studies. A striking example is the markedly increased platelet activation and thrombosis in Scarbi−/− mice.29,36 Scarbi−/− mice have an unusually high plasma ratio of unesterified cholesterol to total cholesterol, reflecting impaired delivery of cholesterol by HDL from plasma and peripheral tissues back to the liver via hepatic scavenger receptor class B type I (SR-BI).37  Although the major function of hepatic SR-BI is to mediate selective uptake of cholesterol and cholesteryl ester from HDL, SR-BI-mediated cellular unesterified cholesterol efflux to HDL has been reported.38,39  The net unesterified cholesterol influx or efflux facilitated by SR-BI likely depends on the type of cells and the relative cholesterol/phospholipid ratio in the cell membrane vs that in HDL.40,41  SR-BI is expressed in platelets, whereas platelet cholesterol overload due to markedly increased unesterified cholesterol content in plasma lipoproteins, but not intrinsic SR-BI deficiency in platelets or other hematopoietic cells, is responsible for the heightened platelet activation in Scarb1−/− mice.29  As a consequence, Scarb1−/− mice developed spontaneous occlusive arterial thrombosis and premature death when introduced into hypercholesterolemic apoE−/− or Ldlr−/− background.42,43  Despite this accumulating evidence indicating a direct effect of cholesterol enrichment on platelet activation, the underlying mechanisms have not been elucidated. Cholesterol accumulation in plasma membranes disturbs membrane structures, particularly the cholesterol-rich specialized microdomains of lipid rafts.44  Enhanced signaling of cell surface receptors located in lipid rafts has been reported in various hematopoietic effector cells in response to membrane cholesterol accumulation.45,46  Thus, plasma membrane cholesterol accumulation in platelets could potentially alter the membrane structure and affect signaling via surface receptors. One study suggests that rHDL infusions suppress platelet activation by reducing lipid raft assembly.35  SR-BI is a receptor for HDL, and there is evidence that HDL suppresses thrombin-induced platelet aggregation by binding to SR-BI and generating inhibitory signals.47  In contrast, rHDL can suppress platelet activation via a mechanism independent of SR-BI,35  likely due to the fact that the cholesterol-free, phospholipid-rich rHDL can promote passive cholesterol efflux independent of transporters such as adenosine triphosphate–binding cassette transporter (ABC)A1, ABCG1, or SR-BI.

Platelets are derived from megakaryocytes, and the latter from MkPs in the bone marrow and spleen.48  Thrombopoietin (TPO) and its cognate receptor MPL proto-oncogene (c-MPL) act as a key growth-factor signaling pathway in megakaryopoiesis and platelet biogenesis.49  Genetic deficiency of TPO or c-MPL causes marked thrombocytopenia,49  whereas increased signaling in this pathway results in elevated platelet production and thrombocytosis.50  Plasma lipid levels have long been linked to platelet biogenesis and/or turnover. An analysis of 2 independent studies involving ∼10 000 participants indicates a positive correlation of non-HDL cholesterol levels with platelet counts,51  consistent with some other studies.52,53  Hyperlipidemia also is associated with shortened platelet survival and increased turnover, particularly in the setting of overt atherosclerosis.54-56  Together, these findings suggest the promotion of platelet production by hypercholesterolemia (Figure 1). TPO and c-MPL function to maintain platelet homeostasis, with TPO/c-MPL signaling in hematopoietic stem cells, MkPs, and megakaryocytes regulating megakaryocyte and platelet production, and megakaryocyte and platelet c-MPL acting as a sink for plasma TPO and mediating its internalization and turnover.49  Recent studies indicate an additional mechanism regulating platelet production in which aged desialylated platelets bind the hepatic Ashwell-Morell receptor and thereby stimulate hepatocyte TPO production.57,58  Increased platelet turnover is often associated with increased platelet biogenesis via elevated TPO. The newly generated platelets are larger, more dense, and RNA rich, and these so-called reticulated platelets are generally more active than the more mature platelets.59  Hypercholesterolemia is positively associated with the mean volume of platelets and ploidy of megakaryocytes in humans,60  suggesting the possibility that hypercholesterolemia primes platelets and increases platelet activity by promoting platelet production. Increased platelet activation is expected to increase the risk of coronary heart disease. Consistently, there is strong evidence that mean platelet volume and counts of reticulated platelets are positively associated with acute coronary syndrome,59  and 1 study shows that baseline platelet count is an independent risk factor for acute coronary syndrome.61  These findings in humans have been recapitulated in animal studies. Unesterified cholesterol accumulation in platelets of Scarbi−/− mice led to increased platelet turnover and clearance from circulation,62  which resulted in increased platelet biogenesis and produced juvenile platelets with greatly increased volume,29,62  likely as a consequence of the feedback regulation. Thus, the highly activated platelets in the circulation in Scarbi−/− mice29  could be the result of the combined effect of increased platelet biogenesis and the direct impact of platelet cholesterol enrichment on platelet activation. Moreover, dietary hypercholesterolemia led to thrombocytosis and leukocytosis as a result of bone marrow hematopoietic progenitor cell mobilization and altered interactions of megakaryocytes with endothelial cells due to disturbed CXCL12 and CXC4 signaling.63  However, the diet used in the study contained a very high cholesterol content, as well as cholate, and was proinflammatory; thus, effects may not be directly attributable to a direct impact of cholesterol in bone marrow progenitor populations. A recent study indicates that damage-associated molecular patterns promote bone marrow hematopoietic progenitor cell mobilization and extramedullary hematopoiesis.45  Nevertheless, another study assessed the impact of hypercholesterolemia induced by a high-fat, high-cholesterol diet in Ldlr−/− mice relative to the chow-fed Ldlr−/− mice on hematopoietic cells and showed an expansion of the pool of bone marrow hematopoietic stem and progenitor cells (HSPCs), in association with increased myelopoiesis.64  This study in younger mice examined platelet counts and found no change. However, we have detected increased platelet counts in ∼12-month-old chow-fed Ldlr−/− mice (∼12% increase; N.W., unpublished observation). Despite these observations, how hypercholesterolemia modulates platelet biogenesis or turnover at the molecular level remains enigmatic.

Cholesterol homeostasis in hematopoietic cells is maintained in part by mechanisms involving ABC transporters and apolipoproteins (such as apolipoprotein E) that promote cellular cholesterol efflux.65  We showed increased myelopoiesis in ABCA1/ABCG1-deficient or apolipoprotein E–deficient mice due to increased cellular cholesterol accumulation in HSPCs as a result of defective cholesterol efflux.46,66  At the molecular level, increased myelopoiesis was attributed to the increased cell surface levels and signaling of common β subunit of interleukin-3 and granulocyte macrophage–colony-stimulating factor receptors.46  Increased myelopoiesis resulted in leukocytosis and accelerated atherosclerosis in these models.46,66  In subsequent studies in hypercholesterolemic Ldlr−/− mice, we found that deficiency of hematopoietic ABCG4, a transporter highly homologous to ABCG1 and actively promoting cholesterol efflux to HDL,67  increased atherosclerosis in association with increased platelet counts but without any change of plasma TPO levels.68  Selective thrombocytosis in bone marrow ABCG4 deficiency in combination with restricted Abcg4 expression in MkPs but with no or low expression in platelets suggested selectively increased megakaryopoiesis and platelet production. Indeed, MkPs and megakaryocytes, but not HSPCs or progenitors of other hematopoietic lineages, were increased in Abcg4−/− mice. As a result, hematopoietic ABCG4 deficiency led to defective cholesterol efflux to HDL and increased free cholesterol accumulation in MkPs (including plasma membranes) in association with increased c-MPL levels on the surface of Abcg4−/− MkPs, increased cell proliferation in response to TPO, and increased megakaryopoiesis and a more pronounced increase in platelet counts in response to TPO injection. The increased cell surface c-MPL levels in Abcg4−/− MkPs were because of blunting of the negative feedback regulation of c-MPL in response to TPO69  and involved a defective activation of Lyn kinase and casitas B-lineage lymphoma (c-CBL) E3 ligase. Lyn kinase, a palmitoylated membrane protein, seems to act as a membrane cholesterol sensor. Increased membrane cholesterol in Abcg4−/− MkPs may increase Lyn association with the membrane and decrease its tyrosine kinase activity in response to TPO,70  causing defective phosphorylation of c-CBL. This disrupts the negative feedback regulation of c-MPL and leads to increased TPO/c-MPL signaling and platelet production (Figure 2). In addition to increased atherosclerosis, Abcg4−/− mice showed accelerated arterial thrombosis in association with increased reticulated platelets, platelet/leukocyte complexes, and platelet-derived microparticles, all with proven proatherosclerotic and prothrombotic properties. These studies link increased platelet production, initiated from aberrant cholesterol metabolism in its lineage progenitor cells, to accelerated atherosclerosis and arterial thrombosis.

Figure 2

Working model. HDL-mediated cholesterol efflux from MkPs via ABCG4 increases activity of the palmitoylated Lyn kinase, c-CBL phosphorylation and activation, and c-MPL degradation, limiting MkP proliferation and platelet production. JAK2, Janus kinase 2; P, phosphate; pY, phosphotyrosine; STAT, signal transducer and activator of transcription.

Figure 2

Working model. HDL-mediated cholesterol efflux from MkPs via ABCG4 increases activity of the palmitoylated Lyn kinase, c-CBL phosphorylation and activation, and c-MPL degradation, limiting MkP proliferation and platelet production. JAK2, Janus kinase 2; P, phosphate; pY, phosphotyrosine; STAT, signal transducer and activator of transcription.

Close modal

Infusion of rHDL reduced MkP proliferation and platelet counts in wild-type mice, but not in Abcg4−/− mice. The therapeutic potential of rHDL infusions in the control of platelet overproduction was exemplified by the finding that in a mouse model of essential thrombocythemia induced by bone marrow cell expression of a mutant form of c-MPL found in human subjects with essential thrombocythemia,71  rHDL reduced the platelet count in mice receiving Abcg4+/+, but not Abcg4−/−, bone marrow cells.68 

Like ABCG4, ABCB6 is highly expressed in MkPs, and hematopoietic ABCB6 deficiency increases atherogenesis in hypercholesterolemic Ldlr−/− mice in association with selectively increased MkPs, megakaryocytes, total and reticulated platelet counts, and platelet activity.72  Unlike ABCG4, ABCB6 is reported to have transporter activity for porphyrin, but not for cholesterol.73  The detailed molecular mechanism linking ABCB6 deficiency to increased MkP proliferation and platelet production is unknown, but increased oxidative stress in Abcb6−/− MkPs has been suggested to contribute to these phenotypes.

In summary, platelets have critical roles in atherosclerosis and atherothrombosis. The accumulation of cholesterol in platelets or their progenitors, reflecting hypercholesterolemia or defective cholesterol efflux pathways, markedly increases platelet biogenesis, turnover, and activity, potentially contributing to atherogenesis and atherothrombosis. Although the mechanistic understanding of these processes is at an early stage, gaining further insight into the regulation of platelet production and activation by cholesterol and other lipids could open a new window on treatments for atherothrombosis.

The authors thank Andrew Murphy, Laurent Yvan-Charvet, and Prabhakara R. Nagareddy for discussion of this manuscript.

This work is supported by National Institutes of Health National Heart, Lung, and Blood Institute grants HL118567 (N.W.) and HL107653 (A.R.T.).

Contribution: N.W. and A.R.T. wrote the manuscript.

Conflict-of-interest-disclosure: The authors declare no competing financial interests.

Correspondence: Nan Wang, Division of Molecular Medicine, Department of Medicine, Columbia University Medical Center, 630 W. 168th St, New York, NY 10032; e-mail: nw30@cumc.columbia.edu.

1
Semple
 
JW
Italiano
 
JE
Freedman
 
J
Platelets and the immune continuum.
Nat Rev Immunol
2011
, vol. 
11
 
4
(pg. 
264
-
274
)
2
Davì
 
G
Patrono
 
C
Platelet activation and atherothrombosis.
N Engl J Med
2007
, vol. 
357
 
24
(pg. 
2482
-
2494
)
3
Engelmann
 
B
Massberg
 
S
Thrombosis as an intravascular effector of innate immunity.
Nat Rev Immunol
2013
, vol. 
13
 
1
(pg. 
34
-
45
)
4
Ross
 
R
Atherosclerosis--an inflammatory disease.
N Engl J Med
1999
, vol. 
340
 
2
(pg. 
115
-
126
)
5
Libby
 
P
Ridker
 
PM
Hansson
 
GK
Progress and challenges in translating the biology of atherosclerosis.
Nature
2011
, vol. 
473
 
7347
(pg. 
317
-
325
)
6
Huo
 
Y
Schober
 
A
Forlow
 
SB
, et al. 
Circulating activated platelets exacerbate atherosclerosis in mice deficient in apolipoprotein E.
Nat Med
2003
, vol. 
9
 
1
(pg. 
61
-
67
)
7
Koenen
 
RR
von Hundelshausen
 
P
Nesmelova
 
IV
, et al. 
Disrupting functional interactions between platelet chemokines inhibits atherosclerosis in hyperlipidemic mice.
Nat Med
2009
, vol. 
15
 
1
(pg. 
97
-
103
)
8
Sreeramkumar
 
V
Adrover
 
JM
Ballesteros
 
I
, et al. 
Neutrophils scan for activated platelets to initiate inflammation.
Science
2014
, vol. 
346
 
6214
(pg. 
1234
-
1238
)
9
Schrottmaier
 
WC
Kral
 
JB
Badrnya
 
S
Assinger
 
A
Aspirin and P2Y12 Inhibitors in platelet-mediated activation of neutrophils and monocytes.
Thromb Haemost
2015
, vol. 
114
 
3
(pg. 
478
-
489
)
10
Ott
 
I
Neumann
 
FJ
Gawaz
 
M
Schmitt
 
M
Schömig
 
A
Increased neutrophil-platelet adhesion in patients with unstable angina.
Circulation
1996
, vol. 
94
 
6
(pg. 
1239
-
1246
)
11
Furman
 
MI
Benoit
 
SE
Barnard
 
MR
, et al. 
Increased platelet reactivity and circulating monocyte-platelet aggregates in patients with stable coronary artery disease.
J Am Coll Cardiol
1998
, vol. 
31
 
2
(pg. 
352
-
358
)
12
Koyama
 
H
Maeno
 
T
Fukumoto
 
S
, et al. 
Platelet P-selectin expression is associated with atherosclerotic wall thickness in carotid artery in humans.
Circulation
2003
, vol. 
108
 
5
(pg. 
524
-
529
)
13
Burger
 
PC
Wagner
 
DD
Platelet P-selectin facilitates atherosclerotic lesion development.
Blood
2003
, vol. 
101
 
7
(pg. 
2661
-
2666
)
14
Massberg
 
S
Brand
 
K
Grüner
 
S
, et al. 
A critical role of platelet adhesion in the initiation of atherosclerotic lesion formation.
J Exp Med
2002
, vol. 
196
 
7
(pg. 
887
-
896
)
15
Fuchs
 
TA
Brill
 
A
Duerschmied
 
D
, et al. 
Extracellular DNA traps promote thrombosis.
Proc Natl Acad Sci USA
2010
, vol. 
107
 
36
(pg. 
15880
-
15885
)
16
Massberg
 
S
Grahl
 
L
von Bruehl
 
ML
, et al. 
Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases.
Nat Med
2010
, vol. 
16
 
8
(pg. 
887
-
896
)
17
Knight
 
JS
Luo
 
W
O’Dell
 
AA
, et al. 
Peptidylarginine deiminase inhibition reduces vascular damage and modulates innate immune responses in murine models of atherosclerosis.
Circ Res
2014
, vol. 
114
 
6
(pg. 
947
-
956
)
18
Warnatsch
 
A
Ioannou
 
M
Wang
 
Q
Papayannopoulos
 
V
Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis.
Science
2015
, vol. 
349
 
6245
(pg. 
316
-
320
)
19
Mangold
 
A
Alias
 
S
Scherz
 
T
, et al. 
Coronary neutrophil extracellular trap burden and deoxyribonuclease activity in ST-elevation acute coronary syndrome are predictors of ST-segment resolution and infarct size.
Circ Res
2015
, vol. 
116
 
7
(pg. 
1182
-
1192
)
20
Riegger
 
J
Byrne
 
RA
Joner
 
M
, et al. 
Prevention of Late Stent Thrombosis by an Interdisciplinary Global European Effort (PRESTIGE) Investigators; Prevention of Late Stent Thrombosis by an Interdisciplinary Global European Effort PRESTIGE Investigators
Histopathological evaluation of thrombus in patients presenting with stent thrombosis. A multicenter European study: a report of the prevention of late stent thrombosis by an interdisciplinary global European effort consortium [published online ahead of print August 30, 2015].
Eur Heart J
 
doi:10.1093/eurheartj/ehv419
21
Megens
 
RT
Vijayan
 
S
Lievens
 
D
, et al. 
Presence of luminal neutrophil extracellular traps in atherosclerosis.
Thromb Haemost
2012
, vol. 
107
 
3
(pg. 
597
-
598
)
22
Etulain
 
J
Martinod
 
K
Wong
 
SL
Cifuni
 
SM
Schattner
 
M
Wagner
 
DD
P-selectin promotes neutrophil extracellular trap formation in mice.
Blood
2015
, vol. 
126
 
2
(pg. 
242
-
246
)
23
Ahmadsei
 
M
Lievens
 
D
Weber
 
C
von Hundelshausen
 
P
Gerdes
 
N
Immune-mediated and lipid-mediated platelet function in atherosclerosis.
Curr Opin Lipidol
2015
, vol. 
26
 
5
(pg. 
438
-
448
)
24
Carvalho
 
AC
Colman
 
RW
Lees
 
RS
Platelet function in hyperlipoproteinemia.
N Engl J Med
1974
, vol. 
290
 
8
(pg. 
434
-
438
)
25
Connor
 
WE
Hoak
 
JC
Warner
 
ED
Massive thrombosis produced by fatty acid infusion.
J Clin Invest
1963
, vol. 
42(6)
 (pg. 
860
-
866
)
26
Owens
 
AP
Passam
 
FH
Antoniak
 
S
, et al. 
Monocyte tissue factor-dependent activation of coagulation in hypercholesterolemic mice and monkeys is inhibited by simvastatin.
J Clin Invest
2012
, vol. 
122
 
2
(pg. 
558
-
568
)
27
Harmon
 
JT
Tandon
 
NN
Hoeg
 
JM
Jamieson
 
GA
Thrombin binding and response in platelets from patients with dyslipoproteinemias: increased stimulus-response coupling in type II hyperlipoproteinemia.
Blood
1986
, vol. 
68
 
2
(pg. 
498
-
505
)
28
Podrez
 
EA
Byzova
 
TV
Febbraio
 
M
, et al. 
Platelet CD36 links hyperlipidemia, oxidant stress and a prothrombotic phenotype.
Nat Med
2007
, vol. 
13
 
9
(pg. 
1086
-
1095
)
29
Korporaal
 
SJ
Meurs
 
I
Hauer
 
AD
, et al. 
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
2011
, vol. 
31
 
1
(pg. 
34
-
42
)
30
Boullier
 
A
Bird
 
DA
Chang
 
MK
, et al. 
 
Scavenger receptors, oxidized LDL, and atherosclerosis. Ann N Y Acad Sci. 2001;947:214-223
31
Magwenzi
 
S
Woodward
 
C
Wraith
 
KS
, et al. 
Oxidized LDL activates blood platelets through CD36/NOX2-mediated inhibition of the cGMP/protein kinase G signaling cascade.
Blood
2015
, vol. 
125
 
17
(pg. 
2693
-
2703
)
32
Shattil
 
SJ
Anaya-Galindo
 
R
Bennett
 
J
Colman
 
RW
Cooper
 
RA
Platelet hypersensitivity induced by cholesterol incorporation.
J Clin Invest
1975
, vol. 
55
 
3
(pg. 
636
-
643
)
33
Siddiqi
 
HK
Kiss
 
D
Rader
 
D
HDL-cholesterol and cardiovascular disease: rethinking our approach.
Curr Opin Cardiol
2015
, vol. 
30
 
5
(pg. 
536
-
542
)
34
Mineo
 
C
Deguchi
 
H
Griffin
 
JH
Shaul
 
PW
Endothelial and antithrombotic actions of HDL.
Circ Res
2006
, vol. 
98
 
11
(pg. 
1352
-
1364
)
35
Calkin
 
AC
Drew
 
BG
Ono
 
A
, et al. 
Reconstituted high-density lipoprotein attenuates platelet function in individuals with type 2 diabetes mellitus by promoting cholesterol efflux.
Circulation
2009
, vol. 
120
 
21
(pg. 
2095
-
2104
)
36
Ma
 
Y
Ashraf
 
MZ
Podrez
 
EA
Scavenger receptor BI modulates platelet reactivity and thrombosis in dyslipidemia.
Blood
2010
, vol. 
116
 
11
(pg. 
1932
-
1941
)
37
Trigatti
 
BL
Krieger
 
M
Rigotti
 
A
Influence of the HDL receptor SR-BI on lipoprotein metabolism and atherosclerosis.
Arterioscler Thromb Vasc Biol
2003
, vol. 
23
 
10
(pg. 
1732
-
1738
)
38
Jian
 
B
de la Llera-Moya
 
M
Ji
 
Y
, et al. 
Scavenger receptor class B type I as a mediator of cellular cholesterol efflux to lipoproteins and phospholipid acceptors.
J Biol Chem
1998
, vol. 
273
 
10
(pg. 
5599
-
5606
)
39
Ji
 
Y
Jian
 
B
Wang
 
N
, et al. 
Scavenger receptor BI promotes high density lipoprotein-mediated cellular cholesterol efflux.
J Biol Chem
1997
, vol. 
272
 
34
(pg. 
20982
-
20985
)
40
Yvan-Charvet
 
L
Pagler
 
TA
Wang
 
N
, et al. 
SR-BI inhibits ABCG1-stimulated net cholesterol efflux from cells to plasma HDL.
J Lipid Res
2008
, vol. 
49
 
1
(pg. 
107
-
114
)
41
Wang
 
X
Collins
 
HL
Ranalletta
 
M
, et al. 
Macrophage ABCA1 and ABCG1, but not SR-BI, promote macrophage reverse cholesterol transport in vivo.
J Clin Invest
2007
, vol. 
117
 
8
(pg. 
2216
-
2224
)
42
Braun
 
A
Trigatti
 
BL
Post
 
MJ
, et al. 
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
2002
, vol. 
90
 
3
(pg. 
270
-
276
)
43
Fuller
 
M
Dadoo
 
O
Serkis
 
V
, et al. 
The effects of diet on occlusive coronary artery atherosclerosis and myocardial infarction in scavenger receptor class B, type 1/low-density lipoprotein receptor double knockout mice.
Arterioscler Thromb Vasc Biol
2014
, vol. 
34
 
11
(pg. 
2394
-
2403
)
44
Lingwood
 
D
Simons
 
K
Lipid rafts as a membrane-organizing principle.
Science
2010
, vol. 
327
 
5961
(pg. 
46
-
50
)
45
Zhu
 
X
Owen
 
JS
Wilson
 
MD
, et al. 
Macrophage ABCA1 reduces MyD88-dependent Toll-like receptor trafficking to lipid rafts by reduction of lipid raft cholesterol.
J Lipid Res
2010
, vol. 
51
 
11
(pg. 
3196
-
3206
)
46
Yvan-Charvet
 
L
Pagler
 
T
Gautier
 
EL
, et al. 
ATP-binding cassette transporters and HDL suppress hematopoietic stem cell proliferation.
Science
2010
, vol. 
328
 
5986
(pg. 
1689
-
1693
)
47
Brodde
 
MF
Korporaal
 
SJ
Herminghaus
 
G
, et al. 
Native high-density lipoproteins inhibit platelet activation via scavenger receptor BI: role of negatively charged phospholipids.
Atherosclerosis
2011
, vol. 
215
 
2
(pg. 
374
-
382
)
48
Machlus
 
KR
Italiano
 
JE
The incredible journey: From megakaryocyte development to platelet formation.
J Cell Biol
2013
, vol. 
201
 
6
(pg. 
785
-
796
)
49
Kaushansky
 
K
Thrombopoiesis.
Semin Hematol
2015
, vol. 
52
 
1
(pg. 
4
-
11
)
50
Viny
 
AD
Levine
 
RL
Genetics of myeloproliferative neoplasms.
Cancer J
2014
, vol. 
20
 
1
(pg. 
61
-
65
)
51
Fessler
 
MB
Rose
 
K
Zhang
 
Y
Jaramillo
 
R
Zeldin
 
DC
Relationship between serum cholesterol and indices of erythrocytes and platelets in the US population.
J Lipid Res
2013
, vol. 
54
 
11
(pg. 
3177
-
3188
)
52
Santimone
 
I
Di Castelnuovo
 
A
De Curtis
 
A
, et al. 
MOLI-SANI Project Investigators
White blood cell count, sex and age are major determinants of heterogeneity of platelet indices in an adult general population: results from the MOLI-SANI project.
Haematologica
2011
, vol. 
96
 
8
(pg. 
1180
-
1188
)
53
Sloan
 
A
Gona
 
P
Johnson
 
AD
Cardiovascular correlates of platelet count and volume in the Framingham Heart Study.
Ann Epidemiol
2015
, vol. 
25
 
7
(pg. 
492
-
498
)
54
Harker
 
LA
Hazzard
 
W
Platelet kinetic studies in patients with hyperlipoproteinemia: effects of clofibrate therapy.
Circulation
1979
, vol. 
60
 
3
(pg. 
492
-
496
)
55
Jäger
 
E
Sinzinger
 
H
Widhalm
 
K
Kaliman
 
J
Höfer
 
R
Platelet half-life in patients with primary hyperlipoproteinemia type IIa, IIb, and IV according to Fredrickson with and without clinical signs of atherosclerosis [in German].
Wien Klin Wochenschr
1982
, vol. 
94
 
16
(pg. 
421
-
425
)
56
Murphy
 
EA
Mustard
 
JF
Coagulation tests and platelet economy in atherosclerotic and control subjects.
Circulation
1962
, vol. 
25
 (pg. 
114
-
125
)
57
Grozovsky
 
R
Begonja
 
AJ
Liu
 
K
, et al. 
The Ashwell-Morell receptor regulates hepatic thrombopoietin production via JAK2-STAT3 signaling.
Nat Med
2015
, vol. 
21
 
1
(pg. 
47
-
54
)
58
Li
 
J
van der Wal
 
DE
Zhu
 
G
, et al. 
Desialylation is a mechanism of Fc-independent platelet clearance and a therapeutic target in immune thrombocytopenia.
Nat Commun
2015
, vol. 
6
 pg. 
7737
 
59
Martin
 
JF
Kristensen
 
SD
Mathur
 
A
Grove
 
EL
Choudry
 
FA
The causal role of megakaryocyte–platelet hyperactivity in acute coronary syndromes.
Nat Rev Cardiol
2012
, vol. 
9
 
11
(pg. 
658
-
670
)
60
Pathansali
 
R
Smith
 
N
Bath
 
P
Altered megakaryocyte-platelet haemostatic axis in hypercholesterolaemia.
Platelets
2001
, vol. 
12
 
5
(pg. 
292
-
297
)
61
Wu
 
Y
Wu
 
H
Mueller
 
C
, et al. 
 
Baseline platelet count and clinical outcome in acute coronary syndrome. Circ J. 2012;76(3):704-711
62
Dole
 
VS
Matuskova
 
J
Vasile
 
E
, et al. 
Thrombocytopenia and platelet abnormalities in high-density lipoprotein receptor-deficient mice.
Arterioscler Thromb Vasc Biol
2008
, vol. 
28
 
6
(pg. 
1111
-
1116
)
63
Gomes
 
AL
Carvalho
 
T
Serpa
 
J
Torre
 
C
Dias
 
S
Hypercholesterolemia promotes bone marrow cell mobilization by perturbing the SDF-1:CXCR4 axis.
Blood
2010
, vol. 
115
 
19
(pg. 
3886
-
3894
)
64
Seijkens
 
T
Hoeksema
 
MA
Beckers
 
L
, et al. 
 
Hypercholesterolemia-induced priming of hematopoietic stem and progenitor cells aggravates atherosclerosis. FASEB J. 2014;28(5):2202-2213
65
Westerterp
 
M
Bochem
 
AE
Yvan-Charvet
 
L
Murphy
 
AJ
Wang
 
N
Tall
 
AR
ATP-binding cassette transporters, atherosclerosis, and inflammation.
Circ Res
2014
, vol. 
114
 
1
(pg. 
157
-
170
)
66
Murphy
 
AJ
Akhtari
 
M
Tolani
 
S
, et al. 
ApoE regulates hematopoietic stem cell proliferation, monocytosis, and monocyte accumulation in atherosclerotic lesions in mice.
J Clin Invest
2011
, vol. 
121
 
10
(pg. 
4138
-
4149
)
67
Wang
 
N
Yvan-Charvet
 
L
Lutjohann
 
D
, et al. 
 
ATP-binding cassette transporters G1 and G4 mediate cholesterol and desmosterol efflux to HDL and regulate sterol accumulation in the brain. FASEB J. 2008;22(4):1073-1082
68
Murphy
 
AJ
Bijl
 
N
Yvan-Charvet
 
L
, et al. 
Cholesterol efflux in megakaryocyte progenitors suppresses platelet production and thrombocytosis.
Nat Med
2013
, vol. 
19
 
5
(pg. 
586
-
594
)
69
Saur
 
SJ
Sangkhae
 
V
Geddis
 
AE
Kaushansky
 
K
Hitchcock
 
IS
Ubiquitination and degradation of the thrombopoietin receptor c-Mpl.
Blood
2010
, vol. 
115
 
6
(pg. 
1254
-
1263
)
70
Oneyama
 
C
Iino
 
T
Saito
 
K
Suzuki
 
K
Ogawa
 
A
Okada
 
M
Transforming potential of Src family kinases is limited by the cholesterol-enriched membrane microdomain.
Mol Cell Biol
2009
, vol. 
29
 
24
(pg. 
6462
-
6472
)
71
Pikman
 
Y
Lee
 
BH
Mercher
 
T
, et al. 
MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia.
PLoS Med
2006
, vol. 
3
 
7
pg. 
e270
 
72
Murphy
 
AJ
Sarrazy
 
V
Wang
 
N
, et al. 
Deficiency of ATP-binding cassette transporter B6 in megakaryocyte progenitors accelerates atherosclerosis in mice.
Arterioscler Thromb Vasc Biol
2014
, vol. 
34
 
4
(pg. 
751
-
758
)
73
Krishnamurthy
 
PC
Du
 
G
Fukuda
 
Y
, et al. 
Identification of a mammalian mitochondrial porphyrin transporter.
Nature
2006
, vol. 
443
 
7111
(pg. 
586
-
589
)
Sign in via your Institution