Despite their small size and anucleate status, platelets have diverse roles in vascular biology. Not only are platelets the cellular mediator of thrombosis, but platelets are also immune cells that initiate and accelerate many vascular inflammatory conditions. Platelets are linked to the pathogenesis of inflammatory diseases such as atherosclerosis, malaria infection, transplant rejection, and rheumatoid arthritis. In some contexts, platelet immune functions are protective, whereas in others platelets contribute to adverse inflammatory outcomes. In this review, we will discuss platelet and platelet-derived mediator interactions with the innate and acquired arms of the immune system and platelet-vessel wall interactions that drive inflammatory disease. There have been many recent publications indicating both important protective and adverse roles for platelets in infectious disease. Because of this new accumulating data, and the fact that infectious disease continues to be a leading cause of death globally, we will also focus on new and emerging concepts related to platelet immune and inflammatory functions in the context of infectious disease.

Platelets are best known as the cellular mediator of thrombosis. There is now a growing appreciation of the important immune and inflammatory roles of platelets in both health and disease. A number of studies have demonstrated that platelets impact inflammatory processes ranging from atherosclerosis to infectious diseases, making platelets the most numerous circulating cell type that has an immune function. Platelets interact with white blood cells and vascular endothelial cells both directly by contact-dependent mechanisms and indirectly through secreted immune mediator-driven mechanisms. Platelet immune effects are therefore noted both locally at sites of platelet activation and deposition or systemically at locations distant from platelet activation itself.1,2  Platelet interactions with inflammatory cells may mediate proinflammatory outcomes, but these interactions have likely evolved to be beneficial in limiting infection. For example, with a breach in the skin there is exposure to pathogens, and by combining thrombotic and immune recruitment functions, platelets may help focus hemostasis and immune responses against potential infectious agents to prevent pathogen invasion. However, continued or chronic platelet interactions with white blood cells or endothelial cells can lead to adverse effects from excessive immune stimulation and inflammatory insult.

Platelets are small in size (∼4 µm), but because they are present in large numbers (≈200 000/µL blood in humans) and have many preformed inflammatory molecules and immune mediators, platelets exert inflammatory effects on a scale that exceeds their individual size. A platelet contains ∼60 granules that store many molecules with immune functions (Table 1, 2-59  partial list of granule contents with inflammatory/immune roles).60  There are 3 types of platelet granules: α-granules, dense granules, and lysomal granules. A recent report has described a possible new type of granule termed a T-granule.61  α-Granules are the most numerous (50-60 per platelet) and largest (200-400 nm) platelet granule and store a large variety of proteins. One proteomic analysis of α-granule content found 284 proteins.62  Dense granules are smaller (∼150 nm), less numerous (3-8 per platelet), and store small molecules (Table 1). Lysosomal granules are sparse and contain glycohydrolases and degradative enzymes.63-66  Upon platelet stimulation, granules undergo regulated exocytosis and release their contents into the extracellular environment, or molecules found on the inner granule membrane become surface expressed. Although granule exocytosis contributes to platelet activation and thrombus formation, many granule-derived mediators also have either primary or secondary roles as immune molecules.

Table 1

Partial list of platelet-derived inflammatory mediators and immune modulators

MoleculeImmune/inflammatory role
α-Granule  
 PF4 Chemokine: monocyte, neutrophil, and T-cell recruitment; Th differentiation2-5  
 Ppbp β-thromboglobulin NAP-2 Chemokine: neutrophil activation and recruitment, macrophage phagocytic activity6-8  
 P-selectin Selectin: leukocyte adhesion, complement activation9-11  
 CD40L TNF superfamily: antigen-presenting cell activation, B-cell responses, endothelial cell activation12-14  
 TGF-β Cytokine: cell proliferation, T-cell differentiation, B-cell and macrophage phenotype regulation15-19  
 PDGF Growth factor: cell growth and differentiation, monocyte/macrophage differentiation20,21  
 VWF Platelet adhesion, PMN extravasation22,23  
 CD63 Tetraspanin: transmembrane adaptor protein, leukocyte recruitment24  
 SDF-1 Chemokine: T-cell, monocyte, and PMN chemotaxis25-27  
 VEGF Growth factor: angiogenesis, adhesion molecule expression28-30  
 Thrombospondins Apoptosis, endothelial cell inflammation, macrophage-platelet aggregates31,32  
 MIP-1α Cytokine: neutrophil and eosinophil activation, B-cell immunoglobulin production33,34  
 MMP-2, MMP-9 Protease: extracellular matrix breakdown, platelet-leukocyte aggregate formation35-38  
 Cyclophilin A Vascular smooth muscle cell growth factor36  
Dense granule  
 Serotonin DC and T-cell functions39,40  
 Glutamate T-cell trafficking41,42  
 Polyphosphates Inflammatory response amplification43,44  
 ADP Platelet, leukocyte, endothelial cell activation45-47  
 Histamine Increased vessel reactivity and degranulation48-50  
Produced or constitutively expressed  
  IL-1β Cytokine: acute phase response, leukocyte and endothelial activation51-54  
 Thromboxane Eicosanoid: T-cell differentiation, monocyte activation55-57  
 Nitric oxide Reactive oxygen species: anti-inflammatory and antithrombotic58  
 GPIbα Adhesion molecule: binds Mac-1 on leukocytes59  
MoleculeImmune/inflammatory role
α-Granule  
 PF4 Chemokine: monocyte, neutrophil, and T-cell recruitment; Th differentiation2-5  
 Ppbp β-thromboglobulin NAP-2 Chemokine: neutrophil activation and recruitment, macrophage phagocytic activity6-8  
 P-selectin Selectin: leukocyte adhesion, complement activation9-11  
 CD40L TNF superfamily: antigen-presenting cell activation, B-cell responses, endothelial cell activation12-14  
 TGF-β Cytokine: cell proliferation, T-cell differentiation, B-cell and macrophage phenotype regulation15-19  
 PDGF Growth factor: cell growth and differentiation, monocyte/macrophage differentiation20,21  
 VWF Platelet adhesion, PMN extravasation22,23  
 CD63 Tetraspanin: transmembrane adaptor protein, leukocyte recruitment24  
 SDF-1 Chemokine: T-cell, monocyte, and PMN chemotaxis25-27  
 VEGF Growth factor: angiogenesis, adhesion molecule expression28-30  
 Thrombospondins Apoptosis, endothelial cell inflammation, macrophage-platelet aggregates31,32  
 MIP-1α Cytokine: neutrophil and eosinophil activation, B-cell immunoglobulin production33,34  
 MMP-2, MMP-9 Protease: extracellular matrix breakdown, platelet-leukocyte aggregate formation35-38  
 Cyclophilin A Vascular smooth muscle cell growth factor36  
Dense granule  
 Serotonin DC and T-cell functions39,40  
 Glutamate T-cell trafficking41,42  
 Polyphosphates Inflammatory response amplification43,44  
 ADP Platelet, leukocyte, endothelial cell activation45-47  
 Histamine Increased vessel reactivity and degranulation48-50  
Produced or constitutively expressed  
  IL-1β Cytokine: acute phase response, leukocyte and endothelial activation51-54  
 Thromboxane Eicosanoid: T-cell differentiation, monocyte activation55-57  
 Nitric oxide Reactive oxygen species: anti-inflammatory and antithrombotic58  
 GPIbα Adhesion molecule: binds Mac-1 on leukocytes59  

ADP, adenosine 5′-diphosphate; CD40L, CD40 ligand; DC, dendritic cell; GPIbα, glycoprotein Ibα; IL, interleukin; MIP, macrophage-inflammatory protein; MMP, metalloproteinase; NAP, neutrophil-activating peptide; PDGF, platelet-derived growth factor; PF4, platelet factor 4; PMN, neutrophil; ppbp, proplatelet basic protein; SDF, stromal cell–derived factor; TGF, transforming growth factor; Th, T helper; TNF, tumor necrosis factor; VEGF, vascular endothelial growth factor; VWF, von Willebrand factor.

α-Granule constituents such as PF4, regulated on activation normal T expressed and secreted (RANTES), SDF-1, and ppbp have limited thrombotic functions and instead are chemokines and cytokines that recruit and activate other immune cells or induce endothelial cell inflammation. The inflammatory roles of most α-granule–derived chemokines, cytokines, and adhesion molecules are well described.67,68  Dense granule constituents such as ADP, serotonin, polyphosphates, and glutamate are more understood as modifiers of platelet activation and thrombus formation, but many have immune cell–modifying effects. DCs express the P2Y12 ADP receptor, and DC P2Y12 activation increases antigen endocytosis and processing.69  Adenosine triphosphate signaling through T-cell P2X7 increases differentiation of CD4+ Th cells toward a proinflammatory Th17 cell type.70  Polyphosphates induce nuclear factor κB activation and the expression of endothelial adhesion molecules,44  and polyphosphates also amplify high-mobility group protein B1–mediated inflammatory signaling through interactions with the receptor for advanced glycosylation endproducts and P2Y1 receptors.43  Platelet dense granules contain glutamate, and glutamate in the periphery can induce T-cell migration.71,72  Platelets are the peripheral source of serotonin, and serotonin increases monocyte differentiation into DCs73  and early naïve T-cell activation.40  Although these dense granule–derived immune modifiers are highly enriched in platelets and have immune functions, the direct contribution of platelet dense granule constituents to immune responses in a disease context is still largely unexplored.

The number of potential interactions, both direct and indirect, between platelets and other cells is extensive, and as a result, a multitude of inflammatory effects can be exerted by platelets both in the local environment and systemically.

The acute phase response (APR) is the earliest response to infection or vascular injury. The APR is typified by the production of acute phase proteins such as C-reactive protein, serum amyloids A and P, complement proteins, and fibrinogen by the liver. Acute phase proteins destroy or inhibit the growth of microbes and exert procoagulant effects that may limit infection by trapping pathogens within local blood clots. Our studies have indicated that platelets induce the APR.51  Following vascular compromise, it may be advantageous to activate the APR simultaneous with platelet activation to localize and limit systemic invasion of potential microbes. interleukin-1β (IL-1β), IL-6, and IL-8 are all potent inducers of the APR. Platelets are not a direct source of IL-6 or IL-8 but are a major source of IL-1β.51  Because IL-1β is not granule stored and is produced upon platelet stimulation, it was surprising to discover IL-1β in platelets, and more unexpected to find pre–messenger RNA (mRNA) for IL-1β expressed by platelets.53  With platelet stimulation, IL-1β pre-mRNA is spliced, IL-1β mRNA translated into pro-IL-1β, and caspase-1 processed, resulting in the release of functional IL-1β.53  Typical markers of platelet activation (granule exocytosis and integrin activation) are increased rapidly after platelet stimulation (seconds to minutes), but the release of IL-1β from stimulated platelets occurs over hours.74  Platelets contain other mRNAs and pre-mRNAs, some of which are used to make proteins after platelet stimulation, but IL-1β is the best described.75  In a mouse model of severe malaria, we found that platelets are activated early postinfection, and platelet-derived IL-1β has a major role in inducing the APR.51  Platelets were also found localized to hepatic sinusoids following Plasmodium infection indicating that platelets may induce the APR in a contact-dependent manner (Figures 12).

Figure 1

Schematic of platelet-derived immune mediators. 5-HT, serotonin; ADP, adenosine 5′-diphosphate; COX-1, cyclooxygenase 1; P-selectin glycoprotein ligand-1; TbxA, thromboxane; WBC, white blood cell; PSGL-1.

Figure 1

Schematic of platelet-derived immune mediators. 5-HT, serotonin; ADP, adenosine 5′-diphosphate; COX-1, cyclooxygenase 1; P-selectin glycoprotein ligand-1; TbxA, thromboxane; WBC, white blood cell; PSGL-1.

Close modal
Figure 2

Platelet and liver sinusoid interactions in a Plasmodium berghei–infected mouse (* denotes platelets).

Figure 2

Platelet and liver sinusoid interactions in a Plasmodium berghei–infected mouse (* denotes platelets).

Close modal

Toll-like receptors (TLRs) are a highly conserved family of pattern recognition receptors expressed by organisms from flies to mammals. TLRs bind pathogen-associated molecular pattern molecules that are broadly expressed by many infectious organisms. Lipopolysaccharide (LPS) is the most studied TLR ligand, and others include unmethylated cytosine guanine dinucleotide, double-stranded RNA, and lipoproteins. Platelets express numerous TLR family members including TLR1, TLR2, TLR4, TLR6, TLR8, and TLR9.76  Because gram-negative sepsis is a major clinical problem with few therapeutic options, much focus has been on the implications and mechanisms of LPS signaling via platelet TLR4 in sepsis-associated thrombocytopenia and platelet activation and accumulation in organs such as the lung.77  CD14 is a coreceptor for LPS signaling through TLR4, but CD14 itself may not be expressed by platelets,78  meaning platelets are dependent on other sources of soluble CD14 to respond to LPS. Platelet TLR4 signaling leads to platelet activation, the shedding of IL-1β-rich microparticles (MPs), and platelet interactions with other cells.74  TLR4-stimulated platelet interactions with adherent neutrophils lead to the formation of neutrophil extracellular traps that may trap bacteria.79  TLR2 also has described roles in platelet inflammatory functions. Stimulation of platelet TLR2 by Porphyromonas gingivalis results in increased platelet-neutrophil adhesion,80  and stimulation of megakaryocytes through TLR2 results in the production of platelets with an increased proinflammatory gene and protein expression.81  Other TLRs, pattern recognition receptors, and scavenger receptors also have functions in platelet activation and thrombosis, including TLR9 and CD36, but their role in inflammation and platelet immune cell interactions remains to be determined.82-84 

Monocytes and neutrophils are the most numerous innate immune cells in the blood. Monocytes have major roles in chronic cardiovascular diseases including atherosclerosis. Some of the earliest studies of monocyte interactions with platelets focused on platelet phagocytosis by monocytes within thrombi and speculated that this may contribute to atherosclerotic lesion development.85  The identification of interactions between platelets and monocytes via platelet P-selectin has historical significance, in part because P-selectin is a mediator of platelet and monocyte interactions, and in part because the recognition of this interaction helped accelerate discovery of other platelet immune functions. P-selectin was once alternatively termed platelet activation-dependent granule-external membrane protein or granule membrane protein 140. In 1985, granule membrane protein 140 was shown to be expressed on the platelet plasma membrane after activation,86  setting the stage for other studies demonstrating that P-selectin mediates platelet and leukocyte interactions. This included the in vivo demonstration in a primate arteriovenous shunt model that adherent platelets express P-selectin and platelet P-selectin immobilizes leukocytes at the site of the lesion,10,87  establishing an important framework for subsequent in vivo and disease-focused studies. The generation of the first P-selectin knockout mouse provided genetic proof of a mechanistic role for platelet interactions with leukocytes and the vessel wall.11  The interaction of platelet P-selectin with its receptor on monocytes, PSGL-1, does more than just localize monocytes. P-selectin and PSGL-1 interactions induce tissue factor–bearing MP formation and monocyte proinflammatory changes.88 

With the identification of critical links between platelets and monocytes and insights into the association between monocytes and atherosclerosis, an expansion of our understanding of platelets as immune mediators in vascular inflammatory diseases has rapidly emerged. The increase in numbers of commercially available genetically modified mice has also facilitated studies focused on platelet and monocyte interactions in atherosclerosis, an inflammatory disease driven in large part by immune cells.89,90  Early investigations into the pathogenesis of atherosclerosis suggested that platelets induced an endothelial cell inflammatory response91,92  and platelet-derived mediators increased endothelial permeability permitting lipid entry into the vessel wall and atherosclerosis development.93,94  Mouse models have provided evidence that platelet-secreted inflammatory molecules such as RANTES and PF4 localize inflammatory molecules to sites of vascular inflammation, accelerating lesion development.95-97  Human studies have identified increased platelet and monocyte aggregates in the circulation of those with atherosclerosis, perhaps accelerating the immune response to vascular lesions and atherosclerotic progression.98-101  Because of the great health impact of atherosclerosis and the role of innate immune cells in its pathogenesis, studies are likely to continue to define the role of platelets in innate immunity.

Although the role of platelets in innate immune responses has received much more attention, platelets also influence acquired immune responses, including T-cell trafficking, activation, and differentiation. T cells are broadly divided into CD8+ or CD4+ cells, and CD4+ T cells further divided into the Th types Th1, Th2, or Th17 as immune effectors and T regulatory cells as immune suppressors. CD8+ T cells are often termed cytotoxic T cells. They recognize antigen presented in major histocompatibility complex class I leading to infected cell death through CD8+ T-cell cytokine secretion. CD4+ cells are master regulators of immune responses. CD4+ cells recognize antigen presented by major histocompatibility complex class II and release cytokines that regulate the activity of other immune cells, such as B cells and innate immune cells. T-cell trafficking to sites of infection or inflammation is an important step in T-cell responses. Chemokine and cytokine gradients and increased expression of T-cell chemokine receptors lead to T-cell trafficking. CXC chemokine receptor (CXCR) 3 is the chemokine receptor highly expressed on activated Th1 cells. CXCR3 ligands include CXC chemokine ligand 10 and PF4.102,103  We have found in a mouse model of cerebral malaria that PF4−/− mice have decreased expression of T-cell CXCR3 indicating that PF4 may directly or indirectly mediate CXCR3 expression and T-cell trafficking.2  Other platelet-derived chemokines recruit and activate T cells at sites of vascular inflammation. For example, CC chemokine receptor 5 is a T-cell receptor for both MIP-1α and RANTES (CC chemokine ligand 5), each of which is present at high concentrations in α-granules. T cells may activate platelets through a T-cell CD40L/platelet CD40 interaction leading to platelet RANTES release and further T-cell recruitment.104  Using an acute skin graft model, we have found that platelets increase T-cell graft infiltrates and rejection, in part via platelet-derived thromboxane.41,105  Prostaglandins such as thromboxane also facilitate Th1 differentiation and Th17 expansion contributing to the development of inflammatory diseases,56  but the relevance of platelet-derived prostaglandins to T-cell development and differentiation remains to be determined.

Platelets are the major source of soluble CD40L (soluble CD40L/soluble CD154). Platelet CD40L is stored in α-granules, and with activation CD40L becomes expressed on the platelet surface or released in soluble form into the extracellular environment. In a cardiac allograft model, platelet-derived soluble CD40L was sufficient to initiate rejection independent of other CD40L cell sources.14  Platelet-derived CD40L can also be delivered by MPs increasing the number and distance of interactions between platelets and other cells.106  Platelet CD40L augments T-cell immunity to viral challenge and is necessary for optimal production of immunoglobulin G by inducing DC maturation and B-cell isotype switching.107,108  It has also been suggested that when antigen-specific B and T cells are rare, platelets enhance signals needed for adaptive humoral immunity and germinal center formation.108 

We have recently discovered in a mouse cardiac transplant model that platelets have a central role in maintaining CD4+ Th cell homeostasis and regulating Th differentiation into effector Th subtypes.109  Platelet-derived PF4 is needed to limit Th17 differentiation and expansion, both basally and following immune stimulation. PF4 effects on Th differentiation may be mediated in part by PF4 limiting TGF-β signaling. These findings greatly expand our understanding of how platelets shape acquired immune development and indicate that platelets help maintain basal Th cell balance.

Platelets influence adaptive immunity through modulation of DC functions. Platelets may recruit and activate DCs through interactions between DC-derived CD11b/CD18 (Mac-1) and platelet junctional adhesion molecule C, thereby increasing DC activation.110  DC expression of T-cell costimulatory molecules CD80 and CD86 is increased by activated platelets in a contact-independent manner111  leading to a stronger and more rapid T-cell response. Platelets also direct pathogen delivery to DCs. Listeria monocytogenes associates with platelets in the blood in a GPIb- and complement-dependent manner leading to targeting of the platelet-Listeria complexes to splenic CD8α+ DCs.112  The result may be a directing of bacterial clearance away from less immunogenic phagocytes to the more immunologically active CD8α+ DCs.

Although it is clear that platelets affect all phases of immune responses, more work remains to fully appreciate how platelets directly and indirectly affect acquired immune responses. Because T cells, and CD4+ T cells in particular, are master regulators of the immune system, a better understanding of platelet and T-cell interactions is likely to impact a broad range of inflammatory and immune conditions.

Because of the great health impact of atherosclerosis, much has been described regarding interactions between platelets and endothelial cells. Atherosclerosis is now clearly defined as an inflammatory disease driven by immune cell interactions with the vessel wall.113  Many reviews have described the activities of platelets at the interface with endothelial cells and atherosclerosis,114  so we will focus on emerging new investigations demonstrating that platelets are mediators of intercellular vascular communication via platelet-derived MPs, and the transfer of platelet-derived microRNA (miRNA) to other vascular cells, because each has been linked to the pathogenesis of vascular inflammation and directly impacts immune cell trafficking.

MPs are released by almost all types of cells. Activated platelets release MPs, and platelets are a major source of circulating MPs.115-117  MPs are commonly defined as lipid membrane vesicles 0.1-1 µM in size. An increased number of circulating platelet MPs (PMPs) correlates with the development of atherosclerosis in patients with diabetes,118  the amount of heart tissue at risk in recent myocardial infarction,119  and the development of acute coronary syndrome and stroke.120-122  PMPs carry adhesion molecules (such as P-selectin) and chemokines (such as RANTES), facilitating monocyte arrest at the site of PMP deposition along the vessel wall, potentially accelerating atherosclerosis.123  Developmental endothelial locus-1 contributes to endothelial uptake of PMPs because Del-1−/− mice had decreased PMP uptake and increased circulating PMPs in an LPS challenge model.124  Statin usage decreases the number of PMPs, potentially providing a potential non-lipid-lowering protective effect of statins.125  The inflammatory effects of PMPs extend to other diseases. PMPs are present at increased numbers in the joint space of individuals with rheumatoid arthritis and are proinflammatory by inducing synovial fibroblast cytokine responses in a PMP IL-1α- and IL-1β-dependent manner.54  PMP IL-1β also contributes to endothelial permeability during dengue virus infection.126,127 

MPs carry miRNAs, which are small RNAs ∼20-22 nucleotides in size that regulate gene expression. Each miRNA has the potential to alter the expression of hundreds of genes, increasing the potential impact that platelet miRNA transfer may have on cardiovascular disease. Increased circulating miRNA has been noted in both coronary artery disease and post myocardial infarction,128,129  and recent evidence has suggested that platelet-derived miR-340 and miR-624 are upregulated in patients with premature atherosclerosis.130  Platelets contain >250 different identified and quantified miRNAs.130-133  Platelet RNA and miRNAs can be functionally transferred to other cell types via PMPs. In an in vitro system, RNA from PMPs altered THP-1 (monocyte cell line) gene expression.134  Recent studies have indicated that platelets transfer miRNA in vivo by activated platelet release of PMP-associated miRNA. PMP-derived Ago2 complexes may be functional and taken up by endothelial cells, regulating endogenous endothelial cell gene expression.135  Platelets activated during myocardial infarction release miR-320b that is taken up by endothelial cells and may regulate intercellular adhesion molecule 1 expression, leukocyte adhesion, and trafficking.136  These emerging studies indicate that platelet miRNAs mediate vascular inflammatory processes in a transcellular manner.

Infectious disease continues to be a leading cause of death globally. Platelets have been associated with vascular inflammatory and immune complications of malaria, sepsis, HIV, and influenza, each of which causes a large public health burden worldwide. Our knowledge of how platelet-pathogen interactions help dictate infectious disease outcomes has rapidly expanded. Studies have indicated a complicated role for platelets in infectious disease pathogenesis; platelets may help limit the growth of many organisms, but platelet-mediated inflammatory responses may complicate disease progression.

Malaria is caused by the mosquito-transmitted Plasmodium parasite. Despite extensive research over many decades, malaria continues to be a significant cause of death globally and has a major economic impact.137  In uncomplicated malaria, symptoms are typically secondary to red blood cell (RBC) destruction and anemia. Thrombocytopenia is noted very early following infection,138-140  and platelet counts return to normal after parasite clearance.141  The cause of the thrombocytopenia (platelet activation and clearance vs spleen sequestration vs lack of production) remains unclear. We have found evidence for platelet activation within 24 hours of infection in a mouse model.51 Plasmodium-induced immune activation and inflammatory cell–mediated tissue injury can lead to vascular compromise that complicates disease progression. This is commonly referred to as severe malaria. Children are at particular risk for cerebral vascular injury following malaria infection (cerebral malaria). In one study, up to 33% of children admitted to a hospital in Kenya had malaria infection, and of those 47% had evidence of neurologic deficits142  demonstrating the clinical importance of gaining a better understanding cerebral malaria pathogenesis. Cerebral vascular thrombosis and multifocal strokelike lesions are often noted on autopsy specimens of those who die of cerebral malaria.143  Many laboratories have demonstrated a central role for platelets in initiating and accelerating this cerebral inflammatory injury,2,144-146  and there is evidence of Plasmodium invasion/uptake by platelets even though the parasite is unable to progress through its life cycle within platelets.147  Grau and van der Heyde have demonstrated that platelets are necessary for the development of experimental cerebral malaria,148-150  leading to a unifying platelet-centered model for its pathogenesis. Grau has proposed that platelet activation induces the expression of endothelial cell adhesion molecules, leading to more platelet-endothelial interactions, infected RBC and leukocyte cerebral vascular localization, and ultimately cerebral vascular compromise.146  Using the murine model of cerebral malaria, we have implicated PF4 as a platelet-derived chemokine that significantly contributes to leukocyte cerebral vascular trafficking.2,151  This has been further validated in clinical studies demonstrating that plasma PF4 is a predictive biomarker of cerebral malaria in humans.152  Other studies have found platelet-mediated endothelial cell destruction both in vitro and in vivo in malaria models, further highlighting the adverse roles for platelets in cerebral malaria.153 

There have also been reports that in uncomplicated malaria (limited inflammation and no cerebral vascular compromise) platelets may have a protective role by directly killing Plasmodium, making the role for platelets in malaria infection confusing and dependent on the inflammatory response to infection.154  Using Plasmodium parasite infection models that do not induce severe malaria or a strong immune response, platelet-deficient mice had increased parasitemia, and platelets were shown to kill intra-RBC parasites in vitro.154  The chemokine we found to be detrimental in experimental cerebral malaria, PF4, was shown to have parasite-killing effects through its antimicrobial domain, independent of its chemokine domain in a nonsevere malaria model.155  This highlights both the complexity of platelets in immune responses, particularly in infectious diseases, and the complicated roles for PF4 in health and disease. Based on these studies, we explored whether platelet-driven outcomes are also inflammation and timing dependent in complicated malaria models. Platelets are activated as early as 24 hours after Plasmodium infection both in murine models and in human disease,51,141  a time when the APR is initiated. The APR response to malaria infection is dependent on platelets, and the platelet-induced APR helps limit the early parasite burden.51  This indicates that platelets may have an early protective role in limiting parasite burden, but with continued platelet activation and the release of platelet-derived immune mediators, platelets accelerate cerebral vascular compromise.

With the development of antiviral therapies and the greatly improved survival of HIV+ individuals, cardiovascular complications of chronic HIV infection and the associated drug regimens have become an important and poorly understood clinical consideration. The risk of thrombosis, including deep vein thrombosis and pulmonary embolism, is increased in HIV+ individuals.156,157  The mechanisms are not clear but may involve increased platelet activation, and with it broad inflammatory consequences. Studies using a primate simian immunodeficiency virus infection model have shown that platelets are activated early in infection, contributing to a drop in platelet count immediately following infection.158,159  Furthermore, platelet-monocyte aggregates are greatly increased in the circulation of both infected primates and humans, and platelets may induce monocyte differentiation into a more inflammatory phenotype.159,160  Thrombocytopenia is associated with increased neurologic complications of HIV infection, and neuro-HIV is hypothesized to be driven by infected macrophage migration into the brain.158,160,161  This has led some to speculate that a platelet-induced macrophage inflammatory phenotype may accelerate neurologic complications of HIV infection, but much remains to definitively demonstrate this. Influenza also exerts a large public health impact every year. In the 2009 H1N1A pandemic, ∼6% of patients experienced a thrombotic event.162  Platelet activation in H1N1A infection exceeded platelet activation noted in bacterial pneumonia.163  Similar to HIV infection, platelet-monocyte aggregates are elevated in influenza infection suggesting that platelets affect viral immune responses.163  Many of these important associations are only beginning to be identified. More basic science study is needed to better define mechanistic roles for platelets in driving influenza-related infection complications.

As noted previously in the discussion of platelet TLRs, platelets interact with and may help clear bacterial infections. As early as 1995, it was recognized that thrombin-stimulated platelets facilitated the clearance of adherent Streptococci in experimental infective endocarditis.164  Specific factors such as β-defensins are released from platelets activated by the Staphylococcus aureus α-toxin, impairing S aureus growth and inducing neutrophil extracellular trap formation.165  Platelets may also “nucleate” or help trap blood pathogens on Kupffer cells, the liver-specific macrophage present in hepatic sinusoids in direct contact with circulating blood.166  Bacteria infection of platelet-depleted and GPIbα−/− mice resulted in increased endothelial damage and vascular leak. In addition, these mice had reduced bacteria trapping in the liver demonstrating a potentially important role for platelets and Kupffer cell interactions in clearing bacteria within liver sinusoids. However, similar to the paradigm discussed with malaria, continued platelet activation in response to a systemic bacterial infection (sepsis) may lead to complications and worsen sepsis outcomes.167 

As an awareness of the nonhemostatic functions of platelets grows, studies into the role of platelets in infectious diseases are also increasing and are likely to continue to be better appreciated.

With the continued discovery of exciting new associations between platelets and inflammatory disease, platelets will continue to become better understood and appreciated as an immune cell. Platelet numbers, the diversity of platelet-derived inflammatory mediators, and the potential for multiple interactions between platelets and other cells, both directly and indirectly, increase the impact of platelets on inflammatory conditions, despite their small anucleate status. As we better understand the immune regulatory functions of platelets, we are also likely to better understand the roles for platelets in multiple inflammatory and infectious diseases.

This work was supported by grants from the American Heart Association (13EIA14250023) (C.N.M.) and the National Institutes of Health, National Center for Research Resources (TL1 RR024135-05) (L.M.C.).

C.N.M., A.A.A., K.L.M., and L.M.C. wrote and researched parts of this manuscript.

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

Correspondence: Craig Morrell, Box CVRI, 601 Elmwood Ave, Rochester, NY 14642; e-mail: Craig_Morrell@URMC.Rochester.edu.

1
Smyth
 
SS
McEver
 
RP
Weyrich
 
AS
, et al. 
2009 Platelet Colloquium Participants
Platelet functions beyond hemostasis.
J Thromb Haemost
2009
, vol. 
7
 
11
(pg. 
1759
-
1766
)
2
Srivastava
 
K
Cockburn
 
IA
Swaim
 
A
, et al. 
Platelet factor 4 mediates inflammation in experimental cerebral malaria.
Cell Host Microbe
2008
, vol. 
4
 
2
(pg. 
179
-
187
)
3
Pitsilos
 
S
Hunt
 
J
Mohler
 
ER
, et al. 
Platelet factor 4 localization in carotid atherosclerotic plaques: correlation with clinical parameters.
Thromb Haemost
2003
, vol. 
90
 
6
(pg. 
1112
-
1120
)
4
Aziz
 
KA
Cawley
 
JC
Zuzel
 
M
Platelets prime PMN via released PF4: mechanism of priming and synergy with GM-CSF.
Br J Haematol
1995
, vol. 
91
 
4
(pg. 
846
-
853
)
5
Gleissner
 
CA
Shaked
 
I
Erbel
 
C
Böckler
 
D
Katus
 
HA
Ley
 
K
CXCL4 downregulates the atheroprotective hemoglobin receptor CD163 in human macrophages.
Circ Res
2010
, vol. 
106
 
1
(pg. 
203
-
211
)
6
González-Cortés
 
C
Diez-Tascón
 
C
Guerra-Laso
 
JM
González-Cocaño
 
MC
Rivero-Lezcano
 
OM
Non-chemotactic influence of CXCL7 on human phagocytes. Modulation of antimicrobial activity against L. pneumophila.
Immunobiology
2012
, vol. 
217
 
4
(pg. 
394
-
401
)
7
Walz
 
A
Baggiolini
 
M
Generation of the neutrophil-activating peptide NAP-2 from platelet basic protein or connective tissue-activating peptide III through monocyte proteases.
J Exp Med
1990
, vol. 
171
 
2
(pg. 
449
-
454
)
8
Walz
 
A
Dewald
 
B
von Tscharner
 
V
Baggiolini
 
M
Effects of the neutrophil-activating peptide NAP-2, platelet basic protein, connective tissue-activating peptide III and platelet factor 4 on human neutrophils.
J Exp Med
1989
, vol. 
170
 
5
(pg. 
1745
-
1750
)
9
Del Conde
 
I
Crúz
 
MA
Zhang
 
H
López
 
JA
Afshar-Kharghan
 
V
Platelet activation leads to activation and propagation of the complement system.
J Exp Med
2005
, vol. 
201
 
6
(pg. 
871
-
879
)
10
Larsen
 
E
Palabrica
 
T
Sajer
 
S
, et al. 
PADGEM-dependent adhesion of platelets to monocytes and neutrophils is mediated by a lineage-specific carbohydrate, LNF III (CD15).
Cell
1990
, vol. 
63
 
3
(pg. 
467
-
474
)
11
Mayadas
 
TN
Johnson
 
RC
Rayburn
 
H
Hynes
 
RO
Wagner
 
DD
Leukocyte rolling and extravasation are severely compromised in P selectin-deficient mice.
Cell
1993
, vol. 
74
 
3
(pg. 
541
-
554
)
12
Czapiga
 
M
Kirk
 
AD
Lekstrom-Himes
 
J
Platelets deliver costimulatory signals to antigen-presenting cells: a potential bridge between injury and immune activation.
Exp Hematol
2004
, vol. 
32
 
2
(pg. 
135
-
139
)
13
Xu
 
H
Arnaud
 
F
Tadaki
 
DK
Burkly
 
LC
Harlan
 
DM
Kirk
 
AD
Human platelets activate porcine endothelial cells through a CD154-dependent pathway.
Transplantation
2001
, vol. 
72
 
11
(pg. 
1858
-
1861
)
14
Xu
 
H
Zhang
 
X
Mannon
 
RB
Kirk
 
AD
Platelet-derived or soluble CD154 induces vascularized allograft rejection independent of cell-bound CD154.
J Clin Invest
2006
, vol. 
116
 
3
(pg. 
769
-
774
)
15
Takimoto
 
T
Wakabayashi
 
Y
Sekiya
 
T
, et al. 
Smad2 and Smad3 are redundantly essential for the TGF-beta-mediated regulation of regulatory T plasticity and Th1 development [published correction appears in J Immunol. 2011;186(1):632].
J Immunol
2010
, vol. 
185
 
2
(pg. 
842
-
855
)
16
Santarlasci
 
V
Maggi
 
L
Capone
 
M
, et al. 
TGF-beta indirectly favors the development of human Th17 cells by inhibiting Th1 cells.
Eur J Immunol
2009
, vol. 
39
 
1
(pg. 
207
-
215
)
17
Sica
 
A
Mantovani
 
A
Macrophage plasticity and polarization: in vivo veritas.
J Clin Invest
2012
, vol. 
122
 
3
(pg. 
787
-
795
)
18
Ehrhardt
 
RO
Strober
 
W
Harriman
 
GR
Effect of transforming growth factor (TGF)-beta 1 on IgA isotype expression. TGF-beta 1 induces a small increase in sIgA+ B cells regardless of the method of B cell activation.
J Immunol
1992
, vol. 
148
 
12
(pg. 
3830
-
3836
)
19
Warner
 
GL
Nelson
 
DO
Scott
 
DW
Synergy between TGF-beta and anti-IgM in growth inhibition of CD5+ B-cell lymphomas.
Ann N Y Acad Sci
1992
, vol. 
651
 (pg. 
274
-
276
)
20
Bezuidenhout
 
L
Bracher
 
M
Davison
 
G
Zilla
 
P
Davies
 
N
Ang-2 and PDGF-BB cooperatively stimulate human peripheral blood monocyte fibrinolysis.
J Leukoc Biol
2007
, vol. 
81
 
6
(pg. 
1496
-
1503
)
21
Krettek
 
A
Ostergren-Lundén
 
G
Fager
 
G
Rosmond
 
C
Bondjers
 
G
Lustig
 
F
Expression of PDGF receptors and ligand-induced migration of partially differentiated human monocyte-derived macrophages. Influence of IFN-gamma and TGF-beta.
Atherosclerosis
2001
, vol. 
156
 
2
(pg. 
267
-
275
)
22
Chauhan
 
AK
Kisucka
 
J
Brill
 
A
Walsh
 
MT
Scheiflinger
 
F
Wagner
 
DD
ADAMTS13: a new link between thrombosis and inflammation.
J Exp Med
2008
, vol. 
205
 
9
(pg. 
2065
-
2074
)
23
Zhao
 
BQ
Chauhan
 
AK
Canault
 
M
, et al. 
von Willebrand factor-cleaving protease ADAMTS13 reduces ischemic brain injury in experimental stroke.
Blood
2009
, vol. 
114
 
15
(pg. 
3329
-
3334
)
24
Doyle
 
EL
Ridger
 
V
Ferraro
 
F
Turmaine
 
M
Saftig
 
P
Cutler
 
DF
CD63 is an essential cofactor to leukocyte recruitment by endothelial P-selectin.
Blood
2011
, vol. 
118
 
15
(pg. 
4265
-
4273
)
25
Stellos
 
K
Rahmann
 
A
Kilias
 
A
, et al. 
Expression of platelet-bound stromal cell-derived factor-1 in patients with non-valvular atrial fibrillation and ischemic heart disease.
J Thromb Haemost
2012
, vol. 
10
 
1
(pg. 
49
-
55
)
26
Dunussi-Joannopoulos
 
K
Zuberek
 
K
Runyon
 
K
, et al. 
Efficacious immunomodulatory activity of the chemokine stromal cell-derived factor 1 (SDF-1): local secretion of SDF-1 at the tumor site serves as T-cell chemoattractant and mediates T-cell-dependent antitumor responses.
Blood
2002
, vol. 
100
 
5
(pg. 
1551
-
1558
)
27
Takahashi
 
M
Role of the SDF-1/CXCR4 system in myocardial infarction.
Circ J
2010
, vol. 
74
 
3
(pg. 
418
-
423
)
28
Lee
 
CG
Link
 
H
Baluk
 
P
, et al. 
Vascular endothelial growth factor (VEGF) induces remodeling and enhances TH2-mediated sensitization and inflammation in the lung.
Nat Med
2004
, vol. 
10
 
10
(pg. 
1095
-
1103
)
29
Waldner
 
MJ
Wirtz
 
S
Jefremow
 
A
, et al. 
VEGF receptor signaling links inflammation and tumorigenesis in colitis-associated cancer.
J Exp Med
2010
, vol. 
207
 
13
(pg. 
2855
-
2868
)
30
Azimi-Nezhad
 
M
Stathopoulou
 
MG
Bonnefond
 
A
, et al. 
Associations of vascular endothelial growth factor (VEGF) with adhesion and inflammation molecules in a healthy population.
Cytokine
2013
, vol. 
61
 
2
(pg. 
602
-
607
)
31
Silverstein
 
RL
Nachman
 
RL
Thrombospondin binds to monocytes-macrophages and mediates platelet-monocyte adhesion.
J Clin Invest
1987
, vol. 
79
 
3
(pg. 
867
-
874
)
32
Lopez-Dee
 
Z
Pidcock
 
K
Gutierrez
 
LS
Thrombospondin-1: multiple paths to inflammation.
Mediators Inflamm
2011
, vol. 
2011
 pg. 
296069
 
33
Kimata
 
H
Yoshida
 
A
Ishioka
 
C
Fujimoto
 
M
Lindley
 
I
Furusho
 
K
RANTES and macrophage inflammatory protein 1 alpha selectively enhance immunoglobulin (IgE) and IgG4 production by human B cells.
J Exp Med
1996
, vol. 
183
 
5
(pg. 
2397
-
2402
)
34
Reichel
 
CA
Rehberg
 
M
Lerchenberger
 
M
, et al. 
Ccl2 and Ccl3 mediate neutrophil recruitment via induction of protein synthesis and generation of lipid mediators.
Arterioscler Thromb Vasc Biol
2009
, vol. 
29
 
11
(pg. 
1787
-
1793
)
35
Santos-Martínez
 
MJ
Medina
 
C
Jurasz
 
P
Radomski
 
MW
Role of metalloproteinases in platelet function.
Thromb Res
2008
, vol. 
121
 
4
(pg. 
535
-
542
)
36
Elvers
 
M
Herrmann
 
A
Seizer
 
P
, et al. 
Intracellular cyclophilin A is an important Ca(2+) regulator in platelets and critically involved in arterial thrombus formation.
Blood
2012
, vol. 
120
 
6
(pg. 
1317
-
1326
)
37
Fernández Bello
 
I
Álvarez
 
MT
López-Longo
 
FJ
, et al. 
Platelet soluble CD40L and matrix metalloproteinase 9 activity are proinflammatory mediators in Behçet disease patients.
Thromb Haemost
2012
, vol. 
107
 
1
(pg. 
88
-
98
)
38
Chung
 
AW
Radomski
 
A
Alonso-Escolano
 
D
, et al. 
Platelet-leukocyte aggregation induced by PAR agonists: regulation by nitric oxide and matrix metalloproteinases.
Br J Pharmacol
2004
, vol. 
143
 
7
(pg. 
845
-
855
)
39
O’Connell
 
PJ
Wang
 
X
Leon-Ponte
 
M
Griffiths
 
C
Pingle
 
SC
Ahern
 
GP
A novel form of immune signaling revealed by transmission of the inflammatory mediator serotonin between dendritic cells and T cells.
Blood
2006
, vol. 
107
 
3
(pg. 
1010
-
1017
)
40
León-Ponte
 
M
Ahern
 
GP
O’Connell
 
PJ
Serotonin provides an accessory signal to enhance T-cell activation by signaling through the 5-HT7 receptor.
Blood
2007
, vol. 
109
 
8
(pg. 
3139
-
3146
)
41
Swaim
 
AF
Field
 
DJ
Fox-Talbot
 
K
Baldwin
 
WM
Morrell
 
CN
Platelets contribute to allograft rejection through glutamate receptor signaling.
J Immunol
2010
, vol. 
185
 
11
(pg. 
6999
-
7006
)
42
Sarchielli
 
P
Di Filippo
 
M
Candeliere
 
A
, et al. 
Expression of ionotropic glutamate receptor GLUR3 and effects of glutamate on MBP- and MOG-specific lymphocyte activation and chemotactic migration in multiple sclerosis patients.
J Neuroimmunol
2007
, vol. 
188
 
1-2
(pg. 
146
-
158
)
43
Dinarvand
 
P
Hassanian
 
SM
Qureshi
 
SH
, et al. 
Polyphosphate amplifies proinflammatory responses of nuclear proteins through interaction with receptor for advanced glycation end products and P2Y1 purinergic receptor.
Blood
2014
, vol. 
123
 
6
(pg. 
935
-
945
)
44
Bae
 
JS
Lee
 
W
Rezaie
 
AR
Polyphosphate elicits pro-inflammatory responses that are counteracted by activated protein C in both cellular and animal models.
J Thromb Haemost
2012
, vol. 
10
 
6
(pg. 
1145
-
1151
)
45
Evangelista
 
V
Manarini
 
S
Dell’Elba
 
G
, et al. 
Clopidogrel inhibits platelet-leukocyte adhesion and platelet-dependent leukocyte activation.
Thromb Haemost
2005
, vol. 
94
 
3
(pg. 
568
-
577
)
46
Kronlage
 
M
Song
 
J
Sorokin
 
L
, et al. 
Autocrine purinergic receptor signaling is essential for macrophage chemotaxis.
Sci Signal
2010
, vol. 
3
 
132
pg. 
ra55
 
47
Riegel
 
AK
Faigle
 
M
Zug
 
S
, et al. 
Selective induction of endothelial P2Y6 nucleotide receptor promotes vascular inflammation.
Blood
2011
, vol. 
117
 
8
(pg. 
2548
-
2555
)
48
Mannaioni
 
PF
Di Bello
 
MG
Masini
 
E
Platelets and inflammation: role of platelet-derived growth factor, adhesion molecules and histamine.
Inflamm Res
1997
, vol. 
46
 
1
(pg. 
4
-
18
)
49
Ferguson
 
MK
Shahinian
 
HK
Michelassi
 
F
Lymphatic smooth muscle responses to leukotrienes, histamine and platelet activating factor.
J Surg Res
1988
, vol. 
44
 
2
(pg. 
172
-
177
)
50
André
 
P
Denis
 
CV
Ware
 
J
, et al. 
Platelets adhere to and translocate on von Willebrand factor presented by endothelium in stimulated veins.
Blood
2000
, vol. 
96
 
10
(pg. 
3322
-
3328
)
51
Aggrey
 
AA
Srivastava
 
K
Ture
 
S
Field
 
DJ
Morrell
 
CN
Platelet induction of the acute-phase response is protective in murine experimental cerebral malaria.
J Immunol
2013
, vol. 
190
 
9
(pg. 
4685
-
4691
)
52
Bustos
 
M
Saadi
 
S
Platt
 
JL
Platelet-mediated activation of endothelial cells: implications for the pathogenesis of transplant rejection.
Transplantation
2001
, vol. 
72
 
3
(pg. 
509
-
515
)
53
Lindemann
 
S
Tolley
 
ND
Dixon
 
DA
, et al. 
Activated platelets mediate inflammatory signaling by regulated interleukin 1beta synthesis.
J Cell Biol
2001
, vol. 
154
 
3
(pg. 
485
-
490
)
54
Boilard
 
E
Nigrovic
 
PA
Larabee
 
K
, et al. 
Platelets amplify inflammation in arthritis via collagen-dependent microparticle production.
Science
2010
, vol. 
327
 
5965
(pg. 
580
-
583
)
55
Yao
 
C
Sakata
 
D
Esaki
 
Y
, et al. 
Prostaglandin E2-EP4 signaling promotes immune inflammation through Th1 cell differentiation and Th17 cell expansion.
Nat Med
2009
, vol. 
15
 
6
(pg. 
633
-
640
)
56
Sakata
 
D
Yao
 
C
Narumiya
 
S
Emerging roles of prostanoids in T cell-mediated immunity.
IUBMB Life
2010
, vol. 
62
 
8
(pg. 
591
-
596
)
57
Capone
 
ML
Tacconelli
 
S
Sciulli
 
MG
, et al. 
Clinical pharmacology of platelet, monocyte, and vascular cyclooxygenase inhibition by naproxen and low-dose aspirin in healthy subjects.
Circulation
2004
, vol. 
109
 
12
(pg. 
1468
-
1471
)
58
Morrell
 
CN
Matsushita
 
K
Chiles
 
K
, et al. 
Regulation of platelet granule exocytosis by S-nitrosylation.
Proc Natl Acad Sci USA
2005
, vol. 
102
 
10
(pg. 
3782
-
3787
)
59
Simon
 
DI
Chen
 
Z
Xu
 
H
, et al. 
Platelet glycoprotein ibalpha is a counterreceptor for the leukocyte integrin Mac-1 (CD11b/CD18).
J Exp Med
2000
, vol. 
192
 
2
(pg. 
193
-
204
)
60
Rumbaut
 
RE
Thiagarajan
 
P
Platelet-Vessel Wall Interactions in Hemostasis and Thrombosis
2010
San Rafael, CA
Morgan & Claypool Life Sciences
61
Thon
 
JN
Peters
 
CG
Machlus
 
KR
, et al. 
T granules in human platelets function in TLR9 organization and signaling.
J Cell Biol
2012
, vol. 
198
 
4
(pg. 
561
-
574
)
62
Maynard
 
DM
Heijnen
 
HF
Horne
 
MK
White
 
JG
Gahl
 
WA
Proteomic analysis of platelet alpha-granules using mass spectrometry.
J Thromb Haemost
2007
, vol. 
5
 
9
(pg. 
1945
-
1955
)
63
Italiano
 
JE
Battinelli
 
EM
Selective sorting of alpha-granule proteins.
J Thromb Haemost
2009
, vol. 
7
 
suppl 1
(pg. 
173
-
176
)
64
Rendu
 
F
Brohard-Bohn
 
B
The platelet release reaction: granules’ constituents, secretion and functions.
Platelets
2001
, vol. 
12
 
5
(pg. 
261
-
273
)
65
Lemons
 
PP
Chen
 
D
Bernstein
 
AM
Bennett
 
MK
Whiteheart
 
SW
Regulated secretion in platelets: identification of elements of the platelet exocytosis machinery.
Blood
1997
, vol. 
90
 
4
(pg. 
1490
-
1500
)
66
Koseoglu
 
S
Flaumenhaft
 
R
Advances in platelet granule biology.
Curr Opin Hematol
2013
, vol. 
20
 
5
(pg. 
464
-
471
)
67
Deppermann
 
C
Cherpokova
 
D
Nurden
 
P
, et al. 
Gray platelet syndrome and defective thrombo-inflammation in Nbeal2-deficient mice.
J Clin Invest
2013
, vol. 
123
 
8
(pg. 
3331
-
3342
)
68
Blair
 
P
Flaumenhaft
 
R
Platelet alpha-granules: basic biology and clinical correlates.
Blood Rev
2009
, vol. 
23
 
4
(pg. 
177
-
189
)
69
Vanderstocken
 
G
Bondue
 
B
Horckmans
 
M
, et al. 
P2Y2 receptor regulates VCAM-1 membrane and soluble forms and eosinophil accumulation during lung inflammation.
J Immunol
2010
, vol. 
185
 
6
(pg. 
3702
-
3707
)
70
Younas
 
M
Hue
 
S
Lacabaratz
 
C
, et al. 
IL-7 modulates in vitro and in vivo human memory T regulatory cell functions through the CD39/ATP axis.
J Immunol
2013
, vol. 
191
 
6
(pg. 
3161
-
3168
)
71
Ganor
 
Y
Besser
 
M
Ben-Zakay
 
N
Unger
 
T
Levite
 
M
Human T cells express a functional ionotropic glutamate receptor GluR3, and glutamate by itself triggers integrin-mediated adhesion to laminin and fibronectin and chemotactic migration.
J Immunol
2003
, vol. 
170
 
8
(pg. 
4362
-
4372
)
72
Ganor
 
Y
Grinberg
 
I
Reis
 
A
Cooper
 
I
Goldstein
 
RS
Levite
 
M
Human T-leukemia and T-lymphoma express glutamate receptor AMPA GluR3, and the neurotransmitter glutamate elevates the cancer-related matrix-metalloproteinases inducer CD147/EMMPRIN, MMP-9 secretion and engraftment of T-leukemia in vivo.
Leuk Lymphoma
2009
, vol. 
50
 
6
(pg. 
985
-
997
)
73
Katoh
 
N
Soga
 
F
Nara
 
T
, et al. 
Effect of serotonin on the differentiation of human monocytes into dendritic cells.
Clin Exp Immunol
2006
, vol. 
146
 
2
(pg. 
354
-
361
)
74
Brown
 
GT
McIntyre
 
TM
Lipopolysaccharide signaling without a nucleus: kinase cascades stimulate platelet shedding of proinflammatory IL-1β-rich microparticles.
J Immunol
2011
, vol. 
186
 
9
(pg. 
5489
-
5496
)
75
Denis
 
MM
Tolley
 
ND
Bunting
 
M
, et al. 
Escaping the nuclear confines: signal-dependent pre-mRNA splicing in anucleate platelets.
Cell
2005
, vol. 
122
 
3
(pg. 
379
-
391
)
76
Aslam
 
R
Speck
 
ER
Kim
 
M
, et al. 
Platelet Toll-like receptor expression modulates lipopolysaccharide-induced thrombocytopenia and tumor necrosis factor-alpha production in vivo.
Blood
2006
, vol. 
107
 
2
(pg. 
637
-
641
)
77
Andonegui
 
G
Kerfoot
 
SM
McNagny
 
K
Ebbert
 
KV
Patel
 
KD
Kubes
 
P
Platelets express functional Toll-like receptor-4.
Blood
2005
, vol. 
106
 
7
(pg. 
2417
-
2423
)
78
Ståhl
 
AL
Svensson
 
M
Mörgelin
 
M
, et al. 
Lipopolysaccharide from enterohemorrhagic Escherichia coli binds to platelets through TLR4 and CD62 and is detected on circulating platelets in patients with hemolytic uremic syndrome.
Blood
2006
, vol. 
108
 
1
(pg. 
167
-
176
)
79
Clark
 
SR
Ma
 
AC
Tavener
 
SA
, et al. 
Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood.
Nat Med
2007
, vol. 
13
 
4
(pg. 
463
-
469
)
80
Blair
 
P
Rex
 
S
Vitseva
 
O
, et al. 
Stimulation of Toll-like receptor 2 in human platelets induces a thromboinflammatory response through activation of phosphoinositide 3-kinase.
Circ Res
2009
, vol. 
104
 
3
(pg. 
346
-
354
)
81
Beaulieu
 
LM
Lin
 
E
Morin
 
KM
Tanriverdi
 
K
Freedman
 
JE
Regulatory effects of TLR2 on megakaryocytic cell function.
Blood
2011
, vol. 
117
 
22
(pg. 
5963
-
5974
)
82
Panigrahi
 
S
Ma
 
Y
Hong
 
L
, et al. 
Engagement of platelet toll-like receptor 9 by novel endogenous ligands promotes platelet hyperreactivity and thrombosis.
Circ Res
2013
, vol. 
112
 
1
(pg. 
103
-
112
)
83
Zhu
 
W
Li
 
W
Silverstein
 
RL
Advanced glycation end products induce a prothrombotic phenotype in mice via interaction with platelet CD36.
Blood
2012
, vol. 
119
 
25
(pg. 
6136
-
6144
)
84
Ghosh
 
A
Murugesan
 
G
Chen
 
K
, et al. 
Platelet CD36 surface expression levels affect functional responses to oxidized LDL and are associated with inheritance of specific genetic polymorphisms.
Blood
2011
, vol. 
117
 
23
(pg. 
6355
-
6366
)
85
Chandler
 
AB
Hand
 
RA
Phagocytized platelets: a source of lipids in human thrombi and atherosclerotic plaques.
Science
1961
, vol. 
134
 
3483
(pg. 
946
-
947
)
86
Bainton
 
CR
Richter
 
DW
Seller
 
H
Ballantyne
 
D
Klein
 
JP
Respiratory modulation of sympathetic activity.
J Auton Nerv Syst
1985
, vol. 
12
 
1
(pg. 
77
-
90
)
87
Palabrica
 
T
Lobb
 
R
Furie
 
BC
, et al. 
Leukocyte accumulation promoting fibrin deposition is mediated in vivo by P-selectin on adherent platelets.
Nature
1992
, vol. 
359
 
6398
(pg. 
848
-
851
)
88
Hrachovinová
 
I
Cambien
 
B
Hafezi-Moghadam
 
A
, et al. 
Interaction of P-selectin and PSGL-1 generates microparticles that correct hemostasis in a mouse model of hemophilia A.
Nat Med
2003
, vol. 
9
 
8
(pg. 
1020
-
1025
)
89
Robbie
 
L
Libby
 
P
Inflammation and atherothrombosis.
Ann N Y Acad Sci
2001
, vol. 
947
 (pg. 
167
-
179, discussion 179-180
)
90
Hansson
 
GK
Libby
 
P
The immune response in atherosclerosis: a double-edged sword.
Nat Rev Immunol
2006
, vol. 
6
 
7
(pg. 
508
-
519
)
91
Duguid
 
JB
Thrombosis as a factor in the pathogenesis of coronary atherosclerosis.
J Pathol Bacteriol
1946
, vol. 
58
 (pg. 
207
-
212
)
92
Duguid
 
JB
Anderson
 
GS
The pathogenesis of hyaline arteriolosclerosis.
J Pathol Bacteriol
1952
, vol. 
64
 
3
(pg. 
519
-
522
)
93
Mustard
 
JF
Packham
 
MA
Rowsell
 
HC
Jorgensen
 
L
The role of platelets in thrombosis and atherosclerosis.
Thromb Diath Haemorrh Suppl
1967
, vol. 
26
 (pg. 
261
-
274
)
94
Renaud
 
S
Kinlough
 
RL
Mustard
 
JF
Relationship between platelet aggregation and the thrombotic tendency in rats fed hyperlipemic diets.
Lab Invest
1970
, vol. 
22
 
4
(pg. 
339
-
343
)
95
Huo
 
Y
Schober
 
A
Forlow
 
SB
, et al. 
Circulating activated platelets exacerbate atherosclerosis in mice deficient in apolipoprotein E.
Nat Med
2003
, vol. 
9
 
1
(pg. 
61
-
67
)
96
Nassar
 
T
Sachais
 
BS
Akkawi
 
S
, et al. 
Platelet factor 4 enhances the binding of oxidized low-density lipoprotein to vascular wall cells.
J Biol Chem
2003
, vol. 
278
 
8
(pg. 
6187
-
6193
)
97
Sachais
 
BS
Turrentine
 
T
Dawicki McKenna
 
JM
Rux
 
AH
Rader
 
D
Kowalska
 
MA
Elimination of platelet factor 4 (PF4) from platelets reduces atherosclerosis in C57Bl/6 and apoE-/- mice.
Thromb Haemost
2007
, vol. 
98
 
5
(pg. 
1108
-
1113
)
98
Dixon
 
DA
Tolley
 
ND
Bemis-Standoli
 
K
, et al. 
Expression of COX-2 in platelet-monocyte interactions occurs via combinatorial regulation involving adhesion and cytokine signaling.
J Clin Invest
2006
, vol. 
116
 
10
(pg. 
2727
-
2738
)
99
Shoji
 
T
Koyama
 
H
Fukumoto
 
S
, et al. 
Platelet-monocyte aggregates are independently associated with occurrence of carotid plaques in type 2 diabetic patients.
J Atheroscler Thromb
2005
, vol. 
12
 
6
(pg. 
344
-
352
)
100
Dotsenko
 
O
Chaturvedi
 
N
Thom
 
SA
, et al. 
Platelet and leukocyte activation, atherosclerosis and inflammation in European and South Asian men.
J Thromb Haemost
2007
, vol. 
5
 
10
(pg. 
2036
-
2042
)
101
Gremmel
 
T
Kopp
 
CW
Seidinger
 
D
, et al. 
The formation of monocyte-platelet aggregates is independent of on-treatment residual agonists’-inducible platelet reactivity.
Atherosclerosis
2009
, vol. 
207
 
2
(pg. 
608
-
613
)
102
Mueller
 
A
Meiser
 
A
McDonagh
 
EM
, et al. 
CXCL4-induced migration of activated T lymphocytes is mediated by the chemokine receptor CXCR3.
J Leukoc Biol
2008
, vol. 
83
 
4
(pg. 
875
-
882
)
103
Lasagni
 
L
Francalanci
 
M
Annunziato
 
F
, et al. 
An alternatively spliced variant of CXCR3 mediates the inhibition of endothelial cell growth induced by IP-10, Mig, and I-TAC, and acts as functional receptor for platelet factor 4.
J Exp Med
2003
, vol. 
197
 
11
(pg. 
1537
-
1549
)
104
Danese
 
S
de la Motte
 
C
Reyes
 
BM
Sans
 
M
Levine
 
AD
Fiocchi
 
C
Cutting edge: T cells trigger CD40-dependent platelet activation and granular RANTES release: a novel pathway for immune response amplification.
J Immunol
2004
, vol. 
172
 
4
(pg. 
2011
-
2015
)
105
Chapman
 
LM
Aggrey
 
AA
Field
 
DJ
, et al. 
Platelets present antigen in the context of MHC class I.
J Immunol
2012
, vol. 
189
 
2
(pg. 
916
-
923
)
106
Sprague
 
DL
Elzey
 
BD
Crist
 
SA
Waldschmidt
 
TJ
Jensen
 
RJ
Ratliff
 
TL
Platelet-mediated modulation of adaptive immunity: unique delivery of CD154 signal by platelet-derived membrane vesicles.
Blood
2008
, vol. 
111
 
10
(pg. 
5028
-
5036
)
107
Elzey
 
BD
Schmidt
 
NW
Crist
 
SA
, et al. 
Platelet-derived CD154 enables T-cell priming and protection against Listeria monocytogenes challenge.
Blood
2008
, vol. 
111
 
7
(pg. 
3684
-
3691
)
108
Elzey
 
BD
Tian
 
J
Jensen
 
RJ
, et al. 
Platelet-mediated modulation of adaptive immunity. A communication link between innate and adaptive immune compartments.
Immunity
2003
, vol. 
19
 
1
(pg. 
9
-
19
)
109
Shi
 
G
Field
 
DJ
Ko
 
KA
, et al. 
Platelet factor 4 limits Th17 differentiation and cardiac allograft rejection.
J Clin Invest
2014
, vol. 
124
 
2
(pg. 
543
-
552
)
110
Langer
 
HF
Daub
 
K
Braun
 
G
, et al. 
Platelets recruit human dendritic cells via Mac-1/JAM-C interaction and modulate dendritic cell function in vitro.
Arterioscler Thromb Vasc Biol
2007
, vol. 
27
 
6
(pg. 
1463
-
1470
)
111
Hagihara
 
M
Higuchi
 
A
Tamura
 
N
, et al. 
Platelets, after exposure to a high shear stress, induce IL-10-producing, mature dendritic cells in vitro.
J Immunol
2004
, vol. 
172
 
9
(pg. 
5297
-
5303
)
112
Verschoor
 
A
Neuenhahn
 
M
Navarini
 
AA
, et al. 
A platelet-mediated system for shuttling blood-borne bacteria to CD8α+ dendritic cells depends on glycoprotein GPIb and complement C3.
Nat Immunol
2011
, vol. 
12
 
12
(pg. 
1194
-
1201
)
113
Libby
 
P
Lichtman
 
AH
Hansson
 
GK
Immune effector mechanisms implicated in atherosclerosis: from mice to humans.
Immunity
2013
, vol. 
38
 
6
(pg. 
1092
-
1104
)
114
von Hundelshausen
 
P
Weber
 
C
Platelets as immune cells: bridging inflammation and cardiovascular disease.
Circ Res
2007
, vol. 
100
 
1
(pg. 
27
-
40
)
115
Zhang
 
X
McGeoch
 
SC
Johnstone
 
AM
, et al. 
Platelet-derived microparticle count and surface molecule expression differ between subjects with and without type 2 diabetes, independently of obesity status [published online ahead of print October 5, 2013].
J Thromb Thrombolysis
116
Guiducci
 
S
Distler
 
JH
Jüngel
 
A
, et al. 
The relationship between plasma microparticles and disease manifestations in patients with systemic sclerosis.
Arthritis Rheum
2008
, vol. 
58
 
9
(pg. 
2845
-
2853
)
117
Trappenburg
 
MC
van Schilfgaarde
 
M
Marchetti
 
M
, et al. 
Elevated procoagulant microparticles expressing endothelial and platelet markers in essential thrombocythemia.
Haematologica
2009
, vol. 
94
 
7
(pg. 
911
-
918
)
118
Nomura
 
S
Suzuki
 
M
Katsura
 
K
, et al. 
Platelet-derived microparticles may influence the development of atherosclerosis in diabetes mellitus.
Atherosclerosis
1995
, vol. 
116
 
2
(pg. 
235
-
240
)
119
Huisse
 
MG
Ajzenberg
 
N
Feldman
 
L
Guillin
 
MC
Steg
 
PG
Microparticle-linked tissue factor activity and increased thrombin activity play a potential role in fibrinolysis failure in ST-segment elevation myocardial infarction.
Thromb Haemost
2009
, vol. 
101
 
4
(pg. 
734
-
740
)
120
Mallat
 
Z
Benamer
 
H
Hugel
 
B
, et al. 
Elevated levels of shed membrane microparticles with procoagulant potential in the peripheral circulating blood of patients with acute coronary syndromes.
Circulation
2000
, vol. 
101
 
8
(pg. 
841
-
843
)
121
Bernal-Mizrachi
 
L
Jy
 
W
Jimenez
 
JJ
, et al. 
High levels of circulating endothelial microparticles in patients with acute coronary syndromes.
Am Heart J
2003
, vol. 
145
 
6
(pg. 
962
-
970
)
122
Montoro-García
 
S
Shantsila
 
E
Tapp
 
LD
, et al. 
Small-size circulating microparticles in acute coronary syndromes: relevance to fibrinolytic status, reparative markers and outcomes.
Atherosclerosis
2013
, vol. 
227
 
2
(pg. 
313
-
322
)
123
Mause
 
SF
von Hundelshausen
 
P
Zernecke
 
A
Koenen
 
RR
Weber
 
C
Platelet microparticles: a transcellular delivery system for RANTES promoting monocyte recruitment on endothelium.
Arterioscler Thromb Vasc Biol
2005
, vol. 
25
 
7
(pg. 
1512
-
1518
)
124
Dasgupta
 
SK
Le
 
A
Chavakis
 
T
Rumbaut
 
RE
Thiagarajan
 
P
Developmental endothelial locus-1 (Del-1) mediates clearance of platelet microparticles by the endothelium.
Circulation
2012
, vol. 
125
 
13
(pg. 
1664
-
1672
)
125
Suades
 
R
Padró
 
T
Alonso
 
R
López-Miranda
 
J
Mata
 
P
Badimon
 
L
Circulating CD45+/CD3+ lymphocyte-derived microparticles map lipid-rich atherosclerotic plaques in familial hypercholesterolaemia patients.
Thromb Haemost
2014
, vol. 
111
 
1
(pg. 
111
-
121
)
126
Hottz
 
ED
Lopes
 
JF
Freitas
 
C
, et al. 
Platelets mediate increased endothelium permeability in dengue through NLRP3-inflammasome activation.
Blood
2013
, vol. 
122
 
20
(pg. 
3405
-
3414
)
127
Hottz
 
ED
Oliveira
 
MF
Nunes
 
PC
, et al. 
Dengue induces platelet activation, mitochondrial dysfunction and cell death through mechanisms that involve DC-SIGN and caspases.
J Thromb Haemost
2013
, vol. 
11
 
5
(pg. 
951
-
962
)
128
De Rosa
 
S
Fichtlscherer
 
S
Lehmann
 
R
Assmus
 
B
Dimmeler
 
S
Zeiher
 
AM
Transcoronary concentration gradients of circulating microRNAs.
Circulation
2011
, vol. 
124
 
18
(pg. 
1936
-
1944
)
129
Fichtlscherer
 
S
De Rosa
 
S
Fox
 
H
, et al. 
Circulating microRNAs in patients with coronary artery disease.
Circ Res
2010
, vol. 
107
 
5
(pg. 
677
-
684
)
130
Sondermeijer
 
BM
Bakker
 
A
Halliani
 
A
, et al. 
Platelets in patients with premature coronary artery disease exhibit upregulation of miRNA340* and miRNA624*.
PLoS ONE
2011
, vol. 
6
 
10
pg. 
e25946
 
131
Nagalla
 
S
Shaw
 
C
Kong
 
X
, et al. 
Platelet microRNA-mRNA coexpression profiles correlate with platelet reactivity.
Blood
2011
, vol. 
117
 
19
(pg. 
5189
-
5197
)
132
Edelstein
 
LC
Bray
 
PF
MicroRNAs in platelet production and activation.
Blood
2011
, vol. 
117
 
20
(pg. 
5289
-
5296
)
133
Landry
 
P
Plante
 
I
Ouellet
 
DL
Perron
 
MP
Rousseau
 
G
Provost
 
P
Existence of a microRNA pathway in anucleate platelets.
Nat Struct Mol Biol
2009
, vol. 
16
 
9
(pg. 
961
-
966
)
134
Risitano
 
A
Beaulieu
 
LM
Vitseva
 
O
Freedman
 
JE
Platelets and platelet-like particles mediate intercellular RNA transfer.
Blood
2012
, vol. 
119
 
26
(pg. 
6288
-
6295
)
135
Laffont
 
B
Corduan
 
A
Plé
 
H
, et al. 
Activated platelets can deliver mRNA regulatory Ago2•microRNA complexes to endothelial cells via microparticles.
Blood
2013
, vol. 
122
 
2
(pg. 
253
-
261
)
136
Gidlöf
 
O
van der Brug
 
M
Ohman
 
J
, et al. 
Platelets activated during myocardial infarction release functional miRNA, which can be taken up by endothelial cells and regulate ICAM1 expression.
Blood
2013
, vol. 
121
 
19
(pg. 
3908
-
3917
)
137
Sicuri
 
E
Vieta
 
A
Lindner
 
L
Constenla
 
D
Sauboin
 
C
The economic costs of malaria in children in three sub-Saharan countries: Ghana, Tanzania and Kenya.
Malar J
2013
, vol. 
12
 
1
pg. 
307
 
138
Kakar
 
A
Bhoi
 
S
Prakash
 
V
Kakar
 
S
Profound thrombocytopenia in Plasmodium vivax malaria.
Diagn Microbiol Infect Dis
1999
, vol. 
35
 
3
(pg. 
243
-
244
)
139
Hill
 
GJ
Knight
 
V
Jeffery
 
GM
Thrombocytopenia in vivax malaria.
Lancet
1964
, vol. 
283
 
7327
(pg. 
240
-
241
)
140
Beale
 
PJ
Cormack
 
JD
Oldrey
 
TB
Thrombocytopenia in malaria with immunoglobulin (IgM) changes.
BMJ
1972
, vol. 
1
 
5796
(pg. 
345
-
349
)
141
Kelton
 
JG
Keystone
 
J
Moore
 
J
, et al. 
Immune-mediated thrombocytopenia of malaria.
J Clin Invest
1983
, vol. 
71
 
4
(pg. 
832
-
836
)
142
Idro
 
R
Ndiritu
 
M
Ogutu
 
B
, et al. 
Burden, features, and outcome of neurological involvement in acute falciparum malaria in Kenyan children.
JAMA
2007
, vol. 
297
 
20
(pg. 
2232
-
2240
)
143
Grau
 
GE
Mackenzie
 
CD
Carr
 
RA
, et al. 
Platelet accumulation in brain microvessels in fatal pediatric cerebral malaria.
J Infect Dis
2003
, vol. 
187
 
3
(pg. 
461
-
466
)
144
Wassmer
 
SC
Combes
 
V
Grau
 
GE
Pathophysiology of cerebral malaria: role of host cells in the modulation of cytoadhesion.
Ann N Y Acad Sci
2003
, vol. 
992
 (pg. 
30
-
38
)
145
Wassmer
 
SC
Lépolard
 
C
Traoré
 
B
Pouvelle
 
B
Gysin
 
J
Grau
 
GE
Platelets reorient Plasmodium falciparum-infected erythrocyte cytoadhesion to activated endothelial cells.
J Infect Dis
2004
, vol. 
189
 
2
(pg. 
180
-
189
)
146
van der Heyde
 
HC
Nolan
 
J
Combes
 
V
Gramaglia
 
I
Grau
 
GE
A unified hypothesis for the genesis of cerebral malaria: sequestration, inflammation and hemostasis leading to microcirculatory dysfunction.
Trends Parasitol
2006
, vol. 
22
 
11
(pg. 
503
-
508
)
147
Perkash
 
A
Kelly
 
NI
Fajardo
 
LF
Enhanced parasitization of platelets by Plasmodium berghei yoelii.
Trans R Soc Trop Med Hyg
1984
, vol. 
78
 
4
(pg. 
451
-
455
)
148
Wassmer
 
SC
de Souza
 
JB
Frère
 
C
Candal
 
FJ
Juhan-Vague
 
I
Grau
 
GE
TGF-beta1 released from activated platelets can induce TNF-stimulated human brain endothelium apoptosis: a new mechanism for microvascular lesion during cerebral malaria.
J Immunol
2006
, vol. 
176
 
2
(pg. 
1180
-
1184
)
149
Sun
 
G
Chang
 
WL
Li
 
J
Berney
 
SM
Kimpel
 
D
van der Heyde
 
HC
Inhibition of platelet adherence to brain microvasculature protects against severe Plasmodium berghei malaria.
Infect Immun
2003
, vol. 
71
 
11
(pg. 
6553
-
6561
)
150
van der Heyde
 
HC
Gramaglia
 
I
Sun
 
G
Woods
 
C
Platelet depletion by anti-CD41 (alphaIIb) mAb injection early but not late in the course of disease protects against Plasmodium berghei pathogenesis by altering the levels of pathogenic cytokines.
Blood
2005
, vol. 
105
 
5
(pg. 
1956
-
1963
)
151
Srivastava
 
K
Field
 
DJ
Aggrey
 
A
Yamakuchi
 
M
Morrell
 
CN
Platelet factor 4 regulation of monocyte KLF4 in experimental cerebral malaria.
PLoS ONE
2010
, vol. 
5
 
5
pg. 
e10413
 
152
Wilson
 
NO
Jain
 
V
Roberts
 
CE
, et al. 
CXCL4 and CXCL10 predict risk of fatal cerebral malaria.
Dis Markers
2011
, vol. 
30
 
1
(pg. 
39
-
49
)
153
Wassmer
 
SC
Combes
 
V
Candal
 
FJ
Juhan-Vague
 
I
Grau
 
GE
Platelets potentiate brain endothelial alterations induced by Plasmodium falciparum.
Infect Immun
2006
, vol. 
74
 
1
(pg. 
645
-
653
)
154
McMorran
 
BJ
Marshall
 
VM
de Graaf
 
C
, et al. 
Platelets kill intraerythrocytic malarial parasites and mediate survival to infection.
Science
2009
, vol. 
323
 
5915
(pg. 
797
-
800
)
155
Love
 
MS
Millholland
 
MG
Mishra
 
S
, et al. 
Platelet factor 4 activity against P. falciparum and its translation to nonpeptidic mimics as antimalarials.
Cell Host Microbe
2012
, vol. 
12
 
6
(pg. 
815
-
823
)
156
Crum-Cianflone
 
NF
Weekes
 
J
Bavaro
 
M
Review: thromboses among HIV-infected patients during the highly active antiretroviral therapy era.
AIDS Patient Care STDS
2008
, vol. 
22
 
10
(pg. 
771
-
778
)
157
Lijfering
 
WM
Ten Kate
 
MK
Sprenger
 
HG
van der Meer
 
J
Absolute risk of venous and arterial thrombosis in HIV-infected patients and effects of combination antiretroviral therapy.
J Thromb Haemost
2006
, vol. 
4
 
9
(pg. 
1928
-
1930
)
158
Wachtman
 
LM
Skolasky
 
RL
Tarwater
 
PM
, et al. 
Platelet decline: an avenue for investigation into the pathogenesis of human immunodeficiency virus -associated dementia.
Arch Neurol
2007
, vol. 
64
 
9
(pg. 
1264
-
1272
)
159
Metcalf Pate
 
KA
Lyons
 
CE
Dorsey
 
JL
, et al. 
Platelet activation and platelet-monocyte aggregate formation contribute to decreased platelet count during acute simian immunodeficiency virus infection in pig-tailed macaques.
J Infect Dis
2013
, vol. 
208
 
6
(pg. 
874
-
883
)
160
Singh
 
MV
Davidson
 
DC
Kiebala
 
M
Maggirwar
 
SB
Detection of circulating platelet-monocyte complexes in persons infected with human immunodeficiency virus type-1.
J Virol Methods
2012
, vol. 
181
 
2
(pg. 
170
-
176
)
161
Wachtman
 
LM
Tarwater
 
PM
Queen
 
SE
Adams
 
RJ
Mankowski
 
JL
Platelet decline: an early predictive hematologic marker of simian immunodeficiency virus central nervous system disease.
J Neurovirol
2006
, vol. 
12
 
1
(pg. 
25
-
33
)
162
Bunce
 
PE
High
 
SM
Nadjafi
 
M
Stanley
 
K
Liles
 
WC
Christian
 
MD
Pandemic H1N1 influenza infection and vascular thrombosis.
Clin Infect Dis
2011
, vol. 
52
 
2
(pg. 
e14
-
e17
)
163
Rondina
 
MT
Brewster
 
B
Grissom
 
CK
, et al. 
In vivo platelet activation in critically ill patients with primary 2009 influenza A(H1N1).
Chest
2012
, vol. 
141
 
6
(pg. 
1490
-
1495
)
164
Dankert
 
J
van der Werff
 
J
Zaat
 
SA
Joldersma
 
W
Klein
 
D
Hess
 
J
Involvement of bactericidal factors from thrombin-stimulated platelets in clearance of adherent viridans streptococci in experimental infective endocarditis.
Infect Immun
1995
, vol. 
63
 
2
(pg. 
663
-
671
)
165
Kraemer
 
BF
Campbell
 
RA
Schwertz
 
H
, et al. 
Novel anti-bacterial activities of β-defensin 1 in human platelets: suppression of pathogen growth and signaling of neutrophil extracellular trap formation.
PLoS Pathog
2011
, vol. 
7
 
11
pg. 
e1002355
 
166
Wong
 
CH
Jenne
 
CN
Petri
 
B
Chrobok
 
NL
Kubes
 
P
Nucleation of platelets with blood-borne pathogens on Kupffer cells precedes other innate immunity and contributes to bacterial clearance.
Nat Immunol
2013
, vol. 
14
 
8
(pg. 
785
-
792
)
167
Patel
 
KN
Soubra
 
SH
Lam
 
FW
Rodriguez
 
MA
Rumbaut
 
RE
Polymicrobial sepsis and endotoxemia promote microvascular thrombosis via distinct mechanisms.
J Thromb Haemost
2010
, vol. 
8
 
6
(pg. 
1403
-
1409
)
Sign in via your Institution