Skip to main content
European Heart Journal logoLink to European Heart Journal
. 2016 Feb 10;37(14):1113–1121. doi: 10.1093/eurheartj/ehv718

Disordered haematopoiesis and athero-thrombosis

Andrew J Murphy 1,2,*, Alan R Tall 3
PMCID: PMC4823636  PMID: 26869607

Abstract

Atherosclerosis, the major underlying cause of cardiovascular disease, is characterized by a lipid-driven infiltration of inflammatory cells in large and medium arteries. Increased production and activation of monocytes, neutrophils, and platelets, driven by hypercholesterolaemia and defective high-density lipoproteins-mediated cholesterol efflux, tissue necrosis and cytokine production after myocardial infarction, or metabolic abnormalities associated with diabetes, contribute to atherogenesis and athero-thrombosis. This suggests that in addition to traditional approaches of low-density lipoproteins lowering and anti-platelet drugs, therapies directed at abnormal haematopoiesis, including anti-inflammatory agents, drugs that suppress myelopoiesis, and excessive platelet production, rHDL infusions and anti-obesity and anti-diabetic agents, may help to prevent athero-thrombosis.

Keywords: Athero-thrombosis, Haematopoiesis, Atherosclerosis, Monocytes, Neutrophils, Platelets


In Western societies, the consumption of diets high in calories, saturated fat, and cholesterol combined with a sedentary lifestyle lead to a high prevalence of atherosclerotic cardiovascular disease (CVD). High-circulating blood lipids, including elevated low-density lipoproteins (LDL) and triglyceride-rich lipoproteins, result in increased entry and retention of these particles in the arterial wall, leading to a macrophage-dominated chronic inflammatory process and eventuating in atherosclerotic plaque rupture or erosion, myocardial infarction, or thrombotic stroke.1 While elevated plasma cholesterol levels have an essential role in atherogenesis, co-morbidities such as smoking, hypertension, and diabetes accelerate atherosclerotic CVD. In addition, chronic kidney disease,2,3 recurrent infections,4,5 myeloproliferative neoplasms (MPNs),6–10 and autoimmune disease such as rheumatoid arthritis11,12 and systemic lupus erythematous13 also greatly increase the risk of athero-thrombosis. A common theme linking these diseases to athero-thrombosis is an overactive immune system, mediated in part by increased production and activation of innate immune cells.

Leukocytosis as a risk factor for cardiovascular disease

There is strong epidemiological evidence detailing the link between elevated white blood cells (WBCs) and CVD.14–16 Numerous studies of people with and without pre-existing CVD at baseline measurements show that WBC counts predict the incidence of cardiac events,17–27 largely irrespective of race or gender15,28–30 and after adjusting for confounding factors such as smoking, age, BMI, and lipids.17,28 It appears that the myeloid compartment of the WBCs, namely monocytes31–33 and neutrophils14,34–37 are the strongest predictors of cardiac events, while lymphocytes generally have no correlation or an inverse relationship.20,32 Preclinical animal models of atherosclerosis have identified monocytes, neutrophils, and platelets as important players in the disease process, where the levels of these cells in the blood influence disease initiation and progression38–45 (Figure 1). Cross-talk between these cells can also promote thrombotic disorders.46–49 In humans, the abundance of leucocytes can affect heart failure following a myocardial infarction,50,51 while also contributing to secondary cardiovascular events.52

Figure 1.

Figure 1

Cardiovascular risk factors promote myelopoiesis and contribute to athero-thrombosis. (A) Increased plasma low-density lipoproteins and decreased high-density lipoproteins levels, (B) hyperglycaemia, and (C) obesity are major cardiovascular risk factors. Through various mechanisms, these risk factors directly or indirectly stimulate the production of myeloid cells (monocytes, neutrophils, and reticulated platelets) increasing the abundance in circulation. Hypercholesterolaemia also promotes the mobilization of haematopoietic stem cells (HSCs) to the spleen resulting in extramedullary haematopoiesis further contributing to the circulating pool of myeloid cells. (1) The increased abundance in circulating myeloid cells enhances the progression and impairs the regression of the atheroma. (2) There is also increased platelet–leucocyte interactions that enhance the recruitment of the leucocytes to the atherosclerotic lesion. (3) Neutrophils activation can also result in the formation of neutrophil extracellular traps (NETs), which contribute to enhanced atherogenesis and athero-thrombosis by binding platelets.

Neutrophils and atherosclerotic cardiovascular disease

Neutrophils are the major WBC in humans accounting for ∼60% of the circulating WBCs. Neutrophils are relatively understudied in the setting of atherosclerosis as they are not always present within stable atherosclerotic lesions. However, it is appreciated that neutrophil activation can influence coronary artery disease (CAD). People with unstable angina have higher levels of neutrophil activation as measured by lower myeloperoxidase (MPO) levels (i.e. degranulation) compared to people with stable angina or healthy controls.53 Further, MPO levels were found to be inversely associated with the inflammatory molecule C-reactive protein (CRP).53 C-reactive protein could be playing a causative role in neutrophil activation particularly when CPR dissociates into a monomeric form.54 This monomeric form of CRP is formed by and activates platelets,55 and in turn platelets via P-selectin can activate neutrophils in acute coronary syndromes.56 Importantly, neutrophils have been identified in human carotid atherosclerotic plaques where their abundance correlates positively with features of vulnerability, i.e. lipid core area, macrophage abundance, vessel density, and negatively with collagen and smooth muscle cells.57 In murine models, neutrophils were shown to play a direct role in initiating atherosclerotic lesion formation.38 Neutrophil levels have also been shown to predict ischaemic heart disease over a 10-year period in the Caerphilly and Speedwell studies14 and also predict the risk of recurrent ischaemic events.58 Conversely, Yemenite Jews, who frequently present with benign hereditary neutropenia,59 rarely suffer from myocardial infarction,60 and those who do are generally not neutropenic.61 Neutropenic preclinical models also support the observations that lower neutrophil levels afford some protection against vascular disease, particularly in the early stages of disease development.38 There may be an important role for co-morbidities in driving the production of neutrophils and how they influence CVD, for example, in Type 1 diabetics (T1D) who have CVD compared with T1D without CVD.43 There are a number of theories as to how neutrophils may contribute to CVD, including releasing proteases that contribute to the rupturing of atherosclerotic lesions62,63 and MPO, which modifies proteins, particularly lipoproteins,64,65 and is raised in people with CAD.66,67

Neutrophil extracellular traps and athero-thrombosis

Recent findings suggest that neutrophils may also play an important role in athero-thrombotic events, by releasing DNA NETs (neutrophil extracellular traps) that may have a primary role in protecting against bacterial infections but also promote thrombus formation.68–71 These NETs, while primarily comprised DNA, also hold proteins of specific neutrophil granules including elastase, MPO, and gelatinase, all of which have atherogenic and plaque-destabilizing properties. NETosis with or without death of the neutrophil leads to release cytoplasmic proteins including damage-associated molecular pattern molecules (DAMPs) such as S100A8/A9 (myeloid-related protein 8/14; MRP8/14) and high mobility box 1 (HMGB1), which can further potentiate inflammation and coagulation. These DAMPS can also be presented by activated platelets to promote NETosis.72 The nucleic and chromatin contents of NETs can be found systemically and are positively associated with CAD, pro-thrombotic state, and adverse vascular events.73 Recent studies have suggested that neutrophil nets are present in atherosclerotic plaques in WTD-fed Apoe−/− mice and that reducing net formation by DNAse injection or by crossing mice with neutrophil elastase/proteinase 3-deficient mice reduced formation of atherosclerotic lesions. However, the major impact was on advanced rather than early lesions, in contrast to earlier studies that had implicated neutrophils in early lesion development.38 Moreover, the critical genetic test, involving atherosclerosis susceptible mice with genetic knockouts of peptidylarginine deiminase 4 (PAD4) (which has an essential role in chromatin decondensation and net formation),74 has not yet been reported. Neutrophils can be stimulated to release NETs in vitro following incubation with cholesterol crystals;71 however, it is uncertain whether neutrophils have significant contact with cholesterol crystals within atherosclerotic lesions. In another study of human atherosclerotic lesions with eroded plaques, nets appeared to be formed at the surface of plaques, where they were proposed to contribute to endothelial cell apoptosis and pro-thrombotic effects.70 Platelet–leucocyte interactions have also been described to trigger neutrophil activation and NET formation in sepsis, and while this was primarily in the liver sinusoids,75 this could be a potential mechanism in athero-thrombotic disease (Figure 1). Thus, additional studies on the role of nets in atherosclerosis may prove informative, especially in settings where neutrophils and platelet/neutrophil aggregates are increased. Along with the strong evidence for a role in thrombosis,69 these studies suggest that NETs may have an important role in promoting athero-thrombosis.

Monocyte subsets and atherosclerotic cardiovascular disease

Monocytes account for around 10% of the WBCs and are also a heterogeneous population of cells. They can be divided into three sub-populations based on the expression of the cell surface markers CD14 (co-LPS receptor) and CD16 (FcγIII receptor). Classical monocytes are defined as CD14+CD16, intermediate are CD14+CD16+, while non-classical are CD14dimCD16+.76 These subsets differ in many respects, including in their expression of adhesion molecules, chemokine receptors, and functionality.76 CD14+ monocytes are more phagocytic, produce larger amounts of ROS and cytokines in response to bacterial cues, while CD16+ monocytes appear to be akin to murine Ly6-Clo monocytes in that they can patrol the endothelium and appear to be adapted for viral rather than bacterial immunity. CD16+ monocytes selectively produce TNF-α, IL-β, and CCL3 in response to viruses and immune complexes containing nucleic acids via TLR7 and TLR8.76 The role of monocyte subsets in CVD is not well established, but there are some reports associating levels of specific subsets with disease. Patients with CAD77–79 or unstable atherosclerotic plaques80 have higher numbers of CD16+ monocytes. On the other hand, a decrease in these CD16+ monocytes is associated with plaque stabilization.81 Elevated levels of CD16+ monocytes have been shown to independently predict cardiovascular events82 and correlate with markers of atherosclerosis including carotid intima media thickness (cIMT) and risk algorithms (Framingham and SCORE).83 These associations with the CD16+ monocytes are observed in people with diseases associated with CVD including obesity83 and chronic kidney disease,84 and abundance in CD16+ monocytes after stroke85 and myocardial infarction86 can predict the clinical course and inform on the prognosis. A recent study has also shown that Ly6-Clo monocytes adhere more readily to endothelium and extravasate into tissues resulting in macrophage accumulation in the setting of hypertriglyceridaemia.87 Given that the CD14+CD16 monocytes are the most abundant monocytes in humans and these cells are similar to the CCR2+Ly6Chi monocytes in mice, which are consistently been reported to more readily enter atherosclerotic lesions,44,45 it seems likely that both CD16 and CD16+ monocytes contribute to atherogenesis in humans and the distinct roles of different subsets require further investigation.

Platelets and platelet–leucocyte aggregates in athero-thrombosis

Platelets also have a major role in the initial and advanced stages of athero-thrombotic disease.39,40,42,88,89 Meta-analyses with combined data from over 140 randomized trials show that anti-platelet therapy reduces the risk of vascular events.90 However, platelet counts as predictors of disease have been far less studied. Thaulow et al.91 found that platelet counts and platelet reactivity independently correlate with CVD mortality in a cohort of healthy males. Infusion of activated platelets into Apoe−/− mice resulted in an increase in atherosclerosis, reflecting binding of platelets and platelet/leucocyte aggregates to arterial endothelium over atherosclerotic plaques with release and binding of chemokines to arterial endothelium. Sreeramkumar et al.92 have recently shown that activated platelets may bind to neutrophils after neutrophils have adhered to activated endothelium (Figure 1). The interaction of platelets with leucocytes is initiated by the binding of platelet P-selectin to P-selectin glycoprotein ligand-1 (PSGL-1), which localizes to the uropod (tail) of neutrophils as they bind to endothelium. Platelets and monocytes may be involved in a similar interaction on arterial endothelium. Platelet–leucocyte interactions trigger a series of events that contribute to the inflammatory reaction of the vessel wall and promotion of atherogenesis. Platelet–leucocyte interactions are also important as this can activate adhesion molecules (i.e. CD11b/c) and trigger cytokine expression (i.e. IL-1β) in leucocytes (i.e. CD11b/c) causing adhesion to arterial endothelium and promotion of atherosclerotic lesion formations.93 This process also activates the platelets and triggers the release of an array of atherogenic chemokines including CCL5, CCL2, and CXCL4, which promote entry of monocytes and neutrophils into lesions93 (Figure 1). Platelet–leucocyte transcellular metabolism of arachidonic acid leads to the synthesis of inflammatory, vasoconstrictive leukotrienes, and thromboxane A2, while also generating pro-resolving mediators such as lipoxins.94,95 The factors determining the balance of these opposing factors are poorly understood but could be important in determining the resolution of inflammatory processes such as atherosclerosis.

Monocyte–platelet interactions are observed in the setting of MI and are increased for at least 1 month after the acute event, suggesting that this mechanism could contribute to a secondary MI.86 There is also a clearly defined relationship between platelet size as determined by mean platelet volume (MPV), a marker of platelet production, and CVD.96–98 Mean platelet volume is increased in high-risk groups including those with diabetes, obesity, metabolic syndrome, after acute MI, and in restenosis of coronary angioplasty.99 In a study of over 200 000 people with a median follow-up of 4.6 years, MPV was found to predict mortality due to ischaemic heart disease.100 Mean platelet volume is also elevated in people with low levels of HDL.101 Increased MPV suggests an enriched population of larger immature or reticulated (RNA rich) platelets that are more reactive than mature platelets102 and do not respond as well as mature platelets to anti-platelet therapies such as aspirin and clopidogrel102 (Figure 1). This is possibly because reticulated platelets carry RNA allowing de novo synthesis of cyclooxygenase (COX)-1 and COX-2, overcoming effects of aspirin on these targets.103 This is particularly relevant in people with diabetes who typically have elevated reticulated platelets,102–106 increased MPV,106–108 and respond poorly to anti-platelet therapies.109,110 A high MPV was also shown to markedly increase CVD in people with diabetes.111 The increase in these platelet parameters appears to be driven by diabetes (hyperglycaemia) as the increase independently correlates with the severity of diabetes.112 Obese individuals also have an elevated MPV,113 which is reduced upon weight loss.114 Significantly elevated platelet counts also correlate with CVD in diabetic subjects.115 These observations suggest the importance of anti-platelet therapy in diabetics. Moreover, a deeper understanding of the mechanisms of platelet overproduction in diabetes could lead to the development of new approaches to preventing CHD in diabetics.

Athero-thrombosis in myeloproliferative disorders

Myeloproliferative neoplasms are blood disorders where increased production of myeloid cells is strongly associated with venous and arterial thrombosis (MI and stroke). These MPNs include essential thrombocytosis (ET), polycythemia vera (PV), and myelofibrosis. Cardiovascular disease is a major cause of morbidity and death in patients with MPNs.6–10 Essential thrombocythemia is also associated with thrombosis including CAD.116–119 Randomized clinical trials have shown that aspirin on top of other anti-thrombotic strategies (thromboprophylaxis) reduced the risk of vascular events;120 however, patients with MPNs still remain at significantly higher risk of thrombotic and athero-thrombotic events. It is becoming apparent that thrombosis in ET is linked to leukocytosis,121 potentially providing a major link to thrombotic events in MPNs as leukocytosis remains the strongest risk factor for thrombosis in people with PV.122–124 When people with PV are intensively treated with phlebotomy and/or hydroxyurea to achieve a haematocrit target of <45%, there was significantly fewer cardiovascular events compared with the less aggressively treated group. Interestingly leucocyte, but not platelet levels, reflected the abundance of haematocrit in these people,125 providing further evidence for the importance of WBC levels in people with MPNs and cardiovascular events. While the mechanisms are not completely understood, it is likely that enhanced leucocyte–platelet aggregates resulting in leucocyte and platelet activation and chemokine/cytokine release (as discussed above) are intimately involved.126–129 In addition, increased circulating leucocytes, possibly via the formation of platelet–leucocyte aggregates in MPN patients may give rise to NETS that likely contribute to both arterial and venous thrombosis as seen in other thrombotic disorders.48 Patients with MPNs often have increased monocytes, which may lead to increased entry into atherosclerotic plaques and contribute to increased macrophage foam cell formation. Interestingly, some genetic changes that promote MPNs, such as in LNK/SH2B3, may also be associated with platelet and leucocyte counts and with increased CVD in the general population,130,131 suggesting common mechanisms promoting athero-thrombosis in MPN and in the general population.

Disordered cholesterol metabolism links haematopoiesis to athero-thrombosis

Monocytosis is prominent in animal models of atherosclerosis and is increased in response to diets high in saturated fat and cholesterol.41,44,45,132 Moreover, monocytosis is associated with increased monocyte entry into plaques, and limitation of monocytosis or entry of monocytes into plaques reduces atherosclerosis, suggesting a causal relationship.41,43–45,133 While monocytosis may be related in part to increased inflammatory cytokines such as IL-3, GM-CSF, M-CSF, IL-1β, etc.,134–138 recent studies have uncovered a role of both hypercholesterolaemia and defective cholesterol efflux pathways in haematopoietic progenitors, both in the bone marrow (BM) and in the spleen, in promoting myelopoiesis in mouse atherosclerosis models.

Active cellular cholesterol efflux is mediated by ATP-binding cassette transporters, including ABCA1 that mediates cholesterol efflux to lipid-poor apoA-1 and minimally lipidated apoA-I particles, and ABCG1 and ABCG4 that mediate cholesterol efflux to HDL particles, especially larger HDL species.139 Unexpectedly, Abca-1/Abcg1 double knockout mice were found to develop marked monocytosis and neutrophilia that were associated with a dramatic expansion of the haematopoietic stem and multipotential progenitor cells (HSPCs) in the BM, uncovering an important role of cholesterol efflux pathways in the regulation of myelopoiesis140 (Figure 2). Another important cholesterol efflux pathway suppressing monocytosis and HSPC proliferation is mediated by apolipoprotein E (apoE), which may interact with ABCA1/G1 in haematopoietic stem cells and in multipotential progenitor cells, i.e. the LinSca1+cKit+ population (HSPCs) to promote cholesterol efflux.41 In both Abca1−/−/Abcg1−/− and Apoe−/− mice, HSPCs showed evidence of increased cholesterol in the plasma membrane associated with increased cell surface levels of the common β-subunit of the IL-3/GM-CSF receptor (CBS), allowing these cells to more readily sense these cytokines.41,140 ABCA1/ABCG1-deficient splenic macrophages also show increased expression of cytokines including M-CSF, which provide an additional stimulus of myelopoiesis141 (Figure 2).

Figure 2.

Figure 2

Defects in cellular cholesterol efflux pathways trigger myelopoiesis, extramedullary haematopoiesis, and enhanced atherosclerosis. In the bone marrow, defects in intrinsic cellular efflux pathways in haematopoietic stem (HSC) and myeloid progenitor cells result in increased membrane cholesterol levels and increased sensitivity to growth factor and cytokines. Deletion of ABCG4 in megakaryocyte progenitors (MkPs) results in increased c-MPL expression and enhanced thrombopoietin (TPO) signalling. This stimulates the production of immature reticulated platelets that can enhance atherogenesis via a number of mechanisms including deposition of cytokines (CCL5) and binding and activating leucocytes. Defective cholesterol efflux in haematopoietic stem cell and myeloid progenitors increased the cell surface abundance of the common β-subunit (CBS) of the IL-3, IL-5, and GM-CSF receptors resulting in enhanced proliferation. Inflammatory stimuli from a myocardial infarction including damage-associated molecular pattern molecules and IL-1β can influence haematopoietic stem cell proliferation and lineage fate. HSCs can also mobilize and migrate to the spleen when efferocytosis fails in macrophages with defective cholesterol efflux as there is a failure to shut down the expression of IL-23; thus, IL-17 and in turn G-CSF levels remain increased. In the spleen, there is an increased abundance of the innate response activator B cells (IRA B-cells) in the setting of hypercholesterolaemia, which produce GM-CSF driving the haematopoietic stem cells to produce monocytes and neutrophils. Defective cholesterol efflux in splenic macrophages also promotes M-CSF production to enhance myelopoiesis and CCL2 to promote monocyte migration. Together, the increased abundance of platelets, monocytes, and neutrophils all contribute to promoting the accumulation of macrophages in the atherosclerotic lesion.

In line with these findings, injection of neutralizing antibodies to IL-3 and GM-CSF in WTD-fed Apoe−/− mice significantly reduced the production of splenic monocytes and in turn triggered apoptosis.142 IL-3 and GM-CSF appear to be made in IRA B-cells, which arise from peritoneal B1a cells.143 The IRA B cells play an important role in the spleen and are not only expanded in Apoe−/− mice but also produce more cytokines134 (Figure 2). Ldlr−/− mice transplanted with Apoe−/−Cbs−/− BM had fewer circulating monocytes and neutrophils, which was accompanied by a reduction in BM and splenic stem and progenitor cells compared with mice that received Apoe−/− BM.138 Interestingly, the IRA B cells are sensitive to the hypercholesterolaemic environment (or the inflammation associated with it), which is dependent on the expression of the CBS. Deletion of the CBS lowered the numbers and proliferation of the IRA B cells. Initially, deletion of the CBS translated into smaller atherosclerotic lesions with fewer macrophages. However, when studies were extended to look at more advanced lesions, it was discovered that deletion of the CBS resulted in lesions of similar size again with fewer macrophages, but these lesions had significantly larger necrotic cores. The decrease in macrophages and larger necrotic cores was linked to increased macrophage apoptosis. This appeared to be due to a complete absence of Abcg1 expression, which has been shown to render macrophages more susceptible to apoptosis.144 Interestingly, GM-CSF has previously been identified to stimulate Abcg1 expression via PPARγ, suggesting an important pro-survival role for this cytokine in atherosclerosis. The increase in necrotic core formation associated with lower GM-CSF signalling in advanced plaques suggests the need for caution when using therapies that inhibit GM-CSF, e.g. in people with autoimmune diseases that are also at high risk of CVD, e.g. rheumatoid arthritis.145

Increased LDL cholesterol has also been suggested to contribute to the production of WBCs (monocytes and neutrophils) in preclinical models,38,41,44,45,146,147 and this can be dampened when cholesterol levels are restored to normal levels by diet147 or statin44 interventions. However, statins do not appear to effectively lower WBCs in people with CVD,148 suggesting that targeting LDL levels alone via statins may not be sufficient to reduce excessive WBC production. Studies in genetically modified mice showed that hypercholesterolaemia (i.e. increased LDL) along with decreased HDL levels due to an Apoa1 gene mutation were both required to influence monocyte counts.149 Similarly, in children with heterozygous familial hypercholesterolemia who were statin naive, HDL cholesterol levels were inversely related to blood monocyte counts.149 Together these findings support the hypothesis that HDL inversely correlates with monocyte levels, particularly in the setting of hypercholesterolaemia, and suggest that to reduce leucocyte production it may be necessary to increase cholesterol efflux in myeloid progenitors as well as lowering LDL levels, for example by rHDL infusion or LXR activator treatment41 (Table 1).

Table 1.

Mechanisms of monocyte production in CVD and potential interventions

Disease complication Initiating cell/ligand Target cell/receptor Intervention References
Hypercholesterolaemia and defective cholesterol efflux (ABCA1/G1 and apoE) Spleen
– IRA B cell/IL-3 and GM-CSF
– Macrophage/M-CSF and CCL2
HSPC/CBS rHDL 41,44,45,134,138,140,141,142
Myocardial infarction Damaged myocardium
– DAMPs
– IL-1β
HSPCs
– TLR4
– IL-1R
?Paquinimod
?Anakinra, ?Canakinumab
52,157,159,162,164,165,166
Diabetes/hyperglycaemia Neutrophil/S100A8/A9

?
CMPs/RAGE
Defective efflux in HSPCs
SGLT2i
?Paquinimod
Anti-miR33
43,192
Obesity ATM/IL-1β CMP and GMP/IL-1R Anakinra
?Paquinimod
167

Enhanced thrombopoiesis and atherosclerosis

As discussed above, platelets play an important role in CVD, and studies have linked their enhanced production and activation to cardiac events.39,40,42,88,89 Platelets promote atherogenesis via a multitude of pathways, from priming circulating monocytes and neutrophils so they are ready to adhere to the endothelium, to depositing potent chemokines such as RANTES (CCL5) and platelet factor 4 (CXCL4) on monocytes and the endothelium lining the atherosclerotic lesion.39 The membrane-bound platelet adhesion molecule P-selectin appears to be required for this to occur, and P-selectin-deficient platelets do not elicit the same response.39 It should also be noted that a soluble form of P-selectin occurs in vivo and has been shown to activate leucocytes from people with peripheral arterial occlusive disease (PAOD).150,151 However, injections of sP-selectin only moderately promote atherosclerosis,152 suggesting the platelet–leucocyte interaction is key in this atherogenic pathway.

While the study by Huo et al. was seminal in understanding the contribution of platelets to atherogenesis, they relied on adoptive transfer of activated platelets. More recently, models of enhanced thrombopoiesis have confirmed the important contribution that platelets play to atherogenesis.42,153 The cholesterol transporter ABCG4, which promotes cholesterol efflux to HDL, was shown to regulate platelet production.42 ABCG4 was highly expressed in MkPs, and haematopoietic deficiency of Abcg4 resulted in enhanced atherogenesis, which again appeared to be attributable to an increase in total platelets, reticulated platelets, enhanced activation, and increased platelet–leucocyte aggregates42 (Figure 2). Deletion of Abcg4−/− resulted in enhanced expression of the thrombopoietin (TPO) receptor, c-MPL on MkPs making them more sensitive to TPO signalling. It was found that LYN kinase acts a sensor of membrane cholesterol, where LYN kinase is protected from phosphorylation when cellular cholesterol levels increase. This precludes LYN kinase from activating the E-3 ubiquitin ligase c-CBL and in turn prevents c-MPL from degradation. Interestingly, SNPs associated with platelet counts revealed a connection to c-CBL, which is in tight linkage disequilibrium with ABCG4. This suggests that SNPs could alter the expression of either or both these genes and could be responsible for altered platelet production.

Infusion of rHDL in WT mice, but not Abcg4−/− mice, significantly lowered platelet numbers, by reducing MkPs and cell surface c-MPL levels. The therapeutic potential of this pathway was demonstrated by infusion of rHDL which suppressed platelet counts in a mouse model of myelofibrosis and ET caused by an activating mutation in c-MPL (c-MPLW515L). These suggest a role for rHDL infusions or therapies that increase HDL production in the treatment of myeloproliferative disorders. While leucocyte levels were not measured in this study, rHDL does reduce monocytes and neutrophils,41 decreases the activation of these cells,154,155 and reduces platelet activation,156 suggesting that HDL therapies could reduce thrombotic risk in people with MPNs.

HSC mobilization, extramedullary haematopoiesis, and monocytosis with myocardial infarction

The high rate of a secondary athero-thrombotic events after an initial myocardial infarction may in part reflect an acceleration of the underlying atherosclerotic process caused by enhanced myelpoiesis.52 Swirski et al.157 initially showed the spleen was home to a population of monocytes that were ready to be mobilized after an inflammatory insult, such as an MI. While splenic reserves dropped during the first 24 h following an infarct, by Day 6 post-MI there was a significant expansion of splenic monocytes fuelling monocytosis and entry into the infarct area.158 While this had a beneficial effect on myocardial function, Dutta et al.52 discovered that after an MI, myeloid cells, particularly Ly6-Chi monocytes, also infiltrated atherosclerotic lesion causing a significant increase in plaque size and a shift towards an unstable phenotype. Interestingly, the continual supply of monocytes reflected an increased number of HSCs in the spleen. In this setting, there was an expansion of a myeloid-biased subset of activated HSCs that express CCR2,159 previously identified to be responsive to inflammatory cues in vivo.160 Genes that are regulated by myeloid translocation gene on chromosome 16 (Mtg16) were found to be enriched in the CCR2+ HSCs, and deletion of Mtg16 resulted in depletion of CCR2+ HSCs and monocytes after an MI. The CCR2+ HSCs have significantly higher TLR2 and 4 expressions in the steady state suggesting that they are ready to respond to inflammatory signals. HSCs express functional TLRs and may directly sense their ligands.161,162 While the endogenous TLR ligand(s) post-MI remains unknown, these could be DAMPs that have been released from dying or activated myocardial cells (Figure 2). Dutta et al. found that injection of HMBG1, a DAMP found to be increased in patients after an MI,163 significantly increased CCR2+ HSC proliferation. Other DAMPs that are increased after MI include S100A8/A9,164 which are correlated with leucocyte counts.165 Additionally, Sager et al.166 has also discovered that IL-1β is increased and can accelerate haematopoiesis, not to dissimilar has to what was discovered with IL-1β in the setting of obesity167 (Table 1). Extramedullary haematopoiesis is not only initiated after an MI, but also in inflammatory diseases that are associated with increased risk of CVD including autoimmune diseases and MPNs, suggesting a pro-atherogenic role in these settings as well.

Promoting cholesterol efflux inhibits haematopoietic stem cell mobilization and extramedullary haematopoiesis

In murine models of defective cholesterol efflux (i.e. Abca1−/−/Abcg1−/− and Apoe−/− mice), there is chronic HSC mobilization into the circulation and spleen168 (Figure 2). Interestingly, the mechanisms contributing to HSC mobilization were not intrinsic to the HSC and instead reflected changes in the BM niche in response to exogenous signals. This process was initiated by splenic macrophages and dendritic cells where defective cholesterol efflux resulted in enhanced production of IL-23. This stimulated a signalling axis involving IL-23, IL-17, and G-CSF,169,170 a well-known HSC-mobilizing cytokine. These processes were reversed by transgenic overexpression of human APOA-I in Abca1−/−/Abcg1−/− or infusion of rHDL in Apoe−/− mice, suggesting a novel therapeutic approach to suppression of extramedullary haematopoiesis in CVD, MPNs, and leukaemia.

Metabolic diseases, myelopoiesis, and atherosclerosis

Metabolic diseases including obesity, insulin resistance, and diabetes (Types 1 and 2) greatly increase the risk of CVD171–174 and are associated with elevations in WBCs.83,175–183 Even when LDL is lowered by statins in diabetics, there is increased residual CVD risk. Using an experimental model of atherosclerotic lesion regression caused by LDL lowering, Parathath et al.184 showed that the induction of diabetes with streptozotocin prevented regression of atherosclerotic lesions. In this model, hyperglycaemia promoted BM myelopoiesis,43 independent of changes in plasma cholesterol or insulin levels. The mechanism involved neutrophils sensing increased levels of blood glucose and responding with production and release of S100A8/A9. S100A8/A9 induced myelopoiesis, via binding to the pattern recognition receptor RAGE on common myeloid progenitor cells (CMPs) in the BM and initiation of an autocrine/paracrine loop via the induction of key myeloid-promoting cytokines M-CSF and GM-CSF in CMPs (Figure 3). As CMPs are upstream of MkPs, and diabetics have more reticulated platelets,102–106 this or a related mechanism could also contribute to the enhanced production of platelets in diabetes (Figure 3). Together, the increased production of myeloid cells and platelets may contribute to athero-thrombotic complications in diabetes. The importance of glycaemic control in promoting atherosclerotic lesion regression was shown by treatment with the SGLT2i dapagliflozin (Farxiga). SGLT2i treatment lowered blood glucose, decreased monocytosis, recruitment of monocytes into lesions, and promoted lesion regression167 (Table 1). These studies imply that tight blood glucose control (without transient hyperglycaemic spikes)185–187 by use of glycosuric and other agents may dampen monocyte production and entry of inflammatory monocytes into lesions, adding to the benefit of lipid-lowering strategies in diabetics. In this regard, a recent CVD outcomes trial (EMPA-REG OUTCOME)188 in over 7000 participants has been reported to show benefit of the SGLT2i empagliflozin (Jardiance).189 However, it should be noted that this was largely attributed to a reduction in heart failure, and more studies are required to explore this anti-atherogenic mechanism in people with diabetes.

Figure 3.

Figure 3

Mechanisms contributing to myeloid production in metabolic disorders. Hyperglycaemia: In the setting of elevated blood glucose, neutrophils are stimulated to produce S100A8/A9, which travels to the bone marrow to interact with RAGE on the surface of macrophages and common myeloid progenitors (CMPs) triggering the production of M-CSF and GM-CSF. These cytokines increase the abundance of common myeloid progenitors and granulocyte–macrophage progenitors (GMPs) promoting the production of monocytes and neutrophils. Obesity: In the context of obesity, local inflammation in the adipose tissue occurs which appears to be initiated by S100A8/A9 interacting with TLR4 on adipose tissue macrophages (ATMs). This induces IL-1β, which is processed by the NLRP3 inflammasome to its mature form. IL-1β then travels to the bone marrow and binds the IL-1 receptor, which is up-regulated on common myeloid progenitors and granulocyte–macrophage progenitors in the obese state. This interaction drives myelopoiesis. As people with diabetes and obesity have increased diabetes and common myeloid progenitor cells are precursors of megakaryocytes, this may be a mechanism contributing to increased platelets. The enhanced production of myeloid cells in diabetes impairs the regression of atherosclerotic lesions due to persistent entry of monocytes.

Decreased levels of ABCA1190 and ABCG1191 in macrophages and myeloid progenitors have been reported in diabetic animal models and could also contribute to enhanced myelopoiesis.192 Conversely, increased Abca1 and Abcg1 expression in mice achieved by administering anti-miR-33 was able to restore the defect in CMP/GMP Abca1 and Abcg1 gene expression and to reduce the abundance of CMPs and GMPs in the BM and monocytes in the blood of diabetic mice. While miR-33 antagonism appears to have pleiotropic effects,193–195 more specific approaches to increasing HDL levels, such as rHDL infusions, might also be effective at reducing monocytosis in diabetic mice.

Obese Ob/Ob leptin-deficient and diet-induced obese mice that are also insulin resistant and models of early T2D also develop profound monocytosis and neutrophilia.167 These animals were only mildly hyperglycaemic, and lowering blood glucose had no effect on monocyte counts indicating an underlying mechanism distinct from the STZ diabetes model (Figure 3). Using a fat transplantation model, visceral adipose tissue was shown to directly contribute to enhanced myelopoiesis. Moreover, the same ligand as in the T1D models, S100A8/A9 was involved, albeit through a different mechanism. Local, but not systemic increases in S100A8/A9, were shown to signal via CD11c+ adipose tissue macrophage (ATM) TLR4/MyD88 to trigger IL-1β expression,167 which promoted proliferation of BM CMPs and GMPs, leading to increased myelopoiesis, which presumably further promoted ATM accumulation and atherogenesis. A second study also corroborated a role for CD11c+ ATMs in promoting leucocyte production in obesity.196 While a variety of cytokines are likely involved in this inflammatory signalling, the effect could be blocked using the IL-1R antagonist, Anakinra, indicating the key role of IL-1β (Table 1). Clinical trials are underway targeting the IL-1 pathway in CVD,197 and these results suggest that there could be a particular benefit in patients with obesity and increased ATMs.198,199 A common molecule identified in diabetes and obesity appears to be S100A8/A9, which may provide a therapeutic target particularly as paquinimod (ABR-215757) that blocks the interaction and function of these DAMPs is an orphan drug for systemic sclerosis and appears to be well tolerated, suggesting that it could be used as a chronic therapy (Table 1).

Conclusion

In conclusion, elevated levels and activation of leucocytes and platelets promote atherosclerosis, arterial and venous thrombosis. Formation of platelet/leucocyte aggregates in the bloodstream and on arterial endothelium promotes the entry of monocytes and neutrophils into atherosclerotic lesions. Recent studies have implicated a role for neutrophil-derived DNA nets in venous and arterial thrombosis and possibly in atherogenesis. While elevated levels of atherogenic lipoproteins and reduced HDL-mediated cholesterol efflux promote macrophage foam cell formation and inflammation at the level of the arterial wall, they also act in the BM and spleen driving myelopoiesis and platelet production and worsening atherogenesis. Cholesterol accumulation in haematopoietic stem and progenitor cells promotes myelopoiesis, while macrophage foam cell formation in the spleen leads to cytokine release, further myelopoiesis and mobilization of haematopoietic stem cells to the spleen, further driving extramedullary haematopoiesis. The ability of the spleen to act as a reservoir for monocytes and neutrophils may be particularly important in the setting of myocardial infarction, and while having a beneficial effect for the healing process, may also promote further entry of leucocytes into atherosclerotic lesions. In addition, Type 1 and 2 diabetes also promote excessive production of inflammatory cells, involving distinct mechanisms invoked by hyperglycaemia or adipose tissue inflammatory macrophages, production of S100A8/A9, and expansion and activation of BM common myeloid progenitors. These mechanistic insights suggest a variety of novel approaches to the prevention of athero-thrombosis. Even with dramatic improvements in LDL lowering, there will be a large burden of residual athero-thrombotic disease, and a need for further novel therapies aimed at limiting the production and activation of inflammatory cells, especially in patients with myeloproliferative disorders, obesity, and diabetes.

Funding

A.J.M. was supported by a fellowship from the Australian National Health and Medical Research Council (NHMRC) and a future leader fellowship from the National Heart Foundation. A.R.T was supported by an NIH grant (HL107653) and the Leducq Foundation.

Conflict of interest: none declared.

References

  • 1. Moore KJ, Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell 2011;145:341–355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Gansevoort RT, Correa-Rotter R, Hemmelgarn BR, Jafar TH, Heerspink HJ, Mann JF, Matsushita K, Wen CP. Chronic kidney disease and cardiovascular risk: epidemiology, mechanisms, and prevention. Lancet 2013;382:339–352. [DOI] [PubMed] [Google Scholar]
  • 3. Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N Engl J Med 2004;351:1296–1305. [DOI] [PubMed] [Google Scholar]
  • 4. Li X, Kolltveit KM, Tronstad L, Olsen I. Systemic diseases caused by oral infection. Clin Microbiol Rev 2000;13:547–558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Madjid M, Miller CC, Zarubaev VV, Marinich IG, Kiselev OI, Lobzin YV, Filippov AE, Casscells SW 3rd. Influenza epidemics and acute respiratory disease activity are associated with a surge in autopsy-confirmed coronary heart disease death: results from 8 years of autopsies in 34 892 subjects. Eur Heart J 2007;28:1205–1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Cervantes F, Dupriez B, Pereira A, Passamonti F, Reilly JT, Morra E, Vannucchi AM, Mesa RA, Demory JL, Barosi G, Rumi E, Tefferi A. New prognostic scoring system for primary myelofibrosis based on a study of the International Working Group for Myelofibrosis Research and Treatment. Blood 2009;113:2895–2901. [DOI] [PubMed] [Google Scholar]
  • 7. Finazzi G, Caruso V, Marchioli R, Capnist G, Chisesi T, Finelli C, Gugliotta L, Landolfi R, Kutti J, Gisslinger H, Marilus R, Patrono C, Pogliani EM, Randi ML, Villegas A, Tognoni G, Barbui T, Investigators E Acute leukemia in polycythemia vera: an analysis of 1638 patients enrolled in a prospective observational study. Blood 2005;105:2664–2670. [DOI] [PubMed] [Google Scholar]
  • 8. Hultcrantz M, Wilkes SR, Kristinsson SY, Andersson TM, Derolf AR, Eloranta S, Samuelsson J, Landgren O, Dickman PW, Lambert PC, Bjorkholm M. Risk and cause of death in patients diagnosed with myeloproliferative neoplasms in Sweden between 1973 and 2005: a population-based study. J Clin Oncol 2015;33:2288–2295. [DOI] [PubMed] [Google Scholar]
  • 9. Marchioli R, Finazzi G, Landolfi R, Kutti J, Gisslinger H, Patrono C, Marilus R, Villegas A, Tognoni G, Barbui T. Vascular and neoplastic risk in a large cohort of patients with polycythemia vera. J Clin Oncol 2005;23:2224–2232. [DOI] [PubMed] [Google Scholar]
  • 10. Polednak AP. Recent decline in the U.S. death rate from myeloproliferative neoplasms, 1999–2006. Cancer Epidemiol 2012;36:133–136. [DOI] [PubMed] [Google Scholar]
  • 11. Cobb S, Anderson F, Bauer W. Length of life and cause of death in rheumatoid arthritis. N Engl J Med 1953;249:553–556. [DOI] [PubMed] [Google Scholar]
  • 12. Van Doornum S, McColl G, Wicks IP. Accelerated atherosclerosis: an extraarticular feature of rheumatoid arthritis? Arthritis Rheum 2002;46:862–873. [DOI] [PubMed] [Google Scholar]
  • 13. Asanuma Y, Oeser A, Shintani AK, Turner E, Olsen N, Fazio S, Linton MF, Raggi P, Stein CM. Premature coronary-artery atherosclerosis in systemic lupus erythematosus. N Engl J Med 2003;349:2407–2415. [DOI] [PubMed] [Google Scholar]
  • 14. Sweetnam PM, Thomas HF, Yarnell JW, Baker IA, Elwood PC. Total and differential leukocyte counts as predictors of ischemic heart disease: the Caerphilly and Speedwell studies. Am J Epidemiol 1997;145:416–421. [DOI] [PubMed] [Google Scholar]
  • 15. Lee CD, Folsom AR, Nieto FJ, Chambless LE, Shahar E, Wolfe DA. White blood cell count and incidence of coronary heart disease and ischemic stroke and mortality from cardiovascular disease in African-American and White men and women: atherosclerosis risk in communities study. Am J Epidemiol 2001;154:758–764. [DOI] [PubMed] [Google Scholar]
  • 16. 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 1998;279:1477–1482. [DOI] [PubMed] [Google Scholar]
  • 17. Zalokar JB, Richard JL, Claude JR. Leukocyte count, smoking, and myocardial infarction. N Engl J Med 1981;304:465–468. [DOI] [PubMed] [Google Scholar]
  • 18. Weijenberg MP, Feskens EJ, Kromhout D. White blood cell count and the risk of coronary heart disease and all-cause mortality in elderly men. Arterioscler Thromb Vasc Biol 1996;16:499–503. [DOI] [PubMed] [Google Scholar]
  • 19. Takeda Y, Suzuki S, Fukutomi T, Kondo H, Sugiura M, Suzumura H, Murasaki G, Okutani H, Itoh M. Elevated white blood cell count as a risk factor of coronary artery disease: inconsistency between forms of the disease. Jpn Heart J 2003;44:201–211. [DOI] [PubMed] [Google Scholar]
  • 20. Rana JS, Boekholdt SM, Ridker PM, Jukema JW, Luben R, Bingham SA, Day NE, Wareham NJ, Kastelein JJ, Khaw KT. Differential leucocyte count and the risk of future coronary artery disease in healthy men and women: the EPIC-Norfolk Prospective Population Study. J Intern Med 2007;262:678–689. [DOI] [PubMed] [Google Scholar]
  • 21. Phillips AN, Neaton JD, Cook DG, Grimm RH, Shaper AG. Leukocyte count and risk of major coronary heart disease events. Am J Epidemiol 1992;136:59–70. [DOI] [PubMed] [Google Scholar]
  • 22. Manttari M, Manninen V, Koskinen P, Huttunen JK, Oksanen E, Tenkanen L, Heinonen OP, Frick MH. Leukocytes as a coronary risk factor in a dyslipidemic male population. Am Heart J 1992;123(4 Pt 1):873–877. [DOI] [PubMed] [Google Scholar]
  • 23. Grimm RH Jr, Neaton JD, Ludwig W. Prognostic importance of the white blood cell count for coronary, cancer, and all-cause mortality. JAMA 1985;254:1932–1937. [PubMed] [Google Scholar]
  • 24. Gillum RF, Mussolino ME, Madans JH. Counts of neutrophils, lymphocytes, and monocytes, cause-specific mortality and coronary heart disease: the NHANES-I epidemiologic follow-up study. Ann Epidemiol 2005;15:266–271. [DOI] [PubMed] [Google Scholar]
  • 25. Folsom AR, Wu KK, Rosamond WD, Sharrett AR, Chambless LE. Prospective study of hemostatic factors and incidence of coronary heart disease: the Atherosclerosis Risk in Communities (ARIC) Study. Circulation 1997;96:1102–1108. [DOI] [PubMed] [Google Scholar]
  • 26. Folsom AR, Szklo M, Stevens J, Liao F, Smith R, Eckfeldt JH. A prospective study of coronary heart disease in relation to fasting insulin, glucose, and diabetes. The Atherosclerosis Risk in Communities (ARIC) Study. Diabetes Care 1997;20:935–942. [DOI] [PubMed] [Google Scholar]
  • 27. Brown DW, Giles WH, Croft JB. White blood cell count: an independent predictor of coronary heart disease mortality among a national cohort. J Clin Epidemiol 2001;54:316–322. [DOI] [PubMed] [Google Scholar]
  • 28. Twig G, Afek A, Shamiss A, Derazne E, Tzur D, Gordon B, Tirosh A. White blood cell count and the risk for coronary artery disease in young adults. PLoS One 2012;7:e47183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Tatsukawa Y, Hsu WL, Yamada M, Cologne JB, Suzuki G, Yamamoto H, Yamane K, Akahoshi M, Fujiwara S, Kohno N. White blood cell count, especially neutrophil count, as a predictor of hypertension in a Japanese population. Hypertens Res 2008;31:1391–1397. [DOI] [PubMed] [Google Scholar]
  • 30. Margolis KL, Manson JE, Greenland P, Rodabough RJ, Bray PF, Safford M, Grimm RH Jr., Howard BV, Assaf AR, Prentice R, Women's Health Initiative Research Group. Leukocyte count as a predictor of cardiovascular events and mortality in postmenopausal women: the Women's Health Initiative Observational Study. Arch Intern Med 2005;165:500–508. [DOI] [PubMed] [Google Scholar]
  • 31. Yun KH, Oh SK, Park EM, Kim HJ, Shin SH, Lee EM, Rhee SJ, Yoo NJ, Kim NH, Jeong JW, Jeong MH. An increased monocyte count predicts coronary artery spasm in patients with resting chest pain and insignificant coronary artery stenosis. Korean J Intern Med 2006;21:97–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Olivares R, Ducimetiere P, Claude JR. Monocyte count: a risk factor for coronary heart disease? Am J Epidemiol 1993;137:49–53. [DOI] [PubMed] [Google Scholar]
  • 33. Ganda A, Magnusson M, Yvan-Charvet L, Hedblad B, Engstrom G, Ai D, Wang TJ, Gerszten RE, Melander O, Tall AR. Mild renal dysfunction and metabolites tied to low HDL cholesterol are associated with monocytosis and atherosclerosis. Circulation 2013;127:988–996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Avanzas P, Quiles J, Lopez de Sa E, Sanchez A, Rubio R, Garcia E, Lopez-Sendon JL. Neutrophil count and infarct size in patients with acute myocardial infarction. Int J Cardiol 2004;97:155–156. [DOI] [PubMed] [Google Scholar]
  • 35. Haumer M, Amighi J, Exner M, Mlekusch W, Sabeti S, Schlager O, Schwarzinger I, Wagner O, Minar E, Schillinger M. Association of neutrophils and future cardiovascular events in patients with peripheral artery disease. J Vasc Surg 2005;41:610–617. [DOI] [PubMed] [Google Scholar]
  • 36. Kawaguchi H, Mori T, Kawano T, Kono S, Sasaki J, Arakawa K. Band neutrophil count and the presence and severity of coronary atherosclerosis. Am Heart J 1996;132(1 Pt 1):9–12. [DOI] [PubMed] [Google Scholar]
  • 37. Wheeler JG, Mussolino ME, Gillum RF, Danesh J. Associations between differential leucocyte count and incident coronary heart disease: 1764 incident cases from seven prospective studies of 30,374 individuals. Eur Heart J 2004;25:1287–1292. [DOI] [PubMed] [Google Scholar]
  • 38. Drechsler M, Megens RT, van Zandvoort M, Weber C, Soehnlein O. Hyperlipidemia-triggered neutrophilia promotes early atherosclerosis. Circulation 2010;122:1837–1845. [DOI] [PubMed] [Google Scholar]
  • 39. 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 2003;9:61–67. [DOI] [PubMed] [Google Scholar]
  • 40. Koenen RR, von Hundelshausen P, Nesmelova IV, Zernecke A, Liehn EA, Sarabi A, Kramp BK, Piccinini AM, Paludan SR, Kowalska MA, Kungl AJ, Hackeng TM, Mayo KH, Weber C. Disrupting functional interactions between platelet chemokines inhibits atherosclerosis in hyperlipidemic mice. Nat Med 2009;15:97–103. [DOI] [PubMed] [Google Scholar]
  • 41. 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 2011;121:4138–4149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Murphy AJ, Bijl N, Yvan-Charvet L, Welch CB, Bhagwat N, Reheman A, Wang Y, Shaw JA, Levine RL, Ni H, Tall AR, Wang N. Cholesterol efflux in megakaryocyte progenitors suppresses platelet production and thrombocytosis. Nat Med 2013;19:586–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Nagareddy PR, Murphy AJ, Stirzaker RA, Hu Y, Yu S, Miller RG, Ramkhelawon B, Distel E, Westerterp M, Huang LS, Schmidt AM, Orchard TJ, Fisher EA, Tall AR, Goldberg IJ. Hyperglycemia promotes myelopoiesis and impairs the resolution of atherosclerosis. Cell Metab 2013;17:695–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Swirski FK, Libby P, Aikawa E, Alcaide P, Luscinskas FW, Weissleder R, Pittet MJ. Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J Clin Invest 2007;117:195–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Tacke F, Alvarez D, Kaplan TJ, Jakubzick C, Spanbroek R, Llodra J, Garin A, Liu J, Mack M, van Rooijen N, Lira SA, Habenicht AJ, Randolph GJ. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest 2007;117:185–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Palabrica T, Lobb R, Furie BC, Aronovitz M, Benjamin C, Hsu YM, Sajer SA, Furie B. Leukocyte accumulation promoting fibrin deposition is mediated in vivo by P-selectin on adherent platelets. Nature 1992;359:848–851. [DOI] [PubMed] [Google Scholar]
  • 47. Sarma J, Laan CA, Alam S, Jha A, Fox KA, Dransfield I. Increased platelet binding to circulating monocytes in acute coronary syndromes. Circulation 2002;105:2166–2171. [DOI] [PubMed] [Google Scholar]
  • 48. von Bruhl ML, Stark K, Steinhart A, Chandraratne S, Konrad I, Lorenz M, Khandoga A, Tirniceriu A, Coletti R, Kollnberger M, Byrne RA, Laitinen I, Walch A, Brill A, Pfeiler S, Manukyan D, Braun S, Lange P, Riegger J, Ware J, Eckart A, Haidari S, Rudelius M, Schulz C, Echtler K, Brinkmann V, Schwaiger M, Preissner KT, Wagner DD, Mackman N, Engelmann B, Massberg S. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 2012;209:819–835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Zarbock A, Polanowska-Grabowska RK, Ley K. Platelet-neutrophil-interactions: linking hemostasis and inflammation. Blood Rev 2007;21:99–111. [DOI] [PubMed] [Google Scholar]
  • 50. Arruda-Olson AM, Reeder GS, Bell MR, Weston SA, Roger VL. Neutrophilia predicts death and heart failure after myocardial infarction: a community-based study. Circ Cardiovasc Qual Outcomes 2009;2:656–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Munir TA, Afzal MN, Habib ur R. Baseline leukocyte count and acute coronary syndrome: predictor of adverse cardiac events, long and short-term mortality and association with traditional risk factors, cardiac biomarkers and C-reactive protein. J Ayub Med Coll Abbottabad 2009;21:46–50. [PubMed] [Google Scholar]
  • 52. Dutta P, Courties G, Wei Y, Leuschner F, Gorbatov R, Robbins CS, Iwamoto Y, Thompson B, Carlson AL, Heidt T, Majmudar MD, Lasitschka F, Etzrodt M, Waterman P, Waring MT, Chicoine AT, van der Laan AM, Niessen HW, Piek JJ, Rubin BB, Butany J, Stone JR, Katus HA, Murphy SA, Morrow DA, Sabatine MS, Vinegoni C, Moskowitz MA, Pittet MJ, Libby P, Lin CP, Swirski FK, Weissleder R, Nahrendorf M. Myocardial infarction accelerates atherosclerosis. Nature 2012;487:325–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Buffon A, Biasucci LM, Liuzzo G, D'Onofrio G, Crea F, Maseri A. Widespread coronary inflammation in unstable angina. N Engl J Med 2002;347:5–12. [DOI] [PubMed] [Google Scholar]
  • 54. Khreiss T, Jozsef L, Potempa LA, Filep JG. Loss of pentameric symmetry in C-reactive protein induces interleukin-8 secretion through peroxynitrite signaling in human neutrophils. Circ Res 2005;97:690–697. [DOI] [PubMed] [Google Scholar]
  • 55. Eisenhardt SU, Habersberger J, Murphy A, Chen YC, Woollard KJ, Bassler N, Qian H, von Zur Muhlen C, Hagemeyer CE, Ahrens I, Chin-Dusting J, Bobik A, Peter K. Dissociation of pentameric to monomeric C-reactive protein on activated platelets localizes inflammation to atherosclerotic plaques. Circ Res 2009;105:128–137. [DOI] [PubMed] [Google Scholar]
  • 56. Maugeri N, Rovere-Querini P, Evangelista V, Godino C, Demetrio M, Baldini M, Figini F, Coppi G, Slavich M, Camera M, Bartorelli A, Marenzi G, Campana L, Baldissera E, Sabbadini MG, Cianflone D, Tremoli E, D'Angelo A, Manfredi AA, Maseri A. An intense and short-lasting burst of neutrophil activation differentiates early acute myocardial infarction from systemic inflammatory syndromes. PLoS One 2012;7:e39484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. 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 2010;30:1842–1848. [DOI] [PubMed] [Google Scholar]
  • 58. Grau AJ, Boddy AW, Dukovic DA, Buggle F, Lichy C, Brandt T, Hacke W, Investigators C Leukocyte count as an independent predictor of recurrent ischemic events. Stroke 2004;35:1147–1152. [DOI] [PubMed] [Google Scholar]
  • 59. Shoenfeld Y, Weinberger A, Avishar R, Zamir R, Gazit E, Joshua H, Pinkhas J. Familial leukopenia among Yemenite Jews. Isr J Med Sci 1978;14:1271–1274. [PubMed] [Google Scholar]
  • 60. Dreyfuss F. The incidence of myocardial infarctions in various communities in Israel. Am Heart J 1953;45:749–755. [DOI] [PubMed] [Google Scholar]
  • 61. Shoenfeld Y, Pinkhas J. Leukopenia and low incidence of myocardial infarction. N Engl J Med 1981;304:1606. [DOI] [PubMed] [Google Scholar]
  • 62. Soehnlein O. Multiple roles for neutrophils in atherosclerosis. Circ Res 2012;110:875–888. [DOI] [PubMed] [Google Scholar]
  • 63. Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med 2011;17:1410–1422. [DOI] [PubMed] [Google Scholar]
  • 64. Hazen SL, Heinecke JW. 3-Chlorotyrosine, a specific marker of myeloperoxidase-catalyzed oxidation, is markedly elevated in low density lipoprotein isolated from human atherosclerotic intima. J Clin Invest 1997;99:2075–2081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Huang Y, Wu Z, Riwanto M, Gao S, Levison BS, Gu X, Fu X, Wagner MA, Besler C, Gerstenecker G, Zhang R, Li XM, DiDonato AJ, Gogonea V, Tang WH, Smith JD, Plow EF, Fox PL, Shih DM, Lusis AJ, Fisher EA, DiDonato JA, Landmesser U, Hazen SL. Myeloperoxidase, paraoxonase-1, and HDL form a functional ternary complex. J Clin Invest 2013;123:3815–3828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Brennan ML, Penn MS, Van Lente F, Nambi V, Shishehbor MH, Aviles RJ, Goormastic M, Pepoy ML, McErlean ES, Topol EJ, Nissen SE, Hazen SL. Prognostic value of myeloperoxidase in patients with chest pain. N Engl J Med 2003;349:1595–1604. [DOI] [PubMed] [Google Scholar]
  • 67. Zhang R, Brennan ML, Fu X, Aviles RJ, Pearce GL, Penn MS, Topol EJ, Sprecher DL, Hazen SL. Association between myeloperoxidase levels and risk of coronary artery disease. JAMA 2001;286:2136–2142. [DOI] [PubMed] [Google Scholar]
  • 68. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science 2004;303:1532–1535. [DOI] [PubMed] [Google Scholar]
  • 69. Martinod K, Wagner DD. Thrombosis: tangled up in NETs. Blood 2014;123:2768–2776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Quillard T, Araujo HA, Franck G, Shvartz E, Sukhova G, Libby P. TLR2 and neutrophils potentiate endothelial stress, apoptosis and detachment: implications for superficial erosion. Eur Heart J 2015;36:1394–1404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Warnatsch A, Ioannou M, Wang Q, Papayannopoulos V. Inflammation. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science 2015;349:316–320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Maugeri N, Campana L, Gavina M, Covino C, De Metrio M, Panciroli C, Maiuri L, Maseri A, D'Angelo A, Bianchi ME, Rovere-Querini P, Manfredi AA. Activated platelets present high mobility group box 1 to neutrophils, inducing autophagy and promoting the extrusion of neutrophil extracellular traps. J Thromb Haemost 2014;12:2074–2088. [DOI] [PubMed] [Google Scholar]
  • 73. Borissoff JI, Joosen IA, Versteylen MO, Brill A, Fuchs TA, Savchenko AS, Gallant M, Martinod K, Ten Cate H, Hofstra L, Crijns HJ, Wagner DD, Kietselaer BL. Elevated levels of circulating DNA and chromatin are independently associated with severe coronary atherosclerosis and a prothrombotic state. Arterioscler Thromb Vasc Biol 2013;33:2032–2040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Li P, Li M, Lindberg MR, Kennett MJ, Xiong N, Wang Y. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J Exp Med 2010;207:1853–1862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KD, Chakrabarti S, McAvoy E, Sinclair GD, Keys EM, Allen-Vercoe E, Devinney R, Doig CJ, Green FH, Kubes P. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med 2007;13:463–469. [DOI] [PubMed] [Google Scholar]
  • 76. Cros J, Cagnard N, Woollard K, Patey N, Zhang SY, Senechal B, Puel A, Biswas SK, Moshous D, Picard C, Jais JP, D'Cruz D, Casanova JL, Trouillet C, Geissmann F. Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors. Immunity 2010;33:375–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Schlitt A, Heine GH, Blankenberg S, Espinola-Klein C, Dopheide JF, Bickel C, Lackner KJ, Iz M, Meyer J, Darius H, Rupprecht HJ. CD14+CD16+ monocytes in coronary artery disease and their relationship to serum TNF-alpha levels. Thromb Haemost 2004;92:419–424. [DOI] [PubMed] [Google Scholar]
  • 78. Tallone T, Turconi G, Soldati G, Pedrazzini G, Moccetti T, Vassalli G. Heterogeneity of human monocytes: an optimized four-color flow cytometry protocol for analysis of monocyte subsets. J Cardiovasc Transl Res 2011;4:211–219. [DOI] [PubMed] [Google Scholar]
  • 79. Wildgruber M, Lee H, Chudnovskiy A, Yoon TJ, Etzrodt M, Pittet MJ, Nahrendorf M, Croce K, Libby P, Weissleder R, Swirski FK. Monocyte subset dynamics in human atherosclerosis can be profiled with magnetic nano-sensors. PLoS One 2009;4:e5663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Kashiwagi M, Imanishi T, Tsujioka H, Ikejima H, Kuroi A, Ozaki Y, Ishibashi K, Komukai K, Tanimoto T, Ino Y, Kitabata H, Hirata K, Akasaka T. Association of monocyte subsets with vulnerability characteristics of coronary plaques as assessed by 64-slice multidetector computed tomography in patients with stable angina pectoris. Atherosclerosis 2010;212:171–176. [DOI] [PubMed] [Google Scholar]
  • 81. Imanishi T, Ikejima H, Tsujioka H, Kuroi A, Ishibashi K, Komukai K, Tanimoto T, Ino Y, Takeshita T, Akasaka T. Association of monocyte subset counts with coronary fibrous cap thickness in patients with unstable angina pectoris. Atherosclerosis 2010;212:628–635. [DOI] [PubMed] [Google Scholar]
  • 82. Rogacev KS, Cremers B, Zawada AM, Seiler S, Binder N, Ege P, Grosse-Dunker G, Heisel I, Hornof F, Jeken J, Rebling NM, Ulrich C, Scheller B, Bohm M, Fliser D, Heine GH. CD14++CD16+ monocytes independently predict cardiovascular events: a cohort study of 951 patients referred for elective coronary angiography. J Am Coll Cardiol 2012;60:1512–1520. [DOI] [PubMed] [Google Scholar]
  • 83. Rogacev KS, Ulrich C, Blomer L, Hornof F, Oster K, Ziegelin M, Cremers B, Grenner Y, Geisel J, Schlitt A, Kohler H, Fliser D, Girndt M, Heine GH. Monocyte heterogeneity in obesity and subclinical atherosclerosis. Eur Heart J 2010;31:369–376. [DOI] [PubMed] [Google Scholar]
  • 84. Rogacev KS, Seiler S, Zawada AM, Reichart B, Herath E, Roth D, Ulrich C, Fliser D, Heine GH. CD14++CD16+ monocytes and cardiovascular outcome in patients with chronic kidney disease. Eur Heart J 2011;32:84–92. [DOI] [PubMed] [Google Scholar]
  • 85. Urra X, Villamor N, Amaro S, Gomez-Choco M, Obach V, Oleaga L, Planas AM, Chamorro A. Monocyte subtypes predict clinical course and prognosis in human stroke. J Cereb Blood Flow Metab 2009;29:994–1002. [DOI] [PubMed] [Google Scholar]
  • 86. Tapp LD, Shantsila E, Wrigley BJ, Pamukcu B, Lip GY. The CD14++CD16+ monocyte subset and monocyte-platelet interactions in patients with ST-elevation myocardial infarction. J Thromb Haemost 2012;10:1231–1241. [DOI] [PubMed] [Google Scholar]
  • 87. Saja MF, Baudino L, Jackson WD, Cook HT, Malik TH, Fossati-Jimack L, Ruseva M, Pickering MC, Woollard KJ, Botto M. Triglyceride-rich lipoproteins modulate the distribution and extravasation of Ly6C/Gr1 monocytes. Cell Rep 2015;12:1802–1815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. 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;9:658–670. [DOI] [PubMed] [Google Scholar]
  • 89. Trip MD, Cats VM, van Capelle FJ, Vreeken J. Platelet hyperreactivity and prognosis in survivors of myocardial infarction. N Engl J Med 1990;322:1549–1554. [DOI] [PubMed] [Google Scholar]
  • 90. Collaboration AT Collaborative overview of randomised trials of antiplatelet therapy—I: Prevention of death, myocardial infarction, and stroke by prolonged antiplatelet therapy in various categories of patients. Antiplatelet Trialists’ Collaboration. BMJ 1994;308:81–106. [PMC free article] [PubMed] [Google Scholar]
  • 91. Thaulow E, Erikssen J, Sandvik L, Stormorken H, Cohn PF. Blood platelet count and function are related to total and cardiovascular death in apparently healthy men. Circulation 1991;84:613–617. [DOI] [PubMed] [Google Scholar]
  • 92. Sreeramkumar V, Adrover JM, Ballesteros I, Cuartero MI, Rossaint J, Bilbao I, Nacher M, Pitaval C, Radovanovic I, Fukui Y, McEver RP, Filippi MD, Lizasoain I, Ruiz-Cabello J, Zarbock A, Moro MA, Hidalgo A. Neutrophils scan for activated platelets to initiate inflammation. Science 2014;346:1234–1238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Totani L, Evangelista V. Platelet-leukocyte interactions in cardiovascular disease and beyond. Arterioscler Thromb Vasc Biol 2010;30:2357–2361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Fiore S, Serhan CN. Formation of lipoxins and leukotrienes during receptor-mediated interactions of human platelets and recombinant human granulocyte/macrophage colony-stimulating factor-primed neutrophils. J Exp Med 1990;172:1451–1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Serhan CN, Sheppard KA. Lipoxin formation during human neutrophil-platelet interactions. Evidence for the transformation of leukotriene A4 by platelet 12-lipoxygenase in vitro. J Clin Invest 1990;85:772–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Berger JS, Eraso LH, Xie D, Sha D, Mohler ER 3rd. Mean platelet volume and prevalence of peripheral artery disease, the National Health and Nutrition Examination Survey, 1999–2004. Atherosclerosis 2010;213:586–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Chu SG, Becker RC, Berger PB, Bhatt DL, Eikelboom JW, Konkle B, Mohler ER, Reilly MP, Berger JS. Mean platelet volume as a predictor of cardiovascular risk: a systematic review and meta-analysis. J Thromb Haemost 2010;8:148–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Sansanayudh N, Anothaisintawee T, Muntham D, McEvoy M, Attia J, Thakkinstian A. Mean platelet volume and coronary artery disease: a systematic review and meta-analysis. Int J Cardiol 2014;175:433–440. [DOI] [PubMed] [Google Scholar]
  • 99. Vizioli L, Muscari S, Muscari A. The relationship of mean platelet volume with the risk and prognosis of cardiovascular diseases. Int J Clin Pract 2009;63:1509–1515. [DOI] [PubMed] [Google Scholar]
  • 100. Slavka G, Perkmann T, Haslacher H, Greisenegger S, Marsik C, Wagner OF, Endler G. Mean platelet volume may represent a predictive parameter for overall vascular mortality and ischemic heart disease. Arterioscler Thromb Vasc Biol 2011;31:1215–1218. [DOI] [PubMed] [Google Scholar]
  • 101. Varol E, Aksoy F, Bas HA, Ari H, Ozaydin M. Mean platelet volume is elevated in patients with low high-density lipoprotein cholesterol. Angiology 2014;65:733–736. [DOI] [PubMed] [Google Scholar]
  • 102. Guthikonda S, Alviar CL, Vaduganathan M, Arikan M, Tellez A, DeLao T, Granada JF, Dong JF, Kleiman NS, Lev EI. Role of reticulated platelets and platelet size heterogeneity on platelet activity after dual antiplatelet therapy with aspirin and clopidogrel in patients with stable coronary artery disease. J Am Coll Cardiol 2008;52:743–749. [DOI] [PubMed] [Google Scholar]
  • 103. Guthikonda S, Lev EI, Patel R, DeLao T, Bergeron AL, Dong JF, Kleiman NS. Reticulated platelets and uninhibited COX-1 and COX-2 decrease the antiplatelet effects of aspirin. J Thromb Haemost 2007;5:490–496. [DOI] [PubMed] [Google Scholar]
  • 104. Vaduganathan M, Alviar CL, Arikan ME, Tellez A, Guthikonda S, DeLao T, Granada JF, Kleiman NS, Ballantyne CM, Lev EI. Platelet reactivity and response to aspirin in subjects with the metabolic syndrome. Am Heart J 2008;156:1002 e1–1002 e7. [DOI] [PubMed] [Google Scholar]
  • 105. Tschoepe D, Roesen P, Esser J, Schwippert B, Nieuwenhuis HK, Kehrel B, Gries FA. Large platelets circulate in an activated state in diabetes mellitus. Semin Thromb Hemost 1991;17:433–438. [DOI] [PubMed] [Google Scholar]
  • 106. Papanas N, Symeonidis G, Maltezos E, Mavridis G, Karavageli E, Vosnakidis T, Lakasas G. Mean platelet volume in patients with type 2 diabetes mellitus. Platelets 2004;15:475–478. [DOI] [PubMed] [Google Scholar]
  • 107. Brown AS, Hong Y, de Belder A, Beacon H, Beeso J, Sherwood R, Edmonds M, Martin JF, Erusalimsky JD. Megakaryocyte ploidy and platelet changes in human diabetes and atherosclerosis. Arterioscler Thromb Vasc Biol 1997;17:802–807. [DOI] [PubMed] [Google Scholar]
  • 108. Muscari A, De Pascalis S, Cenni A, Ludovico C, Castaldini N, Antonelli S, Bianchi G, Magalotti D, Zoli M. Determinants of mean platelet volume (MPV) in an elderly population: relevance of body fat, blood glucose and ischaemic electrocardiographic changes. Thromb Haemost 2008;99:1079–1084. [DOI] [PubMed] [Google Scholar]
  • 109. Nicolucci A, Standl E. Antiplatelet therapy for every diabetic person? Diabetes Care 2011;34(Suppl. 2):S150–S154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Angiolillo DJ. Antiplatelet therapy in diabetes: efficacy and limitations of current treatment strategies and future directions. Diabetes Care 2009;32:531–540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Han JY, Choi DH, Choi SW, Kim BB, Ki YJ, Chung JW, Koh YY, Chang KS, Hong SP. Stroke or coronary artery disease prediction from mean platelet volume in patients with type 2 diabetes mellitus. Platelets 2013;24:401–406. [DOI] [PubMed] [Google Scholar]
  • 112. Shah B, Sha D, Xie D, Mohler ER 3rd, Berger JS. The relationship between diabetes, metabolic syndrome, and platelet activity as measured by mean platelet volume: the National Health And Nutrition Examination Survey, 1999–2004. Diabetes Care 2012;35:1074–1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Coban E, Ozdogan M, Yazicioglu G, Akcit F. The mean platelet volume in patients with obesity. Int J Clin Pract 2005;59:981–982. [DOI] [PubMed] [Google Scholar]
  • 114. Coban E, Yilmaz A, Sari R. The effect of weight loss on the mean platelet volume in obese patients. Platelets 2007;18:212–216. [DOI] [PubMed] [Google Scholar]
  • 115. Sokunbi DO, Wadhwa NK, Suh H. Vascular disease outcome and thrombocytosis in diabetic and nondiabetic end-stage renal disease patients on peritoneal dialysis. Adv Perit Dial 1994;10:77–80. [PubMed] [Google Scholar]
  • 116. Kumagai N, Mitsutake R, Miura S, Kawamura A, Takamiya Y, Nishikawa H, Uehara Y, Saku K. Acute coronary syndrome associated with essential thrombocythemia. J Cardiol 2009;54:485–489. [DOI] [PubMed] [Google Scholar]
  • 117. Natelson EA. Extreme thrombocytosis and cardiovascular surgery: risks and management. Tex Heart Inst J 2012;39:792–798. [PMC free article] [PubMed] [Google Scholar]
  • 118. Saif MW, Khan U, Greenberg BR. Cardiovascular manifestations of myeloporliferative disorders: a review of the liturature. Hospital Physician 1999;60:43–54. [PMC free article] [PubMed] [Google Scholar]
  • 119. Scheffer MG, Michiels JJ, Simoons ML, Roelandt JR. Thrombocythemia and coronary artery disease. Am Heart J 1991;122:573–576. [DOI] [PubMed] [Google Scholar]
  • 120. Landolfi R, Marchioli R, Kutti J, Gisslinger H, Tognoni G, Patrono C, Barbui T, European Collaboration on Low-Dose Aspirin in Polycythemia Vera I. Efficacy and safety of low-dose aspirin in polycythemia vera. N Engl J Med 2004;350:114–124. [DOI] [PubMed] [Google Scholar]
  • 121. Carobbio A, Finazzi G, Guerini V, Spinelli O, Delaini F, Marchioli R, Borrelli G, Rambaldi A, Barbui T. Leukocytosis is a risk factor for thrombosis in essential thrombocythemia: interaction with treatment, standard risk factors, and Jak2 mutation status. Blood 2007;109:2310–2313. [DOI] [PubMed] [Google Scholar]
  • 122. De Stefano V, Za T, Rossi E, Vannucchi AM, Ruggeri M, Elli E, Mico C, Tieghi A, Cacciola RR, Santoro C, Gerli G, Guglielmelli P, Pieri L, Scognamiglio F, Rodeghiero F, Pogliani EM, Finazzi G, Gugliotta L, Leone G, Barbui T, Party GCMNW Leukocytosis is a risk factor for recurrent arterial thrombosis in young patients with polycythemia vera and essential thrombocythemia. Am J Hematol 2010;85:97–100. [DOI] [PubMed] [Google Scholar]
  • 123. Landolfi R, Di Gennaro L, Barbui T, De Stefano V, Finazzi G, Marfisi R, Tognoni G, Marchioli R, European Collaboration on Low-Dose Aspirin in Polycythemia V. Leukocytosis as a major thrombotic risk factor in patients with polycythemia vera. Blood 2007;109:2446–2452. [DOI] [PubMed] [Google Scholar]
  • 124. Tefferi A, Barbui T. Polycythemia vera and essential thrombocythemia: 2015 update on diagnosis, risk-stratification and management. Am J Hematol 2015;90:162–173. [DOI] [PubMed] [Google Scholar]
  • 125. Marchioli R, Finazzi G, Specchia G, Cacciola R, Cavazzina R, Cilloni D, De Stefano V, Elli E, Iurlo A, Latagliata R, Lunghi F, Lunghi M, Marfisi RM, Musto P, Masciulli A, Musolino C, Cascavilla N, Quarta G, Randi ML, Rapezzi D, Ruggeri M, Rumi E, Scortechini AR, Santini S, Scarano M, Siragusa S, Spadea A, Tieghi A, Angelucci E, Visani G, Vannucchi AM, Barbui T, Group C-PC. Cardiovascular events and intensity of treatment in polycythemia vera. N Engl J Med 2013;368:22–33. [DOI] [PubMed] [Google Scholar]
  • 126. Arellano-Rodrigo E, Alvarez-Larran A, Reverter JC, Villamor N, Colomer D, Cervantes F. Increased platelet and leukocyte activation as contributing mechanisms for thrombosis in essential thrombocythemia and correlation with the JAK2 mutational status. Haematologica 2006;91:169–175. [PubMed] [Google Scholar]
  • 127. Falanga A, Marchetti M, Vignoli A, Balducci D, Barbui T. Leukocyte-platelet interaction in patients with essential thrombocythemia and polycythemia vera. Exp Hematol 2005;33:523–530. [DOI] [PubMed] [Google Scholar]
  • 128. Falanga A, Marchetti M, Vignoli A, Balducci D, Russo L, Guerini V, Barbui T. V617F JAK-2 mutation in patients with essential thrombocythemia: relation to platelet, granulocyte, and plasma hemostatic and inflammatory molecules. Exp Hematol 2007;35:702–711. [DOI] [PubMed] [Google Scholar]
  • 129. Marchetti M, Falanga A. Leukocytosis, JAK2V617F mutation, and hemostasis in myeloproliferative disorders. Pathophysiol Haemost Thromb 2008;36:148–159. [DOI] [PubMed] [Google Scholar]
  • 130. Gieger C, Radhakrishnan A, Cvejic A, Tang W, Porcu E, Pistis G, Serbanovic-Canic J, Elling U, Goodall AH, Labrune Y, Lopez LM, Magi R, Meacham S, Okada Y, Pirastu N, Sorice R, Teumer A, Voss K, Zhang W, Ramirez-Solis R, Bis JC, Ellinghaus D, Gogele M, Hottenga JJ, Langenberg C, Kovacs P, O'Reilly PF, Shin SY, Esko T, Hartiala J, Kanoni S, Murgia F, Parsa A, Stephens J, van der Harst P, Ellen van der Schoot C, Allayee H, Attwood A, Balkau B, Bastardot F, Basu S, Baumeister SE, Biino G, Bomba L, Bonnefond A, Cambien F, Chambers JC, Cucca F, D'Adamo P, Davies G, de Boer RA, de Geus EJ, Doring A, Elliott P, Erdmann J, Evans DM, Falchi M, Feng W, Folsom AR, Frazer IH, Gibson QD, Glazer NL, Hammond C, Hartikainen AL, Heckbert SR, Hengstenberg C, Hersch M, Illig T, Loos RJ, Jolley J, Khaw KT, Kuhnel B, Kyrtsonis MC, Lagou V, Lloyd-Jones H, Lumley T, Mangino M, Maschio A, Mateo Leach I, McKnight B, Memari Y, Mitchell BD, Montgomery GW, Nakamura Y, Nauck M, Navis G, Nothlings U, Nolte IM, Porteous DJ, Pouta A, Pramstaller PP, Pullat J, Ring SM, Rotter JI, Ruggiero D, Ruokonen A, Sala C, Samani NJ, Sambrook J, Schlessinger D, Schreiber S, Schunkert H, Scott J, Smith NL, Snieder H, Starr JM, Stumvoll M, Takahashi A, Tang WH, Taylor K, Tenesa A, Lay Thein S, Tonjes A, Uda M, Ulivi S, van Veldhuisen DJ, Visscher PM, Volker U, Wichmann HE, Wiggins KL, Willemsen G, Yang TP, Hua Zhao J, Zitting P, Bradley JR, Dedoussis GV, Gasparini P, Hazen SL, Metspalu A, Pirastu M, Shuldiner AR, Joost van Pelt L, Zwaginga JJ, Boomsma DI, Deary IJ, Franke A, Froguel P, Ganesh SK, Jarvelin MR, Martin NG, Meisinger C, Psaty BM, Spector TD, Wareham NJ, Akkerman JW, Ciullo M, Deloukas P, Greinacher A, Jupe S, Kamatani N, Khadake J, Kooner JS, Penninger J, Prokopenko I, Stemple D, Toniolo D, Wernisch L, Sanna S, Hicks AA, Rendon A, Ferreira MA, Ouwehand WH, Soranzo N. New gene functions in megakaryopoiesis and platelet formation. Nature 2011;480:201–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Soranzo N, Spector TD, Mangino M, Kuhnel B, Rendon A, Teumer A, Willenborg C, Wright B, Chen L, Li M, Salo P, Voight BF, Burns P, Laskowski RA, Xue Y, Menzel S, Altshuler D, Bradley JR, Bumpstead S, Burnett MS, Devaney J, Doring A, Elosua R, Epstein SE, Erber W, Falchi M, Garner SF, Ghori MJ, Goodall AH, Gwilliam R, Hakonarson HH, Hall AS, Hammond N, Hengstenberg C, Illig T, Konig IR, Knouff CW, McPherson R, Melander O, Mooser V, Nauck M, Nieminen MS, O'Donnell CJ, Peltonen L, Potter SC, Prokisch H, Rader DJ, Rice CM, Roberts R, Salomaa V, Sambrook J, Schreiber S, Schunkert H, Schwartz SM, Serbanovic-Canic J, Sinisalo J, Siscovick DS, Stark K, Surakka I, Stephens J, Thompson JR, Volker U, Volzke H, Watkins NA, Wells GA, Wichmann HE, Van Heel DA, Tyler-Smith C, Thein SL, Kathiresan S, Perola M, Reilly MP, Stewart AF, Erdmann J, Samani NJ, Meisinger C, Greinacher A, Deloukas P, Ouwehand WH, Gieger C. A genome-wide meta-analysis identifies 22 loci associated with eight hematological parameters in the HaemGen consortium. Nat Genet 2009;41:1182–1190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. 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 2008;117:1649–1657. [DOI] [PubMed] [Google Scholar]
  • 133. Potteaux S, Gautier EL, Hutchison SB, van Rooijen N, Rader DJ, Thomas MJ, Sorci-Thomas MG, Randolph GJ. Suppressed monocyte recruitment drives macrophage removal from atherosclerotic plaques of Apoe-/- mice during disease regression. J Clin Invest 2011;121:2025–2036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Hilgendorf I, Theurl I, Gerhardt LM, Robbins CS, Weber GF, Gonen A, Iwamoto Y, Degousee N, Holderried TA, Winter C, Zirlik A, Lin HY, Sukhova GK, Butany J, Rubin BB, Witztum JL, Libby P, Nahrendorf M, Weissleder R, Swirski FK. Innate response activator B cells aggravate atherosclerosis by stimulating T helper-1 adaptive immunity. Circulation 2014;129:1677–1687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Hsu LC, Enzler T, Seita J, Timmer AM, Lee CY, Lai TY, Yu GY, Lai LC, Temkin V, Sinzig U, Aung T, Nizet V, Weissman IL, Karin M. IL-1beta-driven neutrophilia preserves antibacterial defense in the absence of the kinase IKKbeta. Nat Immunol 2011;12:144–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. 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 1998;101:2702–2710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Ueda Y, Cain DW, Kuraoka M, Kondo M, Kelsoe G. IL-1R type I-dependent hemopoietic stem cell proliferation is necessary for inflammatory granulopoiesis and reactive neutrophilia. J Immunol 2009;182:6477–6484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Wang M, Subramanian M, Abramowicz S, Murphy AJ, Gonen A, Witztum J, Welch C, Tabas I, Westerterp M, Tall AR. Interleukin-3/granulocyte macrophage colony-stimulating factor receptor promotes stem cell expansion, monocytosis, and atheroma macrophage burden in mice with hematopoietic ApoE deficiency. Arterioscler Thromb Vasc Biol 2014;34:976–984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Wang N, Lan D, Chen W, Matsuura F, Tall AR. ATP-binding cassette transporters G1 and G4 mediate cellular cholesterol efflux to high-density lipoproteins. Proc Natl Acad Sci USA 2004;101:9774–9779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. 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 2010;328:1689–1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Westerterp M, Murphy AJ, Wang M, Pagler TA, Vengrenyuk Y, Kappus MS, Gorman DJ, Nagareddy PR, Zhu X, Abramowicz S, Parks JS, Welch C, Fisher EA, Wang N, Yvan-Charvet L, Tall AR. Deficiency of ATP-binding cassette transporters A1 and G1 in macrophages increases inflammation and accelerates atherosclerosis in mice. Circ Res 2013;112:1456–1465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Robbins CS, Chudnovskiy A, Rauch PJ, Figueiredo JL, Iwamoto Y, Gorbatov R, Etzrodt M, Weber GF, Ueno T, van Rooijen N, Mulligan-Kehoe MJ, Libby P, Nahrendorf M, Pittet MJ, Weissleder R, Swirski FK. Extramedullary hematopoiesis generates Ly-6C(high) monocytes that infiltrate atherosclerotic lesions. Circulation 2012;125:364–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Rauch PJ, Chudnovskiy A, Robbins CS, Weber GF, Etzrodt M, Hilgendorf I, Tiglao E, Figueiredo JL, Iwamoto Y, Theurl I, Gorbatov R, Waring MT, Chicoine AT, Mouded M, Pittet MJ, Nahrendorf M, Weissleder R, Swirski FK. Innate response activator B cells protect against microbial sepsis. Science 2012;335:597–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. 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 2007;104:15093–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. van Nieuwenhuijze A, Koenders M, Roeleveld D, Sleeman MA, van den Berg W, Wicks IP. GM-CSF as a therapeutic target in inflammatory diseases. Mol Immunol 2013;56:675–682. [DOI] [PubMed] [Google Scholar]
  • 146. Averill LE, Meagher RC, Gerrity RG. Enhanced monocyte progenitor cell proliferation in bone marrow of hyperlipemic swine. Am J Pathol 1989;135:369–377. [PMC free article] [PubMed] [Google Scholar]
  • 147. Feldman DL, Mogelesky TC, Liptak BF, Gerrity RG. Leukocytosis in rabbits with diet-induced atherosclerosis. Arterioscler Thromb 1991;11:985–994. [DOI] [PubMed] [Google Scholar]
  • 148. Stewart RA, White HD, Kirby AC, Heritier SR, Simes RJ, Nestel PJ, West MJ, Colquhoun DM, Tonkin AM, Long-Term Intervention With Pravastatin in Ischemic Disease Study I. White blood cell count predicts reduction in coronary heart disease mortality with pravastatin. Circulation 2005;111:1756–1762. [DOI] [PubMed] [Google Scholar]
  • 149. Tolani S, Pagler TA, Murphy AJ, Bochem AE, Abramowicz S, Welch C, Nagareddy PR, Holleran S, Hovingh GK, Kuivenhoven JA, Tall AR. Hypercholesterolemia and reduced HDL-C promote hematopoietic stem cell proliferation and monocytosis: studies in mice and FH children. Atherosclerosis 2013;229:79–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Woollard KJ, Kling D, Kulkarni S, Dart AM, Jackson S, Chin-Dusting J. Raised plasma soluble P-selectin in peripheral arterial occlusive disease enhances leukocyte adhesion. Circ Res 2006;98:149–156. [DOI] [PubMed] [Google Scholar]
  • 151. Woollard KJ, Suhartoyo A, Harris EE, Eisenhardt SU, Jackson SP, Peter K, Dart AM, Hickey MJ, Chin-Dusting JP. Pathophysiological levels of soluble P-selectin mediate adhesion of leukocytes to the endothelium through Mac-1 activation. Circ Res 2008;103:1128–1138. [DOI] [PubMed] [Google Scholar]
  • 152. Woollard KJ, Lumsden NG, Andrews KL, Aprico A, Harris E, Irvine JC, Jefferis AM, Fang L, Kanellakis P, Bobik A, Chin-Dusting JP. Raised soluble P-selectin moderately accelerates atherosclerotic plaque progression. PLoS One 2014;9:e97422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Murphy AJ, Sarrazy V, Wang N, Bijl N, Abramowicz S, Westerterp M, Welch CB, Schuetz JD, Yvan-Charvet L. Deficiency of ATP-binding cassette transporter B6 in megakaryocyte progenitors accelerates atherosclerosis in mice. Arterioscler Thromb Vasc Biol 2014;34:751–758. [DOI] [PubMed] [Google Scholar]
  • 154. Murphy AJ, Woollard KJ, Hoang A, Mukhamedova N, Stirzaker RA, McCormick SP, Remaley AT, Sviridov D, Chin-Dusting J. High-density lipoprotein reduces the human monocyte inflammatory response. Arterioscler Thromb Vasc Biol 2008;28:2071–2077. [DOI] [PubMed] [Google Scholar]
  • 155. Murphy AJ, Woollard KJ, Suhartoyo A, Stirzaker RA, Shaw J, Sviridov D, Chin-Dusting 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 2011;31:1333–1341. [DOI] [PubMed] [Google Scholar]
  • 156. Calkin AC, Drew BG, Ono A, Duffy SJ, Gordon MV, Schoenwaelder SM, Sviridov D, Cooper ME, Kingwell BA, Jackson SP. Reconstituted high-density lipoprotein attenuates platelet function in individuals with type 2 diabetes mellitus by promoting cholesterol efflux. Circulation 2009;120:2095–2104. [DOI] [PubMed] [Google Scholar]
  • 157. Swirski FK, Nahrendorf M, Etzrodt M, Wildgruber M, Cortez-Retamozo V, Panizzi P, Figueiredo JL, Kohler RH, Chudnovskiy A, Waterman P, Aikawa E, Mempel TR, Libby P, Weissleder R, Pittet MJ. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science 2009;325:612–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Leuschner F, Rauch PJ, Ueno T, Gorbatov R, Marinelli B, Lee WW, Dutta P, Wei Y, Robbins C, Iwamoto Y, Sena B, Chudnovskiy A, Panizzi P, Keliher E, Higgins JM, Libby P, Moskowitz MA, Pittet MJ, Swirski FK, Weissleder R, Nahrendorf M. Rapid monocyte kinetics in acute myocardial infarction are sustained by extramedullary monocytopoiesis. J Exp Med 2012;209:123–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Dutta P, Sager HB, Stengel KR, Naxerova K, Courties G, Saez B, Silberstein L, Heidt T, Sebas M, Sun Y, Wojtkiewicz G, Feruglio PF, King K, Baker JN, van der Laan AM, Borodovsky A, Fitzgerald K, Hulsmans M, Hoyer F, Iwamoto Y, Vinegoni C, Brown D, Di Carli M, Libby P, Hiebert SW, Scadden DT, Swirski FK, Weissleder R, Nahrendorf M. Myocardial infarction activates CCR2(+) hematopoietic stem and progenitor cells. Cell Stem Cell 2015;16:477–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Si Y, Tsou CL, Croft K, Charo IF. CCR2 mediates hematopoietic stem and progenitor cell trafficking to sites of inflammation in mice. J Clin Invest 2010;120:1192–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. King KY, Goodell MA. Inflammatory modulation of HSCs: viewing the HSC as a foundation for the immune response. Nat Rev Immunol 2011;11:685–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Nagai Y, Garrett KP, Ohta S, Bahrun U, Kouro T, Akira S, Takatsu K, Kincade PW. Toll-like receptors on hematopoietic progenitor cells stimulate innate immune system replenishment. Immunity 2006;24:801–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Arslan F, de Kleijn DP, Pasterkamp G. Innate immune signaling in cardiac ischemia. Nat Rev Cardiol 2011;8:292–300. [DOI] [PubMed] [Google Scholar]
  • 164. Morrow DA, Wang Y, Croce K, Sakuma M, Sabatine MS, Gao H, Pradhan AD, Healy AM, Buros J, McCabe CH, Libby P, Cannon CP, Braunwald E, Simon DI. Myeloid-related protein 8/14 and the risk of cardiovascular death or myocardial infarction after an acute coronary syndrome in the Pravastatin or Atorvastatin Evaluation and Infection Therapy: Thrombolysis in Myocardial Infarction (PROVE IT-TIMI 22) trial. Am Heart J 2008;155:49–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Katashima T, Naruko T, Terasaki F, Fujita M, Otsuka K, Murakami S, Sato A, Hiroe M, Ikura Y, Ueda M, Ikemoto M, Kitaura Y. Enhanced expression of the S100A8/A9 complex in acute myocardial infarction patients. Circ J 2010;74:741–748. [DOI] [PubMed] [Google Scholar]
  • 166. Sager HB, Heidt T, Hulsmans M, Dutta P, Courties G, Sebas M, Wojtkiewicz GR, Tricot B, Iwamoto Y, Sun Y, Weissleder R, Libby P, Swirski FK, Nahrendorf M. Targeting interleukin-1beta reduces leukocyte production after acute myocardial infarction. Circulation 2015;132:1880–1890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Nagareddy PR, Kraakman M, Masters SL, Stirzaker RA, Gorman DJ, Grant RW, Dragoljevic D, Hong ES, Abdel-Latif A, Smyth SS, Choi SH, Korner J, Bornfeldt KE, Fisher EA, Dixit VD, Tall AR, Goldberg IJ, Murphy AJ. Adipose tissue macrophages promote myelopoiesis and monocytosis in obesity. Cell Metab 2014;19:821–835. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Westerterp M, Gourion-Arsiquaud S, Murphy AJ, Shih A, Cremers S, Levine RL, Tall AR, Yvan-Charvet L. Regulation of hematopoietic stem and progenitor cell mobilization by cholesterol efflux pathways. Cell Stem Cell 2012;11:195–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Fossiez F, Djossou O, Chomarat P, Flores-Romo L, Ait-Yahia S, Maat C, Pin JJ, Garrone P, Garcia E, Saeland S, Blanchard D, Gaillard C, Das Mahapatra B, Rouvier E, Golstein P, Banchereau J, Lebecque S. T cell interleukin-17 induces stromal cells to produce proinflammatory and hematopoietic cytokines. J Exp Med 1996;183:2593–2603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Stark MA, Huo Y, Burcin TL, Morris MA, Olson TS, Ley K. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity 2005;22:285–294. [DOI] [PubMed] [Google Scholar]
  • 171. Haffner SM, Stern MP, Hazuda HP, Mitchell BD, Patterson JK. Cardiovascular risk factors in confirmed prediabetic individuals. Does the clock for coronary heart disease start ticking before the onset of clinical diabetes? JAMA 1990;263:2893–2898. [DOI] [PubMed] [Google Scholar]
  • 172. Lind M, Svensson AM, Kosiborod M, Gudbjornsdottir S, Pivodic A, Wedel H, Dahlqvist S, Clements M, Rosengren A. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med 2014;371:1972–1982. [DOI] [PubMed] [Google Scholar]
  • 173. Nathan DM. Long-term complications of diabetes mellitus. N Engl J Med 1993;328:1676–1685. [DOI] [PubMed] [Google Scholar]
  • 174. Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, Orchard TJ, Raskin P, Zinman B, Diabetes C, Complications Trial/Epidemiology of Diabetes I, Complications Study Research Group. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N Engl J Med 2005;353:2643–2653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Ford ES. Leukocyte count, erythrocyte sedimentation rate, and diabetes incidence in a national sample of US adults. Am J Epidemiol 2002;155:57–64. [DOI] [PubMed] [Google Scholar]
  • 176. Kullo IJ, Hensrud DD, Allison TG. Comparison of numbers of circulating blood monocytes in men grouped by body mass index (<25, 25 to <30, > or =30). Am J Cardiol 2002;89:1441–1443. [DOI] [PubMed] [Google Scholar]
  • 177. Ohshita K, Yamane K, Hanafusa M, Mori H, Mito K, Okubo M, Hara H, Kohno N. Elevated white blood cell count in subjects with impaired glucose tolerance. Diabetes Care 2004;27:491–496. [DOI] [PubMed] [Google Scholar]
  • 178. Orchard TJ, Olson JC, Erbey JR, Williams K, Forrest KY, Smithline Kinder L, Ellis D, Becker DJ. Insulin resistance-related factors, but not glycemia, predict coronary artery disease in type 1 diabetes: 10-year follow-up data from the Pittsburgh Epidemiology of Diabetes Complications Study. Diabetes Care 2003;26:1374–1379. [DOI] [PubMed] [Google Scholar]
  • 179. Persson SU, Larsson H, Odeberg H. Reduced number of circulating monocytes after institution of insulin therapy—relevance for development of atherosclerosis in diabetics? Angiology 1998;49:423–433. [DOI] [PubMed] [Google Scholar]
  • 180. Poitou C, Dalmas E, Renovato M, Benhamo V, Hajduch F, Abdennour M, Kahn JF, Veyrie N, Rizkalla S, Fridman WH, Sautes-Fridman C, Clement K, Cremer I. CD14dimCD16+ and CD14+CD16+ monocytes in obesity and during weight loss: relationships with fat mass and subclinical atherosclerosis. Arterioscler Thromb Vasc Biol 2011;31:2322–2330. [DOI] [PubMed] [Google Scholar]
  • 181. Schmidt MI, Duncan BB, Sharrett AR, Lindberg G, Savage PJ, Offenbacher S, Azambuja MI, Tracy RP, Heiss G. Markers of inflammation and prediction of diabetes mellitus in adults (Atherosclerosis Risk in Communities study): a cohort study. Lancet 1999;353:1649–1652. [DOI] [PubMed] [Google Scholar]
  • 182. Vuckovic S, Withers G, Harris M, Khalil D, Gardiner D, Flesch I, Tepes S, Greer R, Cowley D, Cotterill A, Hart DN. Decreased blood dendritic cell counts in type 1 diabetic children. Clin Immunol 2007;123:281–288. [DOI] [PubMed] [Google Scholar]
  • 183. Woo SJ, Ahn SJ, Ahn J, Park KH, Lee K. Elevated systemic neutrophil count in diabetic retinopathy and diabetes: a hospital-based cross-sectional study of 30,793 Korean subjects. Invest Ophthalmol Vis Sci 2011;52:7697–7703. [DOI] [PubMed] [Google Scholar]
  • 184. Parathath S, Grauer L, Huang LS, Sanson M, Distel E, Goldberg IJ, Fisher EA. Diabetes adversely affects macrophages during atherosclerotic plaque regression in mice. Diabetes 2011;60:1759–1769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Ceriello A. Postprandial hyperglycemia and diabetes complications: is it time to treat? Diabetes 2005;54:1–7. [DOI] [PubMed] [Google Scholar]
  • 186. 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 2008;205:2409–2417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Giacco F, Du X, Carratu A, Gerfen GJ, D'Apolito M, Giardino I, Rasola A, Marin O, Divakaruni AS, Murphy AN, Shah MS, Brownlee M. GLP-1 cleavage product reverses persistent ROS generation after transient hyperglycemia by disrupting an ROS-generating feedback loop. Diabetes 2015;64:3273–3284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Zinman B, Inzucchi SE, Lachin JM, Wanner C, Ferrari R, Fitchett D, Bluhmki E, Hantel S, Kempthorne-Rawson J, Newman J, Johansen OE, Woerle HJ, Broedl UC. Rationale, design, and baseline characteristics of a randomized, placebo-controlled cardiovascular outcome trial of empagliflozin (EMPA-REG OUTCOME). Cardiovasc Diabetol 2014;13:102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 2015;373:2117–2128. [DOI] [PubMed] [Google Scholar]
  • 190. Tang C, Kanter JE, Bornfeldt KE, Leboeuf RC, Oram JF. Diabetes reduces the cholesterol exporter ABCA1 in mouse macrophages and kidneys. J Lipid Res 2010;51:1719–1728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Mauldin JP, Srinivasan S, Mulya A, Gebre A, Parks JS, Daugherty A, Hedrick CC. Reduction in ABCG1 in Type 2 diabetic mice increases macrophage foam cell formation. J Biol Chem 2006;281:21216–21224. [DOI] [PubMed] [Google Scholar]
  • 192. Distel E, Barrett TJ, Chung K, Girgis NM, Parathath S, Essau CC, Murphy AJ, Moore KJ, Fisher EA. miR33 inhibition overcomes deleterious effects of diabetes mellitus on atherosclerosis plaque regression in mice. Circ Res 2014;115:759–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Goedeke L, Salerno A, Ramirez CM, Guo L, Allen RM, Yin X, Langley SR, Esau C, Wanschel A, Fisher EA, Suarez Y, Baldan A, Mayr M, Fernandez-Hernando C. Long-term therapeutic silencing of miR-33 increases circulating triglyceride levels and hepatic lipid accumulation in mice. EMBO Mol Med 2014;6:1133–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Niesor EJ, Schwartz GG, Perez A, Stauffer A, Durrwell A, Bucklar-Suchankova G, Benghozi R, Abt M, Kallend D. Statin-induced decrease in ATP-binding cassette transporter A1 expression via microRNA33 induction may counteract cholesterol efflux to high-density lipoprotein. Cardiovasc Drugs Ther 2015;29:7–14. [DOI] [PubMed] [Google Scholar]
  • 195. Zhou J, Xu D, Xie H, Tang J, Liu R, Li J, Wang S, Chen X, Su J, Zhou X, Xia K, He Q, Chen J, Xiong W, Cao P, Cao K. miR-33a functions as a tumor suppressor in melanoma by targeting HIF-1alpha. Cancer Biol Ther 2015;16:846–855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Singer K, DelProposto J, Morris DL, Zamarron B, Mergian T, Maley N, Cho KW, Geletka L, Subbaiah P, Muir L, Martinez-Santibanez G, Lumeng CN. Diet-induced obesity promotes myelopoiesis in hematopoietic stem cells. Mol Metab 2014;3:664–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. 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 2011;162:597–605. [DOI] [PubMed] [Google Scholar]
  • 198. Masters SL, Latz E, O'Neill LA. The inflammasome in atherosclerosis and type 2 diabetes. Sci Transl Med 2011;3:81ps17. [DOI] [PubMed] [Google Scholar]
  • 199. Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, Ravussin E, Stephens JM, Dixit VD. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med 2011;17:179–188. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from European Heart Journal are provided here courtesy of Oxford University Press

RESOURCES