Skip to main content

New frontiers for platelet CD154

Abstract

The role of platelets extends beyond hemostasis. The pivotal role of platelets in inflammation has shed new light on the natural history of conditions associated with acute or chronic inflammation. Beyond the preservation of vascular integrity, platelets are essential to tissue homeostasis and platelet-derived products are already used in the clinics. Unanticipated was the role of platelets in the adaptative immune response, allowing a renewed conceptual approach of auto-immune diseases. Platelets are also important players in cancer growth and dissemination. Platelets fulfill most of their functions through the expression of still incompletely characterized membrane-bound or soluble mediators. Among them, CD154 holds a peculiar position, as platelets represent a major source of CD154 and as CD154 contributes to most of these new platelet attributes. Here, we provide an overview of some of the new frontiers that the study of platelet CD154 is opening, in inflammation, tissue homeostasis, immune response, hematopoiesis and cancer.

Introduction

Platelets are cytoplasmic fragments released in the bloodstream during the fragmentation of polyploid megakaryocytes (MK), a phenomenon critically dependent on thrombopoietin [1-3]. The mammalian platelet is thought to result from a phylogenic trend to ensure hemostasis under high vascular shear forces; indeed, it can specifically form arterial thrombi sustaining high shear stress [4]. It is thought that the platelet coopted attributes of a nucleated cell ancestor endowed with a multifunctional role in coagulation, inflammation and defense against infections [5,6]. Platelets have a short lifespan, of around 7 days; mechanisms responsible for their clearance are ill-understood; lectin-carbohydrate recognition of aged and damaged platelets by splenic and liver macrophages and hepatocytes is emphasized [7]. The best-defined function of platelets is hemostasis. Disruption of the endothelial cell (EC) lining leads to platelet activation, platelet adherence and aggregation which temporarily plug the damaged vessel. In this process, platelets also drive and confine coagulation at sites of tissue damage. Indeed, deficiencies in platelet production or function are associated to bleeding disorders, while increases in platelet number or gain of function are associated to thrombosis. The role of platelets in health and disease extends beyond hemostasis; non-hemostatic platelet functions include inflammation, innate and adaptative immune responses and tissue homeostasis (Figure 1). Decisive advances in understanding platelet function have been made through the characterization of platelet receptors and their ligands and platelet-derived mediators [8]. Among platelet mediators, CD154, the ligand of CD40, has attracted specific attention as it orchestrates many of these new platelet attributes.

Figure 1
figure 1

Platelets have a pleiotropic range of biological roles that extend beyond hemostasis. The breaching of tissue homeostasis leads to platelet activation, a common event in various causes of tissue injury, traumatic, infectious, ischemic, autoimmune… Platelet activation, apart from its essential role in bleeding arrest, is the source of a flow of information that fuels the inflammatory reaction. Platelets represent host defense machines against infection, via the clearing of pathogens and the expression of membrane-bound and soluble signals that regulate the innate and adaptative arms of the immune response. Pathways activated in inflammation, coagulation, vascular/tissue repair and host defense are connected via soluble and cell-mediated signals, providing a coherent biological response aiming at arresting bleeding, curing infection and reestablishing tissue homeostasis. CD154 interfaces with many of these pathways (see Figures 2 and 3); activated platelets express a membrane-bound form of CD154 and release a soluble form (sCD154). Platelet derived microparticles (PMPs) recapitulate several of activated platelet functions (see text for details). Only some relevant molecules have been depicted. Small circles symbolize secreted molecules, large circles membrane-associated molecules. Abbreviations: CAMs, cell adhesion molecules; Fg, fibrinogen; Fn, fibronectin; ECM, extracellular matrix; NET ind., neutrophil extracellular traps induction; PAF, platelet activating factor; ROS, reactive oxygen species; Vn, vitronectin; vWF, von Willebrand factor.

CD154

CD154, the CD40 ligand, a member of the Tumor Necrosis Factor (TNF) family, is central to the immune response [9,10]. CD154 was discovered as mediating humoral immunity and was originally considered to be restricted to activated helper T cells. The CD154/CD40 interaction drives B cell proliferation, antibody production and isotype switching and is involved in thymic selection. This interaction is required for B memory cell generation and germinal center formation. Accordingly, CD154 deficiency is associated with an impairment of the humoral immune response to T-cell dependent antigens, including defective immunoglobulin class switching; patients with the X-linked hyper-IgM syndrome caused by mutations of the CD154 gene, generally present low serum IgG and IgA, but normal or increased serum IgM, and are susceptible to opportunistic infections. Mice with a disrupted Cd154 gene fail to undergo isotype switching to T-cell dependent antigens while normally responding to T-cell independent antigens. In line with its regulatory role on the adaptative immune response, the CD40/CD154 interaction contributes to autoimmune disorders in a number of animal models [11-15]. Manipulation of the CD154/CD40 interaction has been used in efforts to develop novel strategies in autoimmune diseases, results in animal models being encouraging [13]. Clinical trials have been launched with humanized anti-CD154 monoclonal antibodies. Clinical interest of this strategy remains mixed, and is strongly limited by thrombotic complications [12-14].

Apart from B cells, CD40 is expressed by various cells, including dendritic cells (DC), monocytes, T lymphocytes, EC, a variety of epithelial cells, smooth muscle cells, fibroblasts; its expression is low in basal conditions and is stimulated by inflammatory mediators [16-19]. CD40 expression is increased by CD154, however it is not known whether this induction is direct or indirect [20,21]. CD40 is not the sole receptor for CD154; alternative receptors have been described, such as integrins α5β1, αIIbβ3 and αMβ2; CD154 binding depends on their activation states [22-25]. These additional receptors are of significance in the pathophysiology of atherogenesis and are important to consider when comparing CD40- and CD154-deficient mouse phenotypes.

CD154 is a transmembrane protein and a proteolytic soluble form, sCD154, which keeps the CD40-binding domain, is released by a partially understood mechanism. The release of sCD154 was first documented in activated T-lymphocytes [26]. CD154 has a trimeric configuration, required for functional activity [27-30]. A complex signaling cascade is triggered by CD40 ligation, involving TNF receptor-associated factors (TRAF) as proximal transducing signal initiators [10,20]. Several signaling pathways, including nuclear factor-κB (NF-κB), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase pathways, are activated by CD40 ligation; however, there is a differential outcome depending upon which TRAF member binds preferentially, and which cell/conditions are involved [31]; the binding of TRAF-6 is critical in vascular inflammation and metabolic complications associated with obesity [32,33].

CD154 expression is also observed in natural killer cells, DC, cells of the monocyte/macrophage lineage, endothelial, smooth muscle and epithelial cells [20]. Basal CD154 expression is very low, or undetectable, as in EC and epithelial cells for example [34], and is increased by a variety of stimuli, most notably inflammatory cytokines [20]. This suggests that CD154 expression may mostly have relevance when induced, as in inflammation. CD154 is also expressed by blood platelets, being cryptic in unstimulated platelets and rapidly exposed at the platelet surface following platelet activation [35].

CD154 expression by platelets

The distribution of CD154 in platelets is partly understood. CD154 was found in α-granules, as shown by immunoelectron microscopy or quantitative immunofluorescence approaches [36,37]. Accordingly, patients presenting a Gray-platelet syndrome, are characterized by platelets that lack α-granules, and do not release CD154 upon activation [37]. CD154 is highly coclustered with insulin growth factor in α-granules, the signification of which is unknown [36]. One question is whether CD154 is also cytosolic, as found in resting platelets [38].

Pre-mRNAs and mature mRNAs are present in platelets and a functional spliceosome and translational apparatus allow platelets to process them, in response to platelet-activating signals [39,40]. Detecting CD154 mRNA by RT-PCR in platelets is challenging because of purity issues. However, CD154 mRNA was evidenced in mouse platelets, introducing other potential regulatory layers of CD154 expression by platelets [34].

When activated, platelets express a membrane form and release a soluble form of CD154

Platelets are activated by immobilized or soluble agonists. The activation-driven secretion of granule content is a primary phenomenon [41-46]. Platelets also synthetize mediators, including interleukin-1β, tissue factor (TF), fibrinogen, thrombospondin, von Willebrand Factor, αIIbβ3, through a translational-dependent pathway triggered by platelet activation [47,48].

Soluble CD154 is released by an activation-driven proteolytic mechanism. Agonists, including thrombin, thrombin receptor-agonist peptide, ADP or collagen, stimulate CD154 expression at the platelet membrane and the release of sCD154; long-term platelet activation leads to complete conversion of CD154 to sCD154 [38,49-53]. A matrix metalloproteinase (MMP)-dependent proteolytic event is involved. The involvement of MMPs, MMP-2 and/or MMP-9, [51,54-57], differs from the release of sCD154 by activated T-cells, which involves ADAM10 and 17 [58]. A role for αIIb/β3 has been put forward, as αIIb/β3 antagonists inhibit sCD154 release and as Glanzmann platelets show reduced sCD154 release rate [53,54,59]. An interaction between αIIb/β3 and MMP-2 is involved [57]. The roles of NADPH activation and reactive oxygen species (ROS) generation as well as CD154 binding to platelet CD40 have been underlined [50,60]. The particularity of sCD154 release may explain its specific response to agonists and secretion kinetics [38,53]; however, how sCD154 is released remains be fully understood, as shown for example by the effects of inhibitors added after platelet activation, suggesting complex, intra-platelet mechanisms [53]. A debate remains about the parallel biological activities of platelet-derived soluble and membrane-associated CD154; recombinant soluble forms, particularly trimeric forms, are active [50,61-63]. Finally, sCD154 activates platelets by itself, suggesting feed-back amplification of its secretion [64,65].

The megakaryocytic origin of platelet CD154

The assembly and loading of granules mainly occur in MK; granules are distributed in proplatelets via a microtubule-dependent mechanism [2,66,67]. The main origin of platelet CD154 is likely to be the MK that express CD154 mRNA, as shown in MK derived by differentiation of human and mouse hematopoietic progenitor cells and in MK of immune thrombocytopenic purpura (ITP) patients [68,69]. CD154 mRNA expression is increased upon MK differentiation [69]. CD154 protein is also found in MK cell lines and in MK from ITP patients [38,68,69]. As for T cells, the calcium-dependent activation of nuclear factor of activated T cells-c2 and the early growth response transcription factor EGR-1 contribute to CD154 gene activation in MK [69,70].

Translation from endogenous mRNAs contributes to platelet content. Its significance in quiescent platelets is unclear. However, pre-mRNA processing and mRNA translation are driven by platelet activation [40,48,71]. The contribution of such mechanism in CD154 expression during platelet lifespan is unknown.

Platelets also carry mediators present in plasma and possibly concentrated and/or modified within platelets [72,73]. Fibrinogen, albumin, immunoglobulins, amino acids, inflammatory and angiogenic mediators including vascular endothelial growth factor (VEGF), histamine or serotonin, are among them. Soluble CD154 is not detected in platelets, making unlikely its uptake from plasma.

Platelets are a significant reservoir of CD154 in the organism

Platelets carry approximately 5 ng of CD154/mL of blood [52]. Correlation studies suggest a link between platelet count and plasma or serum sCD154 [37,52,74-78]. Such a correlation is also found in experimental ITP [78]. In ITP, albeit platelet CD154 is elevated [68], plasma sCD154 is reduced [78], again suggesting relationship between the platelet count and circulating sCD154. However, there are contrasting studies, and a correlation between the platelet count and sCD154 is not always found [79,80].

Importantly, platelet activation is associated to elevated sCD154 and, indeed, platelet activation markers correlate with sCD154 in blood [81-83]. For this reason, serum seems inappropriate to evaluate circulating sCD154; in fact, sCD154 levels are higher in serum than in plasma, clotting resulting in increased sCD154 generation [52,79,80,84-88]. Hence the importance of a preanalytical standardization of blood samples processing, conditions such as temperature, length of storage, centrifugation, interfering with measurement [84,89]. Further, plasma/serum sCD154 may correspond to a pool of free soluble and microparticle-bound CD154 [84] and ELISA may not discriminate between sCD154 and platelet microparticles (PMP)-associated CD154 [90]. Circulating sCD154 is linked to platelet activation state; in patients with recent thrombotic events, plasma sCD154 correlates with platelet count, but this correlation is not found in patients with non-thrombotic, non-inflammatory conditions [84]. Finally, in patients with cardiovascular conditions, commonly used drugs such as statins, interfere with sCD154 releasing, a point that has also to be considered [91-93]. The baseline presence of sCD154 in the plasma of healthy subjects may be secondary to basal platelet activation, as in high shear stress flow areas [94]. PMP are released upon platelet activation [95]. A functional CD154 is expressed by PMP [63,96]. The importance of the contribution of PMP-bound CD154, in comparison with the “true” soluble CD154, to plasma sCD154 has been emphasized [90]. Questions also remain on the fate and half-life of sCD154 in blood and how the CD154 information can be delivered at distance from platelet activation sites.

Platelet CD154: a critical mediator of the inflammatory reaction

Platelets orchestrate a subtle balance between tissue injury and repair; they are a key source of material for reestablishing tissue homeostasis but they also contribute to tissue injury. CD154 mediates several platelet functions in tissue homeostasis (Figure 2).

Figure 2
figure 2

CD154 is a universal contributor to platelet functions. Activated platelets display CD154 at their membrane and release a soluble form (sCD154). Platelet CD154, directly or indirectly, is a molecular driver of inflammation, coagulation, tissue remodeling, and host defense, processes that intersect at multiple levels. Endothelium is a primary target. Platelet CD154 induces tissue factor (TF) expression and activity, thereby contributing to thrombin generation and upregulates urokinase plasminogen activator receptor which is at the interface between fibrinolysis/inflammation/tissue remodeling. MMP-9 and MT1-MMP induction contribute to regulate endothelium proteolytic activity [97]. Platelet CD154 also induces pro-inflammatory and chemotactic (dotted semicircles) molecule expression, and adhesion molecule expression (CD62e, CD54, CD106,…), leading to leukocyte recruitment and activation. Once activated, target cells recruit and activate other cells through multiple inputs; several amplification loops are thus generated including platelet activation by sCD154 itself (blue arrow). Platelet CD154 also activates cellular effectors of the innate and adaptative immune responses, polymorphonuclear cells (PMN), monocytes (MNC), macrophages (MΦ), dendritic cells (DC); how platelet CD154 contributes to host defense is schematized in Figure 3. CD154-expressing platelet microparticles (PMPs) share most of these functions. Depicted molecules do not comprehensively represent the range of platelet-derived mediators that are controlled by platelet CD154, and other interfaces, such as with endothelin-1, continue to be identified. Magenta arrows depict interaction with CD40. Dotted line for thrombomodulin (ThMod) represents inhibition; full line for others represents stimulation. Dotted semicircles symbolize chemotaxis. Abbreviations: Ly, lymphocytes; MMP, matrix metalloproteinases.

Platelet CD154 and inflammation

Regardless of its cause, the inflammatory milieu is rich in platelet-activating material, including chemokines [98]. The dialog between EC and platelets in inflammation has been widely studied as EC are primary platelet partners. Upon CD40 ligation, EC switch to an activated phenotype, expressing molecules that contribute to an inflammatory and thrombotic scenario, including cytokines/chemokines, adhesion molecules, and tissue factor [16,20,99]. Platelets/EC reciprocal activation is critical in atherosclerosis and cardiovascular conditions [100-103]. The pathogenic role of platelet CD154 is a major theme in atherosclerosis and cardiovascular diseases [25,62,74,100-109].

The role of platelet CD154 in inflammation extends beyond the dialog with EC, as activated platelets interact with various CD40 expressing-cells. Platelets are brought to inflammatory sites via vascular injury/permeability, attachment to activated leukocytes, and also chemotactic recruitment [110]. CD40 ligation on inflammatory cells at sites of tissue injury is a potent stimulus for the expression of a variety of proinflammatory mediators including cytokines, chemokines, eicosanoids, products of the proteolytic cascades, ROS generation, and of adhesion molecules [49,111], making platelet CD154 a versatile fuel for inflammation. The platelet contribution in many inflammation-associated disorders, including rheumatic, lung, gastrointestinal, neuro-inflammatory and metabolic diseases is actively studied [112-120] and the specific pathogenic role played by platelet CD154 in these disorders is a recently opened frontier. Soluble CD154 levels were found to correlate with disease activity as in systemic lupus erythematosus [121]; whether sCD154 could represent a potential useful marker in inflammation-associated disorders is an interesting question. PMP also contribute to inflammatory disorders [122-128]; the specific role of PMP-associated CD154 remains however to be fully understood.

Platelet CD154 and tissue repair

The effectors of inflammation are orchestrated to cure infection and restore tissue integrity [129-131]. At various steps of tissue repair, platelets are a source of relevant material, including growth factors, pro- and anti-apoptotic mediators, matrix and matrix remodeling proteins [132-135] (Figure 1). Platelets contribute to maintain resting and injured endothelium integrity [136]. On injured endothelium, platelets provide EC growth-promoting and anti-apoptotic mediators, attractants for progenitor cells endowed with vascular healing properties [135]. They contribute to restoring the vascular network, by secreting regulators of angiogenesis [137-139]. Beyond endothelium, a remarkable role for platelets in organ regeneration has been substantiated. Platelets contribute to liver regeneration, serotonin being essential [140-142]. It is tempting to speculate that platelets will be found to have a broader role in organ regeneration by providing key mitogenic signals in various organs, such as for example fibroblast growth factor or platelet-derived growth factor that contribute to muscle or brain repair [143,144]. This is also in line with the known ability of platelet lysates to sustain the growth of primary cell cultures. PMP also contribute to vascular integrity [145-148] and promote tissue repair [128,149]. Platelet products have already found various applications in the clinics [150-154].

The specific role of CD154 has been mainly studied in EC. CD154 promotes EC survival, proliferation and migration, capillary-like tube formation in vitro and angiogenesis in vivo. Mechanisms include activation of the phosphatidylinositol-3 kinase/Akt pathway, induction of angiogenic mediators and matrix remodeling protein production [155-157]. CD40 signaling contributes to neointima repair, TRAF6 signaling intermediate being critical [32,158,159]. However, platelet CD154 was shown to inhibit the VEGF-induced EC migration via increased ROS generation, and sCD154 to inhibit VEGF-induced angiogenesis [160]. Soluble CD154 also promotes oxidative stress in endothelial outgrowth cells (EOC), reducing their viability and proliferation [161], while promoting endothelial repair via increased production of MMP-9 by EOC [162]. These findings may be context-dependent; they emphasize the importance of platelet CD154 in vascular homeostasis and the complexity of its biological interfaces. Other tissues for which platelet CD154 is likely to show importance for repair are skin and bone. CD40 ligation stimulates keratinocyte differentiation, suggesting contribution to skin wound repair [163]. Regulation of osteoclastogenesis by CD154 is suggested by the reduced bone mineral density together with elevated urine markers of osteoclast activity in patients with the X-linked hyper-IgM syndrome, and the reduced bone mineral density in CD154 deficient mice [164,165]. CD40 is expressed by osteoblastic cells and CD154 is anti-apoptotic in these cells [166]. Therefore, much remains to be found about the role of platelet CD154 in tissue repair. As CD40 is largely distributed, platelet CD154 could be conjectured to be generally involved, to one degree or another, in tissue repair.

Platelet CD154 as a mediator of tissue injury

The model of platelets promoting tissue repair is to be compared to their deleterious role in acute and chronic tissue injury. Difficult points are raised by this friend or foe facet, implicating balanced therapeutic approaches [119]. Ischemia/reperfusion (I/R) underscores platelet deleterious role, and the importance to control platelet activation in this context. In I/R, platelet activation in the microcirculation vascular bed leads to tissue injury, as shown in lung, liver or kidney. Platelet depletion or antiplatelet treatments are protective in several experimental I/R models [167-169]; CD154 is contributing: mice deficient in CD154 are protected from I/R-mediated injury in brain, lung, liver or intestine; in lung I/R-mediated injury platelet CD154 is specifically contributing [170-172].

Platelet CD154 and the immune response: unanticipated new frontiers

Platelets participate to the control of infection via direct and indirect mechanisms [6,173-178]. The significance of platelet Toll-like receptors (TLR) has been emphasized; TLR ligation activates platelet secretion of mediators regulating the immune response, including sCD154 [6,179-184]. Platelets also regulate several steps of the adaptative immune response [6,182-194]. Moreover, platelets can present antigen [195]; they express MHC class I molecules and T cell costimulatory molecules, including CD86 and CD40 and harbor a functional proteasome [196-199]. Among platelet mediators, CD154 proved to be critical in linking platelet and immunity (Figure 3).

Figure 3
figure 3

Platelet CD154 contributes to the host defense against infections. Infection triggers inflammation and coagulation. The interaction with pathogens, pathogen-derived molecules such as lipopolysaccharide (LPS), inflammation and coagulation concur to activate platelets, leading to CD154 display at the platelet membrane and the release of soluble CD154 (sCD154). Multiple inputs amplify the platelet activation scenario, including soluble and cellular effectors of the inflammatory network. Platelet CD154 targets several immune response effectors, including contribution to the chemotactic recruitment (dotted semicircles symbolize chemotaxis) of leukocytes to sites of infection, e.g. through the induction of adhesion molecules on EC (CD62e, CD54, CD106) and activation/upregulation of integrins such as αMβ2 on neutrophils [158,200]. CD40 triggering is a major inducer of pathogen-killing mechanisms by phagocytic cells. These responses are amplified by inflammatory mediators generated upon CD40 ligation; this schematic representation does not represent all interfaces that are directly or indirectly regulated by platelet CD154. Platelet CD154 influences the adaptative immune response, through several mechanisms, including the activation/maturation of antigen presenting cells (see text for details). Magenta arrows depict interaction with CD40. Abbreviations: PAMPs, pathogen-associated molecular patterns; PRR, pathogen recognition receptors; TLR, Toll-like receptors.

Although much remains to be understood, particularly with reference to the innate immune response, the specific role of platelet CD154 in immunity is strengthening. Several pathogen-clearing mechanisms are stimulated by CD154, including platelet aggregation [173], phagocytosis and production of defense proteins, such as complement proteins and interferon-α, by cells of the innate immune system [6,20,201]. CD40 contributes to the regulation of innate immune response, including induction of TLR expression, cooperation in TLR-mediated B cell activation, engagement in the crosstalk between intracellular MHC class II molecules and TLR signaling pathway [202-204]. The specific role of platelet CD154 in these mechanisms remains to be precised. However, it is now appreciated that platelet CD154 controls many facets of the interface between innate and adaptive immune responses [173,187,191,205]. Platelet CD154 induces DC maturation, can activate B cells, antibody production and isotype switching, contributes to germinal center formation, and enhances CD8+ T cell responses [188,206-213]. Platelet CD154 helps mounting a protective cytotoxic T cell immune response to viral or bacterial challenge [206,214]. Platelet CD154 may promote the immune response in the context of low antigen challenge by lowering the antigen threshold, and improve B cell response in regulatory T-cell limiting settings [210,215]. Further, sCD154 per se induces cardiac allograft rejection [212]. Many questions remain. How platelet CD154 enters the draining lymph nodes to regulate the adaptive immune response machinery is not known; PMP may convey this information, as CD154 associated to PMP is functional: it enhances DC activation, germinal center formation, B cell proliferation and IgG production [63,216]. Several questions are also raised with reference to platelet CD154 in autoimmunity; this “dark side” [14,217] feature of platelet CD154 is a recently opened frontier. Platelet CD154 is competent to increase production of antiplatelet antibodies in immune thrombocytopenic purpura [68] and, in systemic lupus erythematosus, platelet CD154 activates antigen presenting cells contributing to enhanced interferon-α production [218].

Platelet CD154: a new hematopoietic regulator?

Hematopoiesis can be adapted in response to inflammation/infection by signals generated at bone marrow distal sites [219-224]. Platelets are activated at sites of inflammation/infection and are a major source of circulating sCD154. Could platelets deliver a CD154 signal, through sCD154, platelet- or PMP-associated CD154 that regulates hematopoiesis? Platelet mediators enhance hematopoietic stem cell proliferation and platelet-derived signals may contribute to CD34+ cell mobilization [225,226]. Several studies have demonstrated CD154 involvement in hematopoiesis. CD154 regulation of early B cell lymphopoiesis is suggested by the sCD154-induced increased number of B cell progenitors (BCP) in mice after bone marrow transplantation (BMT) [227]. CD40 is expressed on BCP, and a positive effect of CD40 ligation on BCP proliferation can be observed on pre- and immature B cells in human and pro-B cells in the mouse [228,229]. In the mouse, there is clear experimental evidence for a positive role of CD154 in B cell hematopoiesis and, particularly in stress conditions, as after BMT [229]. However, normal numbers of circulating B cells in patients with X-linked hyper-IgM syndrome would rule out an absolute requirement for the CD154/CD40 signaling in early B cell development. CD154 may therefore mostly play a significant role in emergency B cell hematopoiesis [229]. More is known about CD154 regulation of the lymphoid system maturation, which has been fully reviewed [230]. A role for platelet CD154 on myelopoiesis is suggested by the sCD154-mediated increased granulocyte and platelet recovery after BMT in the mouse and by the neutropenia and thrombocytopenia observed in patients with X-linked hyper-IgM syndrome [227]. In vitro, sCD154 promotes the differentiation of CD34+ cells towards the granulocytic/monocytic and megakaryocytic lineages in CD34+/stromal cell cocultures. The mode of action of sCD154 appears to be essentially indirect, through the induction of hematopoietic cytokines by bone marrow stromal cells [231,232]. Platelet CD154 may therefore play a role in regulating emergency hematopoiesis. However, many questions remain unsolved, particularly which and how platelet CD154 signals could be delivered and interact with bone marrow stem/progenitor cells.

Platelet CD154 and cancer: a rapidly expanding frontier

There is strong evidence for the involvement of platelets in cancer progression; mechanisms are multiple [233-240]. Platelets are activated in the tumor environment and bind tumor cells. Mediators released upon platelet activation are key to tumor angiogenesis [241,242] and are likely to contribute to the tumor-supporting inflammatory environment [243,244]. Platelets play a positive role in metastasis [234,238,245-249]. However, this may not be true for all organs [250]. In hematogenous dissemination, platelet/cancer cell microthrombi provide protection, including shielding from shear flow, or immune evasion; during the arrest and extravasation phases, platelet mediators facilitate tumor cell arrest on EC, extravasation, survival and growth after seeding [251]. Platelet MPs are also contributing [124,252,253].

Many tumor cells express CD40. The outcomes of CD40 ligation on tumor cells are ambivalent depending on the models studied. In one hand, CD40 ligation promotes anti-tumor immune surveillance through a variety of mechanisms including antigen-presenting cell activation, restoration of malignant cell immune recognition, activation of tumoricidal-infiltrating macrophages, immunostimulatory cytokine production. CD40 ligation also induces tumor growth arrest and sensitization to apoptotic signals. On the other hand, CD40 ligation has positive consequences on tumor growth, survival and resistance to chemotherapy and metastatic potential. The interpretation of CD154 effects on cancer cells is made complex, first by the existence of several receptors for CD154, potentially explaining variable outcomes of CD154 treatment of tumor cells, and second, by the difficulty in assessing direct versus indirect effects. The contribution of the CD40 signaling in cancer, and prospects offered by targeting the CD40 signaling for cancer treatment have recently been underlined and reviewed [254-258]. However, the specific role played by platelet CD154 remains a new important frontier. If platelet activation is likely to result in expression of CD154 and generation of sCD154 in the tumor cell environment, this study is made complex as there are extra platelet sources of CD154.

Conclusion

There have been recent and rapid advances in our current knowledge of the non-hemostatic functions of platelets, placing them in the middle of the spectrum of mechanisms that maintain homeostasis, and highlighting their role in a variety of inflammatory and immune disorders. However, platelets store and release such a wide diversity of biologically active mediators that major gaps remain in our understanding of which and how these mediators collectively fulfill these functions. Platelet CD154 has attracted considerable attention as it recapitulates several of non-hemostatic platelet attributes. Considering the large number of different cells expressing CD40, the complex signaling cascade and the wide range of effectors activated by the CD154/CD40 interaction, it can be anticipated that future investigations will further extend the contribution of platelet CD154 in health and disease. For example, recent publications on the CD154/CD40 dyad have pointed to its role in obesity and hepatic steatosis [259-263], and it is tempting to speculate that platelet CD154 contributes to metabolic homeostasis. In the same direction, the number of physiological or pathological conditions associated with platelet activation is enlarging. For example, platelet activation has been found associated to aging, to emotional or environmental stresses…; platelet CD154 might represent a significant link between these conditions and accompanying pathologies, such as cardiovascular events [264]. However, platelet CD154 is always acting in a multicytokine context, including inhibitors and activators released at the same time by platelets; understanding how this complexity is tuned and evidencing the specific role of platelet CD154 remains a difficult challenge.

References

  1. Kaushansky K. The molecular mechanisms that control thrombopoiesis. J Clin Invest. 2005;115(12):3339–47.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  2. Thon JN, Italiano JE. Platelet formation. Semin Hematol. 2010;47(3):220–6.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  3. Machlus KR, Italiano Jr JE. The incredible journey: From megakaryocyte development to platelet formation. J Cell Biol. 2013;201(6):785–96.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  4. Schmaier AA, Stalker TJ, Runge JJ, Lee D, Nagaswami C, Mericko P, et al. Occlusive thrombi arise in mammals but not birds in response to arterial injury: evolutionary insight into human cardiovascular disease. Blood. 2011;118(13):3661–9.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  5. Weyrich AS, Lindemann S, Zimmerman GA. The evolving role of platelets in inflammation. J Thromb Haemost. 2003;1(9):1897–905.

    Article  CAS  PubMed  Google Scholar 

  6. Semple JW, Italiano Jr JE, Freedman J. Platelets and the immune continuum. Nat Rev Immunol. 2011;11(4):264–74.

    Article  CAS  PubMed  Google Scholar 

  7. Grozovsky R, Hoffmeister KM, Falet H. Novel clearance mechanisms of platelets. Curr Opin Hematol. 2010;17(6):585–9.

    Article  PubMed Central  PubMed  Google Scholar 

  8. Coller BS. Historical perspective and future directions in platelet research. J Thromb Haemost. 2011;9 Suppl 1:374–95.

    Article  PubMed Central  PubMed  Google Scholar 

  9. Grewal IS, Flavell RA. CD40 and CD154 in cell-mediated immunity. Annu Rev Immunol. 1998;16:111–35.

    Article  CAS  PubMed  Google Scholar 

  10. van Kooten C, Banchereau J. CD40-CD40 ligand. J Leukoc Biol. 2000;67(1):2–17.

    PubMed  Google Scholar 

  11. Howard LM, Miller SD. Immunotherapy targeting the CD40/CD154 costimulatory pathway for treatment of autoimmune disease. Autoimmunity. 2004;37(5):411–8.

    Article  CAS  PubMed  Google Scholar 

  12. Toubi E, Shoenfeld Y. The role of CD40-CD154 interactions in autoimmunity and the benefit of disrupting this pathway. Autoimmunity. 2004;37(6–7):457–64.

    Article  CAS  PubMed  Google Scholar 

  13. Law CL, Grewal IS. Therapeutic interventions targeting CD40L (CD154) and CD40: the opportunities and challenges. Adv Exp Med Biol. 2009;647:8–36.

    Article  CAS  PubMed  Google Scholar 

  14. Peters AL, Stunz LL, Bishop GA. CD40 and autoimmunity: the dark side of a great activator. Semin Immunol. 2009;21(5):293–300.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  15. Alaaeddine N, Hassan GS, Yacoub D, Mourad W. CD154: an immunoinflammatory mediator in systemic lupus erythematosus and rheumatoid arthritis. Clin Dev Immunol. 2012;2012:490148.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  16. Hollenbaugh D, Mischel-Petty N, Edwards CP, Simon JC, Denfeld RW, Kiener PA, et al. Expression of functional CD40 by vascular endothelial cells. J Exp Med. 1995;182(1):33–40.

    Article  CAS  PubMed  Google Scholar 

  17. Karmann K, Hughes CC, Schechner J, Fanslow WC, Pober JS. CD40 on human endothelial cells: inducibility by cytokines and functional regulation of adhesion molecule expression. Proc Natl Acad Sci U S A. 1995;92(10):4342–6.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  18. Yellin MJ, Brett J, Baum D, Matsushima A, Szabolcs M, Stern D, et al. Functional interactions of T cells with endothelial cells: the role of CD40L-CD40-mediated signals. J Exp Med. 1995;182(6):1857–64.

    Article  CAS  PubMed  Google Scholar 

  19. Schonbeck U, Libby P. CD40 signaling and plaque instability. Circ Res. 2001;89(12):1092–103.

    Article  CAS  PubMed  Google Scholar 

  20. Schonbeck U, Libby P. The CD40/CD154 receptor/ligand dyad. Cell Mol Life Sci. 2001;58(1):4–43.

    Article  CAS  PubMed  Google Scholar 

  21. Delmas Y, Viallard JF, Solanilla A, Villeneuve J, Pasquet JM, Belloc F, et al. Activation of mesangial cells by platelets in systemic lupus erythematosus via a CD154-dependent induction of CD40. Kidney Int. 2005;68(5):2068–78.

    Article  CAS  PubMed  Google Scholar 

  22. Andre P, Prasad KS, Denis CV, He M, Papalia JM, Hynes RO, et al. CD40L stabilizes arterial thrombi by a beta3 integrin–dependent mechanism. Nat Med. 2002;8(3):247–52.

    Article  CAS  PubMed  Google Scholar 

  23. Leveille C, Bouillon M, Guo W, Bolduc J, Sharif-Askari E, El-Fakhry Y, et al. CD40 ligand binds to alpha5beta1 integrin and triggers cell signaling. J Biol Chem. 2007;282(8):5143–51.

    Article  CAS  PubMed  Google Scholar 

  24. Zirlik A, Maier C, Gerdes N, MacFarlane L, Soosairajah J, Bavendiek U, et al. CD40 ligand mediates inflammation independently of CD40 by interaction with Mac-1. Circulation. 2007;115(12):1571–80.

    Article  CAS  PubMed  Google Scholar 

  25. Hassan GS, Merhi Y, Mourad WM. CD154 and its receptors in inflammatory vascular pathologies. Trends Immunol. 2009;30(4):165–72.

    Article  CAS  PubMed  Google Scholar 

  26. Graf D, Muller S, Korthauer U, van Kooten C, Weise C, Kroczek RA. A soluble form of TRAP (CD40 ligand) is rapidly released after T cell activation. Eur J Immunol. 1995;25(6):1749–54.

    Article  CAS  PubMed  Google Scholar 

  27. Peitsch MC, Jongeneel CV. A 3-D model for the CD40 ligand predicts that it is a compact trimer similar to the tumor necrosis factors. Int Immunol. 1993;5(2):233–8.

    Article  CAS  PubMed  Google Scholar 

  28. Fanslow WC, Srinivasan S, Paxton R, Gibson MG, Spriggs MK, Armitage RJ. Structural characteristics of CD40 ligand that determine biological function. Semin Immunol. 1994;6(5):267–78.

    Article  CAS  PubMed  Google Scholar 

  29. Karpusas M, Hsu YM, Wang JH, Thompson J, Lederman S, Chess L, et al. 2 A crystal structure of an extracellular fragment of human CD40 ligand. Structure. 1995;3(10):1031–9.

    Article  CAS  PubMed  Google Scholar 

  30. Pietravalle F, Lecoanet-Henchoz S, Blasey H, Aubry JP, Elson G, Edgerton MD, et al. Human native soluble CD40L is a biologically active trimer, processed inside microsomes. J Biol Chem. 1996;271(11):5965–7.

    Article  CAS  PubMed  Google Scholar 

  31. Bishop GA, Moore CR, Xie P, Stunz LL, Kraus ZJ. TRAF proteins in CD40 signaling. Adv Exp Med Biol. 2007;597:131–51.

    Article  PubMed  Google Scholar 

  32. Donners MM, Beckers L, Lievens D, Munnix I, Heemskerk J, Janssen BJ, et al. The CD40-TRAF6 axis is the key regulator of the CD40/CD40L system in neointima formation and arterial remodeling. Blood. 2008;111(9):4596–604.

    Article  CAS  PubMed  Google Scholar 

  33. Chatzigeorgiou A, Seijkens T, Zarzycka B, Engel D, Poggi M, van den Berg S, et al. Blocking CD40-TRAF6 signaling is a therapeutic target in obesity-associated insulin resistance. Proc Natl Acad Sci U S A. 2014;111(7):2686–91.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  34. Horrillo A, Fontela T, Arias-Salgado EG, Llobat D, Porras G, Ayuso MS, et al. Generation of mice with conditional ablation of the Cd40lg gene: new insights on the role of CD40L. Transgenic Res. 2014;23(1):53–66.

    Article  CAS  PubMed  Google Scholar 

  35. Henn V, Slupsky JR, Grafe M, Anagnostopoulos I, Forster R, Muller-Berghaus G, et al. CD40 ligand on activated platelets triggers an inflammatory reaction of endothelial cells. Nature. 1998;391(6667):591–4.

    Article  CAS  PubMed  Google Scholar 

  36. Kamykowski J, Carlton P, Sehgal S, Storrie B. Quantitative immunofluorescence mapping reveals little functional coclustering of proteins within platelet alpha-granules. Blood. 2011;118(5):1370–3.

    Article  CAS  PubMed  Google Scholar 

  37. Charafeddine AH, Kim EJ, Maynard DM, Yi H, Weaver TA, Gunay-Aygun M, et al. Platelet-derived CD154: ultrastructural localization and clinical correlation in organ transplantation. Am J Transplant. 2012;12(11):3143–51.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  38. Hermann A, Rauch BH, Braun M, Schror K, Weber AA. Platelet CD40 ligand (CD40L)–subcellular localization, regulation of expression, and inhibition by clopidogrel. Platelets. 2001;12(2):74–82.

    Article  CAS  PubMed  Google Scholar 

  39. Denis MM, Tolley ND, Bunting M, Schwertz H, Jiang H, Lindemann S, et al. Escaping the nuclear confines: signal-dependent pre-mRNA splicing in anucleate platelets. Cell. 2005;122(3):379–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Rowley JW, Schwertz H, Weyrich AS. Platelet mRNA: the meaning behind the message. Curr Opin Hematol. 2012;19(5):385–91.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  41. Reed GL, Fitzgerald ML, Polgar J. Molecular mechanisms of platelet exocytosis: insights into the “secrete” life of thrombocytes. Blood. 2000;96(10):3334–42.

    CAS  PubMed  Google Scholar 

  42. Jurk K, Kehrel BE. Platelets: physiology and biochemistry. Semin Thromb Hemost. 2005;31(4):381–92.

    Article  CAS  PubMed  Google Scholar 

  43. Ren Q, Ye S, Whiteheart SW. The platelet release reaction: just when you thought platelet secretion was simple. Curr Opin Hematol. 2008;15(5):537–41.

    Article  PubMed Central  PubMed  Google Scholar 

  44. Koseoglu S, Flaumenhaft R. Advances in platelet granule biology. Curr Opin Hematol. 2013;20(5):464–71.

    Article  PubMed  Google Scholar 

  45. Wijten P, van Holten T, Woo LL, Bleijerveld OB, Roest M, Heck AJ, et al. High precision platelet releasate definition by quantitative reversed protein profiling–brief report. Arterioscler Thromb Vasc Biol. 2013;33(7):1635–8.

    Article  CAS  PubMed  Google Scholar 

  46. Golebiewska EM, Poole AW. Secrets of platelet exocytosis - what do we really know about platelet secretion mechanisms? Br J Haematol. 2013;165(2):204–16.

    Article  PubMed Central  Google Scholar 

  47. Lindemann S, Gawaz M. The active platelet: translation and protein synthesis in an anucleate cell. Semin Thromb Hemost. 2007;33(2):144–50.

    Article  CAS  PubMed  Google Scholar 

  48. Weyrich AS, Schwertz H, Kraiss LW, Zimmerman GA. Protein synthesis by platelets: historical and new perspectives. J Thromb Haemost. 2009;7(2):241–6.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  49. Aukrust P, Muller F, Ueland T, Berget T, Aaser E, Brunsvig A, et al. Enhanced levels of soluble and membrane-bound CD40 ligand in patients with unstable angina. Possible reflection of T lymphocyte and platelet involvement in the pathogenesis of acute coronary syndromes. Circulation. 1999;100(6):614–20.

    Article  CAS  PubMed  Google Scholar 

  50. Henn V, Steinbach S, Buchner K, Presek P, Kroczek RA. The inflammatory action of CD40 ligand (CD154) expressed on activated human platelets is temporally limited by coexpressed CD40. Blood. 2001;98(4):1047–54.

    Article  CAS  PubMed  Google Scholar 

  51. Jin Y, Nonoyama S, Morio T, Imai K, Ochs HD, Mizutani S. Characterization of soluble CD40 ligand released from human activated platelets. J Med Dent Sci. 2001;48(1):23–7.

    CAS  PubMed  Google Scholar 

  52. Nannizzi-Alaimo L, Rubenstein MH, Alves VL, Leong GY, Phillips DR, Gold HK. Cardiopulmonary bypass induces release of soluble CD40 ligand. Circulation. 2002;105(24):2849–54.

    Article  CAS  PubMed  Google Scholar 

  53. Otterdal K, Pedersen TM, Solum NO. Release of soluble CD40 ligand after platelet activation: studies on the solubilization phase. Thromb Res. 2004;114(3):167–77.

    CAS  PubMed  Google Scholar 

  54. Furman MI, Krueger LA, Linden MD, Barnard MR, Frelinger 3rd AL, Michelson AD. Release of soluble CD40L from platelets is regulated by glycoprotein IIb/IIIa and actin polymerization. J Am Coll Cardiol. 2004;43(12):2319–25.

    Article  CAS  PubMed  Google Scholar 

  55. Menchen L, Marin-Jimenez I, Arias-Salgado EG, Fontela T, Hernandez-Sampelayo P, Rodriguez MC, et al. Matrix metalloproteinase 9 is involved in Crohn’s disease-associated platelet hyperactivation through the release of soluble CD40 ligand. Gut. 2009;58(7):920–8.

    Article  CAS  PubMed  Google Scholar 

  56. Reinboldt S, Wenzel F, Rauch BH, Hohlfeld T, Grandoch M, Fischer JW, et al. Preliminary evidence for a matrix metalloproteinase-2 (MMP-2)-dependent shedding of soluble CD40 ligand (sCD40L) from activated platelets. Platelets. 2009;20(6):441–4.

    Article  CAS  PubMed  Google Scholar 

  57. Choi WS, Jeon OH, Kim DS. CD40 ligand shedding is regulated by interaction between matrix metalloproteinase-2 and platelet integrin alpha(IIb)beta(3). J Thromb Haemost. 2010;8(6):1364–71.

    Article  CAS  PubMed  Google Scholar 

  58. Yacoub D, Benslimane N, Al-Zoobi L, Hassan G, Nadiri A, Mourad W. CD154 Is Released from T-cells by a Disintegrin and Metalloproteinase Domain-containing Protein 10 (ADAM10) and ADAM17 in a CD40 Protein-dependent Manner. J Biol Chem. 2013;288(50):36083–93.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  59. Nannizzi-Alaimo L, Alves VL, Phillips DR. Inhibitory effects of glycoprotein IIb/IIIa antagonists and aspirin on the release of soluble CD40 ligand during platelet stimulation. Circulation. 2003;107(8):1123–8.

    Article  CAS  PubMed  Google Scholar 

  60. Pignatelli P, Sanguigni V, Lenti L, Ferro D, Finocchi A, Rossi P, et al. gp91phox-dependent expression of platelet CD40 ligand. Circulation. 2004;110(10):1326–9.

    Article  CAS  PubMed  Google Scholar 

  61. Mazzei GJ, Edgerton MD, Losberger C, Lecoanet-Henchoz S, Graber P, Durandy A, et al. Recombinant soluble trimeric CD40 ligand is biologically active. J Biol Chem. 1995;270(13):7025–8.

    Article  CAS  PubMed  Google Scholar 

  62. Anand SX, Viles-Gonzalez JF, Badimon JJ, Cavusoglu E, Marmur JD. Membrane-associated CD40L and sCD40L in atherothrombotic disease. Thromb Haemost. 2003;90(3):377–84.

    CAS  PubMed  Google Scholar 

  63. 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;111(10):5028–36.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  64. Inwald DP, McDowall A, Peters MJ, Callard RE, Klein NJ. CD40 is constitutively expressed on platelets and provides a novel mechanism for platelet activation. Circ Res. 2003;92(9):1041–8.

    Article  CAS  PubMed  Google Scholar 

  65. Prasad KS, Andre P, He M, Bao M, Manganello J, Phillips DR. Soluble CD40 ligand induces beta3 integrin tyrosine phosphorylation and triggers platelet activation by outside-in signaling. Proc Natl Acad Sci U S A. 2003;100(21):12367–71.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  66. King SM, Reed GL. Development of platelet secretory granules. Semin Cell Dev Biol. 2002;13(4):293–302.

    Article  CAS  PubMed  Google Scholar 

  67. Schulze H, Shivdasani RA. Mechanisms of thrombopoiesis. J Thromb Haemost. 2005;3(8):1717–24.

    Article  CAS  PubMed  Google Scholar 

  68. Solanilla A, Pasquet JM, Viallard JF, Contin C, Grosset C, Dechanet-Merville J, et al. Platelet-associated CD154 in immune thrombocytopenic purpura. Blood. 2005;105(1):215–8.

    Article  CAS  PubMed  Google Scholar 

  69. Crist SA, Sprague DL, Ratliff TL. Nuclear factor of activated T cells (NFAT) mediates CD154 expression in megakaryocytes. Blood. 2008;111(7):3553–61.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  70. Crist SA, Elzey BD, Ahmann MT, Ratliff TL. Early growth response-1 (EGR-1) and nuclear factor of activated T cells (NFAT) cooperate to mediate CD40L expression in megakaryocytes and platelets. J Biol Chem. 2013;288(47):33985–96.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  71. Weyrich AS, Dixon DA, Pabla R, Elstad MR, McIntyre TM, Prescott SM, et al. Signal-dependent translation of a regulatory protein, Bcl-3, in activated human platelets. Proc Natl Acad Sci U S A. 1998;95(10):5556–61.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  72. Maguire PB, Fitzgerald DJ. Platelet proteomics. J Thromb Haemost. 2003;1(7):1593–601.

    Article  CAS  PubMed  Google Scholar 

  73. Gnatenko DV, Perrotta PL, Bahou WF. Proteomic approaches to dissect platelet function: Half the story. Blood. 2006;108(13):3983–91.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  74. Andre P, Nannizzi-Alaimo L, Prasad SK, Phillips DR. Platelet-derived CD40L: the switch-hitting player of cardiovascular disease. Circulation. 2002;106(8):896–9.

    Article  PubMed  Google Scholar 

  75. Viallard JF, Solanilla A, Gauthier B, Contin C, Dechanet J, Grosset C, et al. Increased soluble and platelet-associated CD40 ligand in essential thrombocythemia and reactive thrombocytosis. Blood. 2002;99(7):2612–4.

    Article  CAS  PubMed  Google Scholar 

  76. Nagasawa M, Zhu Y, Isoda T, Tomizawa D, Itoh S, Kajiwara M, et al. Analysis of serum soluble CD40 ligand (sCD40L) in the patients undergoing allogeneic stem cell transplantation: platelet is a major source of serum sCD40L. Eur J Haematol. 2005;74(1):54–60.

    Article  CAS  PubMed  Google Scholar 

  77. Feng X, Scheinberg P, Wu CO, Samsel L, Nunez O, Prince C, et al. Cytokine signature profiles in acquired aplastic anemia and myelodysplastic syndromes. Haematologica. 2011;96(4):602–6.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  78. Feng X, Scheinberg P, Samsel L, Rios O, Chen J, McCoy Jr JP, et al. Decreased plasma cytokines are associated with low platelet counts in aplastic anemia and immune thrombocytopenic purpura. J Thromb Haemost. 2012;10(8):1616–23.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  79. Fan Y, Ge Y, Zhu H, Wang Y, Yang B, Zhuang Y, et al. Characterization and application of two novel monoclonal antibodies against CD40L: epitope and functional studies on cell membrane CD40L and studies on the origin of soluble serum CD40L. Tissue Antigens. 2004;64(3):257–63.

    Article  CAS  PubMed  Google Scholar 

  80. Mason PJ, Chakrabarti S, Albers AA, Rex S, Vitseva O, Varghese S, et al. Plasma, serum, and platelet expression of CD40 ligand in adults with cardiovascular disease. Am J Cardiol. 2005;96(10):1365–9.

    Article  CAS  PubMed  Google Scholar 

  81. Cipollone F, Mezzetti A, Porreca E, Di Febbo C, Nutini M, Fazia M, et al. Association between enhanced soluble CD40L and prothrombotic state in hypercholesterolemia: effects of statin therapy. Circulation. 2002;106(4):399–402.

    Article  CAS  PubMed  Google Scholar 

  82. Riondino S, Martini F, La Farina F, Spila A, Guadagni F, Ferroni P. Increased plasma levels of soluble CD40 ligand correlate with platelet activation markers and underline the need for standardized pre-analytical conditions. Clin Biochem. 2010;43(7–8):666–70.

    Article  CAS  PubMed  Google Scholar 

  83. Burdess A, Michelsen AE, Brosstad F, Fox KA, Newby DE, Nimmo AF. Platelet activation in patients with peripheral vascular disease: reproducibility and comparability of platelet markers. Thromb Res. 2012;129(1):50–5.

    Article  CAS  PubMed  Google Scholar 

  84. Ahn ER, Lander G, Jy W, Bidot CJ, Jimenez JJ, Horstman LL, et al. Differences of soluble CD40L in sera and plasma: implications on CD40L assay as a marker of thrombotic risk. Thromb Res. 2004;114(2):143–8.

    Article  CAS  PubMed  Google Scholar 

  85. Thom J, Gilmore G, Yi Q, Hankey GJ, Eikelboom JW. Measurement of soluble P-selectin and soluble CD40 ligand in serum and plasma. J Thromb Haemost. 2004;2(11):2067–9.

    Article  CAS  PubMed  Google Scholar 

  86. Varo N, Nuzzo R, Natal C, Libby P, Schonbeck U. Influence of pre-analytical and analytical factors on soluble CD40L measurements. Clin Sci (Lond). 2006;111(5):341–7.

    Article  CAS  Google Scholar 

  87. Weber M, Rabenau B, Stanisch M, Elsaesser A, Mitrovic V, Heeschen C, et al. Influence of sample type and storage conditions on soluble CD40 ligand assessment. Clin Chem. 2006;52(5):888–91.

    Article  CAS  PubMed  Google Scholar 

  88. Weber M, Rabenau B, Stanisch M, Nef HM, Mollmann H, Elsasser A, et al. Influence of sample type on soluble CD40 ligand assessment in patients with acute coronary syndromes. Thromb Res. 2007;120(6):811–4.

    Article  CAS  PubMed  Google Scholar 

  89. Ivandic BT, Spanuth E, Haase D, Lestin HG, Katus HA. Increased plasma concentrations of soluble CD40 ligand in acute coronary syndrome depend on in vitro platelet activation. Clin Chem. 2007;53(7):1231–4.

    Article  CAS  PubMed  Google Scholar 

  90. Mobarrez F, Sjovik C, Soop A, Hallstrom L, Frostell C, Pisetsky DS et al. CD40L expression in plasma of volunteers following LPS administration: A comparison between assay of CD40L on platelet microvesicles and soluble CD40L. Platelets. 2014:1–5. [Epub ahead of print]

  91. Schonbeck U, Gerdes N, Varo N, Reynolds RS, Horton DB, Bavendiek U, et al. Oxidized low-density lipoprotein augments and 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors limit CD40 and CD40L expression in human vascular cells. Circulation. 2002;106(23):2888–93.

    Article  PubMed  CAS  Google Scholar 

  92. Semb AG, van Wissen S, Ueland T, Smilde T, Waehre T, Tripp MD, et al. Raised serum levels of soluble CD40 ligand in patients with familial hypercholesterolemia: downregulatory effect of statin therapy. J Am Coll Cardiol. 2003;41(2):275–9.

    Article  CAS  PubMed  Google Scholar 

  93. Li J, Zhao SP, Peng DQ, Xu ZM, Zhou HN. Early effect of pravastatin on serum soluble CD40L, matrix metalloproteinase-9, and C-reactive protein in patients with acute myocardial infarction. Clin Chem. 2004;50(9):1696–9.

    Article  CAS  PubMed  Google Scholar 

  94. Tamura N, Yoshida M, Ichikawa N, Handa M, Ikeda Y, Tanabe T, et al. Shear-induced von Willebrand factor-mediated platelet surface translocation of the CD40 ligand. Thromb Res. 2002;108(5–6):311–5.

    Article  PubMed  CAS  Google Scholar 

  95. Heijnen HF, Schiel AE, Fijnheer R, Geuze HJ, Sixma JJ. Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules. Blood. 1999;94(11):3791–9.

    CAS  PubMed  Google Scholar 

  96. Baj-Krzyworzeka M, Majka M, Pratico D, Ratajczak J, Vilaire G, Kijowski J, et al. Platelet-derived microparticles stimulate proliferation, survival, adhesion, and chemotaxis of hematopoietic cells. Exp Hematol. 2002;30(5):450–9.

    Article  CAS  PubMed  Google Scholar 

  97. May AE, Kälsch T, Massberg S, Herouy Y, Schmidt R, Gawaz M. Engagement of glycoprotein IIb/IIIa (aIIbb3) on platelets upregulates CD40L and triggers CD40L-dependent matrix degradation by endothelial cells. Circulation. 2002;106(16):2111–7.

    Article  CAS  PubMed  Google Scholar 

  98. Gear AR, Camerini D. Platelet chemokines and chemokine receptors: linking hemostasis, inflammation, and host defense. Microcirculation. 2003;10(3–4):335–50.

    Article  CAS  PubMed  Google Scholar 

  99. Dechanet J, Grosset C, Taupin JL, Merville P, Banchereau J, Ripoche J, et al. CD40 ligand stimulates proinflammatory cytokine production by human endothelial cells. J Immunol. 1997;159(11):5640–7.

    CAS  PubMed  Google Scholar 

  100. Gawaz M, Langer H, May AE. Platelets in inflammation and atherogenesis. J Clin Invest. 2005;115(12):3378–84.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  101. Davi G, Patrono C. Platelet activation and atherothrombosis. N Engl J Med. 2007;357(24):2482–94.

    Article  CAS  PubMed  Google Scholar 

  102. Projahn D, Koenen RR. Platelets: key players in vascular inflammation. J Leukoc Biol. 2012;92(6):1167–75.

    Article  CAS  PubMed  Google Scholar 

  103. Rondina MT, Weyrich AS, Zimmerman GA. Platelets as cellular effectors of inflammation in vascular diseases. Circ Res. 2013;112(11):1506–19.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  104. Mach F, Schonbeck U, Libby P. CD40 signaling in vascular cells: a key role in atherosclerosis? Atherosclerosis. 1998;137(Suppl):S89–95.

    Article  CAS  PubMed  Google Scholar 

  105. Mach F, Schonbeck U, Sukhova GK, Atkinson E, Libby P. Reduction of atherosclerosis in mice by inhibition of CD40 signalling. Nature. 1998;394(6689):200–3.

    Article  CAS  PubMed  Google Scholar 

  106. Danese S, Fiocchi C. Platelet activation and the CD40/CD40 ligand pathway: mechanisms and implications for human disease. Crit Rev Immunol. 2005;25(2):103–21.

    Article  CAS  PubMed  Google Scholar 

  107. Antoniades C, Bakogiannis C, Tousoulis D, Antonopoulos AS, Stefanadis C. The CD40/CD40 ligand system: linking inflammation with atherothrombosis. J Am Coll Cardiol. 2009;54(8):669–77.

    Article  CAS  PubMed  Google Scholar 

  108. Lievens D, Eijgelaar WJ, Biessen EA, Daemen MJ, Lutgens E. The multi-functionality of CD40L and its receptor CD40 in atherosclerosis. Thromb Haemost. 2009;102(2):206–14.

    CAS  PubMed  Google Scholar 

  109. Lievens D, Zernecke A, Seijkens T, Soehnlein O, Beckers L, Munnix IC, et al. Platelet CD40L mediates thrombotic and inflammatory processes in atherosclerosis. Blood. 2010;116(20):4317–27.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  110. Czapiga M, Gao JL, Kirk A, Lekstrom-Himes J. Human platelets exhibit chemotaxis using functional N-formyl peptide receptors. Exp Hematol. 2005;33(1):73–84.

    Article  CAS  PubMed  Google Scholar 

  111. Kiener PA, Moran-Davis P, Rankin BM, Wahl AF, Aruffo A, Hollenbaugh D. Stimulation of CD40 with purified soluble gp39 induces proinflammatory responses in human monocytes. J Immunol. 1995;155(10):4917–25.

    CAS  PubMed  Google Scholar 

  112. Danese S, de la Motte C, Sturm A, Vogel JD, West GA, Strong SA, et al. Platelets trigger a CD40-dependent inflammatory response in the microvasculature of inflammatory bowel disease patients. Gastroenterology. 2003;124(5):1249–64.

    Article  CAS  PubMed  Google Scholar 

  113. Kornerup KN, Page CP. The role of platelets in the pathophysiology of asthma. Platelets. 2007;18(5):319–28.

    Article  CAS  PubMed  Google Scholar 

  114. Tabuchi A, Kuebler WM. Endothelium-platelet interactions in inflammatory lung disease. Vascul Pharmacol. 2008;49(4–6):141–50.

    Article  CAS  PubMed  Google Scholar 

  115. Yoshida H, Granger DN. Inflammatory bowel disease: a paradigm for the link between coagulation and inflammation. Inflamm Bowel Dis. 2009;15(8):1245–55.

    Article  PubMed Central  PubMed  Google Scholar 

  116. Ripoche J. Blood platelets and inflammation: their relationship with liver and digestive diseases. Clin Res Hepatol Gastroenterol. 2011;35(5):353–7.

    Article  CAS  PubMed  Google Scholar 

  117. Boilard E, Blanco P, Nigrovic PA. Platelets: active players in the pathogenesis of arthritis and SLE. Nat Rev Rheumatol. 2012;8(9):534–42.

    Article  CAS  PubMed  Google Scholar 

  118. Santilli F, Vazzana N, Liani R, Guagnano MT, Davi G. Platelet activation in obesity and metabolic syndrome. Obes Rev. 2012;13(1):27–42.

    Article  CAS  PubMed  Google Scholar 

  119. Gasparyan AY, Ayvazyan L, Pretorius E, Kitas GD. Platelets in Rheumatic Diseases: Friend or Foe? Curr Pharm Des. 2014;20(4):552–66.

    Article  CAS  PubMed  Google Scholar 

  120. Langer HF, Chavakis T. Platelets and neurovascular inflammation. Thromb Haemost. 2013;110(5):888–93.

    Article  CAS  PubMed  Google Scholar 

  121. Kato K, Santana-Sahagùn E, Rassenti LZ, Weisman MH, Tamura N, Kobayashi S, et al. The soluble CD40 ligand sCD154 in systemic lupus erythematosus. J Clin Invest. 1999;104(7):947–55.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  122. Diamant M, Tushuizen ME, Sturk A, Nieuwland R. Cellular microparticles: new players in the field of vascular disease? Eur J Clin Invest. 2004;34(6):392–401.

    Article  CAS  PubMed  Google Scholar 

  123. Tan KT, Lip GY. The potential role of platelet microparticles in atherosclerosis. Thromb Haemost. 2005;94(3):488–92.

    CAS  PubMed  Google Scholar 

  124. Varon D, Shai E. Role of platelet-derived microparticles in angiogenesis and tumor progression. Discov Med. 2009;8(43):237–41.

    PubMed  Google Scholar 

  125. Boilard E, Nigrovic PA, Larabee K, Watts GF, Coblyn JS, Weinblatt ME, et al. Platelets amplify inflammation in arthritis via collagen-dependent microparticle production. Science. 2010;327(5965):580–3.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  126. Shantsila E, Kamphuisen PW, Lip GY. Circulating microparticles in cardiovascular disease: implications for atherogenesis and atherothrombosis. J Thromb Haemost. 2010;8(11):2358–68.

    Article  CAS  PubMed  Google Scholar 

  127. Burger D, Schock S, Thompson CS, Montezano AC, Hakim AM, Touyz RM. Microparticles: biomarkers and beyond. Clin Sci (Lond). 2013;124(7):423–41.

    Article  CAS  Google Scholar 

  128. Burnouf T, Goubran HA, Chou ML, Devos D, Radosevic M. Platelet microparticles: detection and assessment of their paradoxical functional roles in disease and regenerative medicine. Blood Rev. 2014;28(4):155–66.

    Article  CAS  PubMed  Google Scholar 

  129. Nathan C. Points of control in inflammation. Nature. 2002;420(6917):846–52.

    Article  CAS  PubMed  Google Scholar 

  130. Barton GM. A calculated response: control of inflammation by the innate immune system. J Clin Invest. 2008;118(2):413–20.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  131. Medzhitov R. Origin and physiological roles of inflammation. Nature. 2008;454(7203):428–35.

    Article  CAS  PubMed  Google Scholar 

  132. Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol. 2005;6(12):1191–7.

    Article  CAS  PubMed  Google Scholar 

  133. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature. 2008;453(7193):314–21.

    Article  CAS  PubMed  Google Scholar 

  134. Nurden AT. Platelets, inflammation and tissue regeneration. Thromb Haemost. 2011;105 Suppl 1:S13–33.

    Article  CAS  PubMed  Google Scholar 

  135. Gawaz M, Vogel S. Platelets in tissue repair: control of apoptosis and interactions with regenerative cells. Blood. 2013;122(15):2550–4.

    Article  CAS  PubMed  Google Scholar 

  136. Ho-Tin-Noe B, Demers M, Wagner DD. How platelets safeguard vascular integrity. J Thromb Haemost. 2011;9 Suppl 1:56–65.

    Article  PubMed Central  PubMed  Google Scholar 

  137. Verheul HM, Jorna AS, Hoekman K, Broxterman HJ, Gebbink MF, Pinedo HM. Vascular endothelial growth factor-stimulated endothelial cells promote adhesion and activation of platelets. Blood. 2000;96(13):4216–21.

    CAS  PubMed  Google Scholar 

  138. Brill A, Elinav H, Varon D. Differential role of platelet granular mediators in angiogenesis. Cardiovasc Res. 2004;63(2):226–35.

    Article  CAS  PubMed  Google Scholar 

  139. Klement GL, Yip TT, Cassiola F, Kikuchi L, Cervi D, Podust V, et al. Platelets actively sequester angiogenesis regulators. Blood. 2009;113(12):2835–42.

    Article  CAS  PubMed  Google Scholar 

  140. Lesurtel M, Graf R, Aleil B, Walther DJ, Tian Y, Jochum W, et al. Platelet-derived serotonin mediates liver regeneration. Science. 2006;312(5770):104–7.

    Article  CAS  PubMed  Google Scholar 

  141. Markiewski MM, DeAngelis RA, Lambris JD. Liver inflammation and regeneration: two distinct biological phenomena or parallel pathophysiologic processes? Mol Immunol. 2006;43(1–2):45–56.

    Article  CAS  PubMed  Google Scholar 

  142. Nocito A, Georgiev P, Dahm F, Jochum W, Bader M, Graf R, et al. Platelets and platelet-derived serotonin promote tissue repair after normothermic hepatic ischemia in mice. Hepatology. 2007;45(2):369–76.

    Article  CAS  PubMed  Google Scholar 

  143. Doukas J, Blease K, Craig D, Ma C, Chandler LA, Sosnowski BA, et al. Delivery of FGF genes to wound repair cells enhances arteriogenesis and myogenesis in skeletal muscle. Mol Ther. 2002;5(5 Pt 1):517–27.

    Article  CAS  PubMed  Google Scholar 

  144. Norazit A, Nguyen MN, Dickson CG, Tuxworth G, Goss B, Mackay-Sim A, et al. Vascular endothelial growth factor and platelet derived growth factor modulates the glial response to a cortical stab injury. Neuroscience. 2011;192:652–60.

    Article  CAS  PubMed  Google Scholar 

  145. Kim HK, Song KS, Chung JH, Lee KR, Lee SN. Platelet microparticles induce angiogenesis in vitro. Br J Haematol. 2004;124(3):376–84.

    Article  PubMed  Google Scholar 

  146. Brill A, Dashevsky O, Rivo J, Gozal Y, Varon D. Platelet-derived microparticles induce angiogenesis and stimulate post-ischemic revascularization. Cardiovasc Res. 2005;67(1):30–8.

    Article  CAS  PubMed  Google Scholar 

  147. Italiano Jr JE, Mairuhu AT, Flaumenhaft R. Clinical relevance of microparticles from platelets and megakaryocytes. Curr Opin Hematol. 2010;17(6):578–84.

    Article  PubMed Central  PubMed  Google Scholar 

  148. Mause SF, Ritzel E, Liehn EA, Hristov M, Bidzhekov K, Muller-Newen G, et al. Platelet microparticles enhance the vasoregenerative potential of angiogenic early outgrowth cells after vascular injury. Circulation. 2010;122(5):495–506.

    Article  PubMed  Google Scholar 

  149. Hayon Y, Shai E, Varon D, Leker RR. The role of platelets and their microparticles in rehabilitation of ischemic brain tissue. CNS Neurol Disord Drug Targets. 2012;11(7):921–5.

    Article  CAS  PubMed  Google Scholar 

  150. Anitua E, Andia I, Ardanza B, Nurden P, Nurden AT. Autologous platelets as a source of proteins for healing and tissue regeneration. Thromb Haemost. 2004;91(1):4–15.

    CAS  PubMed  Google Scholar 

  151. Langer HF, Gawaz M. Platelets in regenerative medicine. Basic Res Cardiol. 2008;103(4):299–307.

    Article  CAS  PubMed  Google Scholar 

  152. Nurden AT, Nurden P, Sanchez M, Andia I, Anitua E. Platelets and wound healing. Front Biosci. 2008;13:3532–48.

    PubMed  Google Scholar 

  153. Burnouf T, Goubran HA, Chen TM, Ou KL, El-Ekiaby M, Radosevic M. Blood-derived biomaterials and platelet growth factors in regenerative medicine. Blood Rev. 2013;27(2):77–89.

    Article  CAS  PubMed  Google Scholar 

  154. Textor J. Platelet-Rich Plasma (PRP) as a Therapeutic Agent: Platelet Biology, Growth Factors and a Review of the Literature. In: Andrade Santana MH, Dias Belangero W, Malheiros Luzo AC, editors. Lana JFSD. Springer Berlin Heidelberg: Platelet-Rich Plasma. Lecture Notes in Bioengineering; 2014. p. 61–94.

    Google Scholar 

  155. Mach F, Schonbeck U, Fabunmi RP, Murphy C, Atkinson E, Bonnefoy JY, et al. T lymphocytes induce endothelial cell matrix metalloproteinase expression by a CD40L-dependent mechanism: implications for tubule formation. Am J Pathol. 1999;154(1):229–38.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  156. Melter M, Reinders ME, Sho M, Pal S, Geehan C, Denton MD, et al. Ligation of CD40 induces the expression of vascular endothelial growth factor by endothelial cells and monocytes and promotes angiogenesis in vivo. Blood. 2000;96(12):3801–8.

    CAS  PubMed  Google Scholar 

  157. Deregibus MC, Buttiglieri S, Russo S, Bussolati B, Camussi G. CD40-dependent activation of phosphatidylinositol 3-kinase/Akt pathway mediates endothelial cell survival and in vitro angiogenesis. J Biol Chem. 2003;278(20):18008–14.

    Article  CAS  PubMed  Google Scholar 

  158. Li G, Sanders JM, Bevard MH, Sun Z, Chumley JW, Galkina EV, et al. CD40 ligand promotes Mac-1 expression, leukocyte recruitment, and neointima formation after vascular injury. Am J Pathol. 2008;172(4):1141–52.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  159. Song Z, Jin R, Yu S, Nanda A, Granger DN, Li G. Crucial role of CD40 signaling in vascular wall cells in neointimal formation and vascular remodeling after vascular interventions. Arterioscler Thromb Vasc Biol. 2012;32(1):50–64.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  160. Urbich C, Dernbach E, Aicher A, Zeiher AM, Dimmeler S. CD40 ligand inhibits endothelial cell migration by increasing production of endothelial reactive oxygen species. Circulation. 2002;106(8):981–6.

    Article  CAS  PubMed  Google Scholar 

  161. Hristov M, Gumbel D, Lutgens E, Zernecke A, Weber C. Soluble CD40 ligand impairs the function of peripheral blood angiogenic outgrowth cells and increases neointimal formation after arterial injury. Circulation. 2010;121(2):315–24.

    Article  CAS  PubMed  Google Scholar 

  162. Bou Khzam L, Boulahya R, Abou-Saleh H, Hachem A, Zaid Y, Merhi Y. Soluble CD40 ligand stimulates the pro-angiogenic function of peripheral blood angiogenic outgrowth cells via increased release of matrix metalloproteinase-9. PLoS One. 2013;8(12):e84289.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  163. Peguet-Navarro J, Dalbiez-Gauthier C, Moulon C, Berthier O, Reano A, Gaucherand M, et al. CD40 ligation of human keratinocytes inhibits their proliferation and induces their differentiation. J Immunol. 1997;158(1):144–52.

    CAS  PubMed  Google Scholar 

  164. Lopez-Granados E, Temmerman ST, Wu L, Reynolds JC, Follmann D, Liu S, et al. Osteopenia in X-linked hyper-IgM syndrome reveals a regulatory role for CD40 ligand in osteoclastogenesis. Proc Natl Acad Sci U S A. 2007;104(12):5056–61.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  165. Li Y, Toraldo G, Li A, Yang X, Zhang H, Qian W, et al. B cells and T cells are critical for the preservation of bone homeostasis and attainment of peak bone mass in vivo. Blood. 2007;109(9):3839–48.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  166. Ahuja SS, Zhao S, Bellido T, Plotkin LI, Jimenez F, Bonewald LF. CD40 ligand blocks apoptosis induced by tumor necrosis factor alpha, glucocorticoids, and etoposide in osteoblasts and the osteocyte-like cell line murine long bone osteocyte-Y4. Endocrinology. 2003;144(5):1761–9.

    Article  CAS  PubMed  Google Scholar 

  167. Bozza FA, Shah AM, Weyrich AS, Zimmerman GA. Amicus or adversary: platelets in lung biology, acute injury, and inflammation. Am J Respir Cell Mol Biol. 2009;40(2):123–34.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  168. Hu H, Batteux F, Chereau C, Kavian N, Marut W, Gobeaux C, et al. Clopidogrel protects from cell apoptosis and oxidative damage in a mouse model of renal ischaemia-reperfusion injury. J Pathol. 2011;225(2):265–75.

    Article  CAS  PubMed  Google Scholar 

  169. Dixon JT, Gozal E, Roberts AM. Platelet-mediated vascular dysfunction during acute lung injury. Arch Physiol Biochem. 2012;118(2):72–82.

    Article  CAS  PubMed  Google Scholar 

  170. Ishikawa M, Vowinkel T, Stokes KY, Arumugam TV, Yilmaz G, Nanda A, et al. CD40/CD40 ligand signaling in mouse cerebral microvasculature after focal ischemia/reperfusion. Circulation. 2005;111(13):1690–6.

    Article  CAS  PubMed  Google Scholar 

  171. Ke B, Shen XD, Gao F, Tsuchihashi S, Farmer DG, Briscoe D, et al. The CD154-CD40 T-cell co-stimulation pathway in liver ischemia and reperfusion inflammatory responses. Transplantation. 2005;79(9):1078–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Lapchak PH, Ioannou A, Kannan L, Rani P, Dalle Lucca JJ, Tsokos GC. Platelet-associated CD40/CD154 mediates remote tissue damage after mesenteric ischemia/reperfusion injury. PLoS One. 2012;7(2):e32260.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  173. Weyrich AS, Zimmerman GA. Platelets: signaling cells in the immune continuum. Trends Immunol. 2004;25(9):489–95.

    Article  CAS  PubMed  Google Scholar 

  174. Fitzgerald JR, Foster TJ, Cox D. The interaction of bacterial pathogens with platelets. Nat Rev Microbiol. 2006;4(6):445–57.

    Article  CAS  PubMed  Google Scholar 

  175. Flaujac C, Boukour S, Cramer-Borde E. Platelets and viruses: an ambivalent relationship. Cell Mol Life Sci. 2010;67(4):545–56.

    Article  CAS  PubMed  Google Scholar 

  176. Speth C, Loffler J, Krappmann S, Lass-Florl C, Rambach G. Platelets as immune cells in infectious diseases. Future Microbiol. 2013;8(11):1431–51.

    Article  CAS  PubMed  Google Scholar 

  177. Herter JM, Rossaint J, Zarbock A. Platelets in inflammation and immunity. J Thromb Haemost. 2014;12(11):1764–75.

    Article  CAS  PubMed  Google Scholar 

  178. Yeaman MR. Platelets: at the nexus of antimicrobial defence. Nat Rev Microbiol. 2014;12(6):426–37.

    Article  CAS  PubMed  Google Scholar 

  179. Klinger MH, Jelkmann W. Role of blood platelets in infection and inflammation. J Interferon Cytokine Res. 2002;22(9):913–22.

    Article  CAS  PubMed  Google Scholar 

  180. Shiraki R, Inoue N, Kawasaki S, Takei A, Kadotani M, Ohnishi Y, et al. Expression of Toll-like receptors on human platelets. Thromb Res. 2004;113(6):379–85.

    Article  CAS  PubMed  Google Scholar 

  181. Cognasse F, Hamzeh-Cognasse H, Lafarge S, Delezay O, Pozzetto B, McNicol A, et al. Toll-like receptor 4 ligand can differentially modulate the release of cytokines by human platelets. Br J Haematol. 2008;141(1):84–91.

    Article  CAS  PubMed  Google Scholar 

  182. Semple JW, Freedman J. Platelets and innate immunity. Cell Mol Life Sci. 2010;67(4):499–511.

    Article  CAS  PubMed  Google Scholar 

  183. Vieira-de-Abreu A, Campbell RA, Weyrich AS, Zimmerman GA. Platelets: versatile effector cells in hemostasis, inflammation, and the immune continuum. Semin Immunopathol. 2012;34(1):5–30.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  184. Jenne CN, Urrutia R, Kubes P. Platelets: bridging hemostasis, inflammation, and immunity. Int J Lab Hematol. 2013;35(3):254–61.

    Article  CAS  PubMed  Google Scholar 

  185. Diacovo TG, Puri KD, Warnock RA, Springer TA, von Andrian UH. Platelet-mediated lymphocyte delivery to high endothelial venules. Science. 1996;273(5272):252–5.

    Article  CAS  PubMed  Google Scholar 

  186. Diacovo TG, Catalina MD, Siegelman MH, von Andrian UH. Circulating activated platelets reconstitute lymphocyte homing and immunity in L-selectin-deficient mice. J Exp Med. 1998;187(2):197–204.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  187. Elzey BD, Sprague DL, Ratliff TL. The emerging role of platelets in adaptive immunity. Cell Immunol. 2005;238(1):1–9.

    Article  CAS  PubMed  Google Scholar 

  188. Li N. Platelet-lymphocyte cross-talk. J Leukoc Biol. 2008;83(5):1069–78.

    Article  CAS  PubMed  Google Scholar 

  189. McNicol A, Israels SJ. Beyond hemostasis: the role of platelets in inflammation, malignancy and infection. Cardiovasc Hematol Disord Drug Targets. 2008;8(2):99–117.

    Article  CAS  PubMed  Google Scholar 

  190. Smyth SS, McEver RP, Weyrich AS, Morrell CN, Hoffman MR, Arepally GM, et al. Platelet functions beyond hemostasis. J Thromb Haemost. 2009;7(11):1759–66.

    Article  CAS  PubMed  Google Scholar 

  191. Sowa JM, Crist SA, Ratliff TL, Elzey BD. Platelet influence on T- and B-cell responses. Arch Immunol Ther Exp (Warsz). 2009;57(4):235–41.

    Article  CAS  Google Scholar 

  192. Qu Z, Chaikof EL. Interface between hemostasis and adaptive immunity. Curr Opin Immunol. 2010;22(5):634–42.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  193. Li C, Li J, Li Y, Lang S, Yougbare I, Zhu G, et al. Crosstalk between Platelets and the Immune System: Old Systems with New Discoveries. Adv Hematol. 2012;2012:384685.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  194. Garraud O, Hamzeh-Cognasse H, Pozzetto B, Cavaillon JM, Cognasse F. Bench-to-bedside review: Platelets and active immune functions - new clues for immunopathology? Crit Care. 2013;17(4):236.

    Article  PubMed Central  PubMed  Google Scholar 

  195. Chapman LM, Aggrey AA, Field DJ, Srivastava K, Ture S, Yui K, et al. Platelets present antigen in the context of MHC class I. J Immunol. 2012;189(2):916–23.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  196. Kao KJ, Cook DJ, Scornik JC. Quantitative analysis of platelet surface HLA by W6/32 anti-HLA monoclonal antibody. Blood. 1986;68(3):627–32.

    CAS  PubMed  Google Scholar 

  197. Yukawa M, Sakon M, Kambayashi J, Shiba E, Kawasaki T, Ariyoshi H, et al. Proteasome and its novel endogeneous activator in human platelets. Biochem Biophys Res Commun. 1991;178(1):256–62.

    Article  CAS  PubMed  Google Scholar 

  198. Gupta N, Li W, Willard B, Silverstein RL, McIntyre TM. Proteasome proteolysis supports stimulated platelet function and thrombosis. Arterioscler Thromb Vasc Biol. 2014;34(1):160–8.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  199. Zufferey A, Schvartz D, Nolli S, Reny JL, Sanchez JC, Fontana P. Characterization of the platelet granule proteome: Evidence of the presence of MHC1 in alpha-granules. J Proteomics. 2014;101:130–40.

    Article  CAS  PubMed  Google Scholar 

  200. Jin R, Yu S, Song Z, Zhu X, Wang C, Yan J, et al. Soluble CD40 ligand stimulates CD40-dependent activation of the beta2 integrin Mac-1 and protein kinase C zeda (PKCzeta) in neutrophils: implications for neutrophil-platelet interactions and neutrophil oxidative burst. PLoS One. 2013;8(6):e64631.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  201. Suttles J, Stout RD. Macrophage CD40 signaling: a pivotal regulator of disease protection and pathogenesis. Semin Immunol. 2009;21(5):257–64.

    Article  CAS  PubMed  Google Scholar 

  202. Hassan GS, Mourad W. An unexpected role for MHC class II. Nat Immunol. 2011;12(5):375–6.

    Article  CAS  PubMed  Google Scholar 

  203. Jain S, Chodisetti SB, Agrewala JN. CD40 signaling synergizes with TLR-2 in the BCR independent activation of resting B cells. PLoS One. 2011;6(6):e20651.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  204. Liu X, Zhan Z, Li D, Xu L, Ma F, Zhang P, et al. Intracellular MHC class II molecules promote TLR-triggered innate immune responses by maintaining activation of the kinase Btk. Nat Immunol. 2011;12(5):416–24.

    Article  CAS  PubMed  Google Scholar 

  205. von Hundelshausen P, Weber C. Platelets as immune cells: bridging inflammation and cardiovascular disease. Circ Res. 2007;100(1):27–40.

    Article  CAS  Google Scholar 

  206. Elzey BD, Tian J, Jensen RJ, Swanson AK, Lees JR, Lentz SR, et al. Platelet-mediated modulation of adaptive immunity. A communication link between innate and adaptive immune compartments. Immunity. 2003;19(1):9–19.

    Article  CAS  PubMed  Google Scholar 

  207. Kaneider NC, Kaser A, Tilg H, Ricevuti G, Wiedermann CJ. CD40 ligand-dependent maturation of human monocyte-derived dendritic cells by activated platelets. Int J Immunopathol Pharmacol. 2003;16(3):225–31.

    CAS  PubMed  Google Scholar 

  208. 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;32(2):135–9.

    Article  CAS  PubMed  Google Scholar 

  209. Martinson J, Bae J, Klingemann HG, Tam Y. Activated platelets rapidly up-regulate CD40L expression and can effectively mature and activate autologous ex vivo differentiated DC. Cytotherapy. 2004;6(5):487–97.

    Article  CAS  PubMed  Google Scholar 

  210. Elzey BD, Grant JF, Sinn HW, Nieswandt B, Waldschmidt TJ, Ratliff TL. Cooperation between platelet-derived CD154 and CD4+ T cells for enhanced germinal center formation. J Leukoc Biol. 2005;78(1):80–4.

    Article  CAS  PubMed  Google Scholar 

  211. Solpov A, Shenkman B, Vitkovsky Y, Brill G, Koltakov A, Farzam N, et al. Platelets enhance CD4+ lymphocyte adhesion to extracellular matrix under flow conditions: role of platelet aggregation, integrins, and non-integrin receptors. Thromb Haemost. 2006;95(5):815–21.

    CAS  PubMed  Google Scholar 

  212. 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;116(3):769–74.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  213. Cognasse F, Hamzeh-Cognasse H, Lafarge S, Chavarin P, Cogne M, Richard Y, et al. Human platelets can activate peripheral blood B cells and increase production of immunoglobulins. Exp Hematol. 2007;35(9):1376–87.

    Article  CAS  PubMed  Google Scholar 

  214. Iannacone M, Sitia G, Isogawa M, Whitmire JK, Marchese P, Chisari FV, et al. Platelets prevent IFN-alpha/beta-induced lethal hemorrhage promoting CTL-dependent clearance of lymphocytic choriomeningitis virus. Proc Natl Acad Sci U S A. 2008;105(2):629–34.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  215. Elzey BD, Schmidt NW, Crist SA, Kresowik TP, Harty JT, Nieswandt B, et al. Platelet-derived CD154 enables T-cell priming and protection against Listeria monocytogenes challenge. Blood. 2008;111(7):3684–91.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  216. Nomura S, Fujita S, Nakanishi T, Yokoi T, Shimamoto K, Miyamoto R, et al. Platelet-derived microparticles cause CD154-dependent activation of dendritic cells. Platelets. 2012;23(1):81–2.

    Article  CAS  PubMed  Google Scholar 

  217. Elzey BD, Ratliff TL, Sowa JM, Crist SA. Platelet CD40L at the interface of adaptive immunity. Thromb Res. 2011;127(3):180–3.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  218. Duffau P, Seneschal J, Nicco C, Richez C, Lazaro E, Douchet I, et al. Platelet CD154 potentiates interferon-alpha secretion by plasmacytoid dendritic cells in systemic lupus erythematosus. Sci Transl Med. 2010;2(47):47ra63.

    Article  PubMed  CAS  Google Scholar 

  219. Metcalf D. Hematopoietic cytokines. Blood. 2008;111(2):485–91.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  220. Baldridge MT, King KY, Goodell MA. Inflammatory signals regulate hematopoietic stem cells. Trends Immunol. 2011;32(2):57–65.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  221. Takizawa H, Boettcher S, Manz MG. Demand-adapted regulation of early hematopoiesis in infection and inflammation. Blood. 2012;119(13):2991–3002.

    Article  CAS  PubMed  Google Scholar 

  222. Schuettpelz LG, Link DC. Regulation of hematopoietic stem cell activity by inflammation. Front Immunol. 2013;4:204.

    Article  PubMed Central  PubMed  CAS  Google Scholar 

  223. Libregts SF, Nolte MA. Parallels between immune driven-hematopoiesis and T cell activation: 3 signals that relay inflammatory stress to the bone marrow. Exp Cell Res. 2014;329(2):239–47.

    Article  CAS  PubMed  Google Scholar 

  224. Manz MG, Boettcher S. Emergency granulopoiesis. Nat Rev Immunol. 2014;14(5):302–14.

    Article  CAS  PubMed  Google Scholar 

  225. Foss B, Bruserud O, Hervig T. Platelet-released supernatants enhance hematopoietic stem cell proliferation in vitro. Platelets. 2008;19(2):155–9.

    Article  CAS  PubMed  Google Scholar 

  226. de Boer HC, van Oeveren-Rietdijk AM, Rotmans JI, Dekkers OM, Rabelink TJ, van Zonneveld AJ. Activated platelets correlate with mobilization of naive CD34(+) cells and generation of CD34(+) /KDR(+) cells in the circulation. A meta-regression analysis. J Thromb Haemost. 2013;11(8):1583–92.

    Article  PubMed  CAS  Google Scholar 

  227. Funakoshi S, Taub DD, Anver MR, Raziuddin A, Asai O, Reddy V, et al. Immunologic and hematopoietic effects of CD40 stimulation after syngeneic bone marrow transplantation in mice. J Clin Invest. 1997;99(3):484–91.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  228. Larson AW, LeBien TW. Cross-linking CD40 on human B cell precursors inhibits or enhances growth depending on the stage of development and the IL costimulus. J Immunol. 1994;153(2):584–94.

    CAS  PubMed  Google Scholar 

  229. Carlring J, Altaher HM, Clark S, Chen X, Latimer SL, Jenner T, et al. CD154-CD40 interactions in the control of murine B cell hematopoiesis. J Leukoc Biol. 2011;89(5):697–706.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  230. Seijkens T, Engel D, Tjwa M, Lutgens E. The role of CD154 in haematopoietic development. Thromb Haemost. 2010;104(4):693–701.

    Article  CAS  PubMed  Google Scholar 

  231. Solanilla A, Dechanet J, El Andaloussi A, Dupouy M, Godard F, Chabrol J, et al. CD40-ligand stimulates myelopoiesis by regulating flt3-ligand and thrombopoietin production in bone marrow stromal cells. Blood. 2000;95(12):3758–64.

    CAS  PubMed  Google Scholar 

  232. Mavroudi I, Papadaki V, Pyrovolaki K, Katonis P, Eliopoulos AG, Papadaki HA. The CD40/CD40 ligand interactions exert pleiotropic effects on bone marrow granulopoiesis. J Leukoc Biol. 2011;89(5):771–83.

    Article  CAS  PubMed  Google Scholar 

  233. Honn KV, Tang DG, Chen YQ. Platelets and cancer metastasis: more than an epiphenomenon. Semin Thromb Hemost. 1992;18(4):392–415.

    Article  CAS  PubMed  Google Scholar 

  234. Honn KV, Tang DG, Crissman JD. Platelets and cancer metastasis: a causal relationship? Cancer Metastasis Rev. 1992;11(3–4):325–51.

    Article  CAS  PubMed  Google Scholar 

  235. Nash GF, Turner LF, Scully MF, Kakkar AK. Platelets and cancer. Lancet Oncol. 2002;3(7):425–30.

    Article  CAS  PubMed  Google Scholar 

  236. Nierodzik ML, Karpatkin S. Thrombin induces tumor growth, metastasis, and angiogenesis: Evidence for a thrombin-regulated dormant tumor phenotype. Cancer Cell. 2006;10(5):355–62.

    Article  CAS  PubMed  Google Scholar 

  237. Jain S, Harris J, Ware J. Platelets: linking hemostasis and cancer. Arterioscler Thromb Vasc Biol. 2010;30(12):2362–7.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  238. Gay LJ, Felding-Habermann B. Contribution of platelets to tumour metastasis. Nat Rev Cancer. 2011;11(2):123–34.

    Article  CAS  PubMed  Google Scholar 

  239. Goubran HA, Burnouf T, Radosevic M, El-Ekiaby M. The platelet-cancer loop. Eur J Intern Med. 2013;24(5):393–400.

    Article  CAS  PubMed  Google Scholar 

  240. Menter DG, Tucker SC, Kopetz S, Sood AK, Crissman JD, Honn KV. Platelets and cancer: a casual or causal relationship: revisited. Cancer Metastasis Rev. 2014;33(1):231–69.

    Article  PubMed Central  PubMed  Google Scholar 

  241. Pinedo HM, Verheul HM, D’Amato RJ, Folkman J. Involvement of platelets in tumour angiogenesis? Lancet. 1998;352(9142):1775–7.

    Article  CAS  PubMed  Google Scholar 

  242. Sabrkhany S, Griffioen AW, Oude Egbrink MG. The role of blood platelets in tumor angiogenesis. Biochim Biophys Acta. 2011;1815(2):189–96.

    CAS  PubMed  Google Scholar 

  243. Lin WW, Karin M. A cytokine-mediated link between innate immunity, inflammation, and cancer. J Clin Invest. 2007;117(5):1175–83.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  244. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454(7203):436–44.

    Article  CAS  PubMed  Google Scholar 

  245. Gasic GJ, Gasic TB, Stewart CC. Antimetastatic effects associated with platelet reduction. Proc Natl Acad Sci U S A. 1968;61(1):46–52.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  246. Karpatkin S, Pearlstein E, Ambrogio C, Coller BS. Role of adhesive proteins in platelet tumor interaction in vitro and metastasis formation in vivo. J Clin Invest. 1988;81(4):1012–9.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  247. Borsig L, Wong R, Feramisco J, Nadeau DR, Varki NM, Varki A. Heparin and cancer revisited: mechanistic connections involving platelets, P-selectin, carcinoma mucins, and tumor metastasis. Proc Natl Acad Sci U S A. 2001;98(6):3352–7.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  248. Erpenbeck L, Schon MP. Deadly allies: the fatal interplay between platelets and metastasizing cancer cells. Blood. 2010;115(17):3427–36.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  249. Labelle M, Begum S, Hynes RO. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell. 2011;20(5):576–90.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  250. Coupland LA, Chong BH, Parish CR. Platelets and P-selectin control tumor cell metastasis in an organ-specific manner and independently of NK cells. Cancer Res. 2012;72(18):4662–71.

    Article  CAS  PubMed  Google Scholar 

  251. Schumacher D, Strilic B, Sivaraj KK, Wettschureck N, Offermanns S. Platelet-derived nucleotides promote tumor-cell transendothelial migration and metastasis via P2Y2 receptor. Cancer Cell. 2013;24(1):130–7.

    Article  CAS  PubMed  Google Scholar 

  252. Janowska-Wieczorek A, Wysoczynski M, Kijowski J, Marquez-Curtis L, Machalinski B, Ratajczak J, et al. Microvesicles derived from activated platelets induce metastasis and angiogenesis in lung cancer. Int J Cancer. 2005;113(5):752–60.

    Article  CAS  PubMed  Google Scholar 

  253. Varon D, Hayon Y, Dashevsky O, Shai E. Involvement of platelet derived microparticles in tumor metastasis and tissue regeneration. Thromb Res. 2012;130 Suppl 1:S98–9.

    Article  PubMed  Google Scholar 

  254. Tong AW, Stone MJ. Prospects for CD40-directed experimental therapy of human cancer. Cancer Gene Ther. 2003;10(1):1–13.

    Article  CAS  PubMed  Google Scholar 

  255. Vonderheide RH. Prospect of targeting the CD40 pathway for cancer therapy. Clin Cancer Res. 2007;13(4):1083–8.

    Article  CAS  PubMed  Google Scholar 

  256. Loskog AS, Eliopoulos AG. The Janus faces of CD40 in cancer. Semin Immunol. 2009;21(5):301–7.

    Article  CAS  PubMed  Google Scholar 

  257. Beatty GL, Chiorean EG, Fishman MP, Saboury B, Teitelbaum UR, Sun W, et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science. 2011;331(6024):1612–6.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  258. Korniluk A, Kemona H, Dymicka-Piekarska V. Multifunctional CD40L: pro- and anti-neoplastic activity. Tumour Biol. 2014;35(10):9447–57.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  259. Villeneuve J, Lepreux S, Mulot A, Berard AM, Higa-Nishiyama A, Costet P, et al. A protective role for CD154 in hepatic steatosis in mice. Hepatology. 2010;52(6):1968–79.

    Article  CAS  PubMed  Google Scholar 

  260. Poggi M, Engel D, Christ A, Beckers L, Wijnands E, Boon L, et al. CD40L deficiency ameliorates adipose tissue inflammation and metabolic manifestations of obesity in mice. Arterioscler Thromb Vasc Biol. 2011;31(10):2251–60.

    Article  CAS  PubMed  Google Scholar 

  261. Wolf D, Jehle F, Ortiz Rodriguez A, Dufner B, Hoppe N, Colberg C, et al. CD40L deficiency attenuates diet-induced adipose tissue inflammation by impairing immune cell accumulation and production of pathogenic IgG-antibodies. PLoS One. 2012;7(3):e33026.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  262. Guo CA, Kogan S, Amano SU, Wang M, Dagdeviren S, Friedline RH, et al. CD40 deficiency in mice exacerbates obesity-induced adipose tissue inflammation, hepatic steatosis, and insulin resistance. Am J Physiol Endocrinol Metab. 2013;304(9):E951–63.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  263. Wolf D, Jehle F, Michel NA, Bukosza EN, Rivera J, Chen YC, et al. Coinhibitory suppression of T cell activation by CD40 protects against obesity and adipose tissue inflammation in mice. Circulation. 2014;129(23):2414–25.

    Article  CAS  PubMed  Google Scholar 

  264. Franchini M, Mannucci PM. Thrombogenicity and cardiovascular effects of ambient air pollution. Blood. 2011;118(9):2405–12.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

A.T. acknowledges support from the Amadeus LabEx, Université de Bordeaux. J.V. acknowledges support from a Marie Curie international outgoing fellowship within the 7th European community framework program. The support of the Association pour la Recherche en Néphrologie is acknowledged.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jean Ripoche.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

All authors contributed to the writing of the manuscript. All authors read and approved the manuscript.

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dewitte, A., Tanga, A., Villeneuve, J. et al. New frontiers for platelet CD154. Exp Hematol Oncol 4, 6 (2015). https://doi.org/10.1186/s40164-015-0001-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40164-015-0001-6

Keywords