51
|
Toll like receptor 2/1 mediated platelet adhesion and activation on bacterial mimetic surfaces is dependent on src/Syk-signaling and purinergic receptor P2X1 and P2Y12 activation. Biointerphases 2015; 9:041003. [PMID: 25553878 DOI: 10.1116/1.4901135] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Platelets are considered to have important functions in inflammatory processes as key players in innate immunity. Toll like receptors (TLRs), expressed on platelets, recognize pathogen associated molecular patterns and trigger immune responses. Pathogens are able to adhere to human tissues and form biofilms which cause a continuous activation of the immune system. The authors aimed to investigate how immobilized Pam3CSK4 (a synthetic TLR2/1 agonist) and IgG, respectively, resembling a bacterial focus, affects adhesion and activation of platelets including release of two cytokines, regulated on activation normal T-cell expressed and secreted (RANTES) and macrophage migration inhibitory factor (MIF). The authors also aim to clarify the signaling downstream of TLR2/1 and FcγRII (IgG receptor) and the role of adenine nucleotides in this process. Biolayers of Pam3CSK4 and IgG, respectively, were confirmed by null-ellipsometry and contact angle measurements. Platelets were preincubated with signaling inhibitors for scr and Syk and antagonists for P2X1 or P2Y1 [adenosine triphosphate (ATP), adenosine diphosphate (ADP) receptors] prior to addition to the surfaces. The authors show that platelets adhere and spread on both Pam3CSK4- and IgG-coated surfaces and that this process is antagonized by scr and Syc inhibitors as well as P2X1 and P2Y antagonists. This suggests that Pam3CSK4 activated platelets utilize the same pathway as FcγRII. Moreover, the authors show that ATP-ligation of P2X1 is of importance for further platelet activation after TLR2/1-activation, and that P2Y12 is the prominent ADP-receptor involved in adhesion and spreading. RANTES and MIF were secreted over time from platelets adhering to the coated surfaces, but no MIF was released upon stimulation with soluble Pam3CSK4. These results clarify the importance of TLR2/1 and FcγRII in platelet adhesion and activation, and strengthen the role of platelets as an active player in sensing bacterial infections.
Collapse
|
52
|
Platelet Activation and Thrombus Formation over IgG Immune Complexes Requires Integrin αIIbβ3 and Lyn Kinase. PLoS One 2015; 10:e0135738. [PMID: 26291522 PMCID: PMC4546160 DOI: 10.1371/journal.pone.0135738] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 07/25/2015] [Indexed: 12/15/2022] Open
Abstract
IgG immune complexes contribute to the etiology and pathogenesis of numerous autoimmune disorders, including heparin-induced thrombocytopenia, systemic lupus erythematosus, rheumatoid- and collagen-induced arthritis, and chronic glomerulonephritis. Patients suffering from immune complex-related disorders are known to be susceptible to platelet-mediated thrombotic events. Though the role of the Fc receptor, FcγRIIa, in initiating platelet activation is well understood, the role of the major platelet adhesion receptor, integrin αIIbβ3, in amplifying platelet activation and mediating adhesion and aggregation downstream of encountering IgG immune complexes is poorly understood. The goal of this investigation was to gain a better understanding of the relative roles of these two receptor systems in immune complex-mediated thrombotic complications. Human platelets, and mouse platelets genetically engineered to differentially express FcγRIIa and αIIbβ3, were allowed to interact with IgG-coated surfaces under both static and flow conditions, and their ability to spread and form thrombi evaluated in the presence and absence of clinically-used fibrinogen receptor antagonists. Although binding of IgG immune complexes to FcγRIIa was sufficient for platelet adhesion and initial signal transduction events, platelet spreading and thrombus formation over IgG-coated surfaces showed an absolute requirement for αIIbβ3 and its ligands. Tyrosine kinases Lyn and Syk were found to play key roles in IgG-induced platelet activation events. Taken together, our data suggest a complex functional interplay between FcγRIIa, Lyn, and αIIbβ3 in immune complex-induced platelet activation. Future studies may be warranted to determine whether patients suffering from immune complex disorders might benefit from treatment with anti-αIIbβ3-directed therapeutics.
Collapse
|
53
|
Arman M, Krauel K. Human platelet IgG Fc receptor FcγRIIA in immunity and thrombosis. J Thromb Haemost 2015; 13:893-908. [PMID: 25900780 DOI: 10.1111/jth.12905] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/10/2015] [Indexed: 01/23/2023]
Abstract
Beyond their prominent role in hemostasis and thrombosis, platelets are increasingly recognized as having immunologic functions. Supporting this, human platelets express FcγRIIA (CD32a), a low-affinity Fc receptor (FcR) for the constant region of IgG that recognizes immune complexes (ICs) and IgG-opsonized cells with high avidity. In leukocytes, FcγRIIA engagement initiates strong effector functions that are key for immune and inflammatory responses, including cytokine release, antibody-dependent cell-mediated killing of pathogens, and internalization of ICs. However, the physiologic relevance of platelet-expressed FcγRIIA has received little attention in previous reviews on FcRs. This article summarizes and discusses the available information on human platelet FcγRIIA. The importance of this receptor in heparin-induced thrombocytopenia, a prothrombotic adverse drug effect, is well documented. However, studies demonstrating platelet activation by IgG-opsonized bacteria point to the physiologic relevance of platelet FcγRIIA in immunity. In this context, platelet activation and secretion may facilitate both a direct antimicrobial function of platelets and crosstalk with other immune cells. Additionally, a role for platelet FcγRIIA in IgG-independent hemostasis and physiologic thrombosis, by means of amplifying integrin αII b β3 outside-in signaling, has also been proposed. Nonetheless, the thrombotic complications found in some infective and autoimmune diseases may result from unbalanced FcγRIIA-mediated platelet aggregation. Moreover, FcγRIIA is not expressed in mice, and thrombocytopenia and/or thrombotic events found after drug administration can only be recapitulated by the use of human FcγRIIA-transgenic mice. Altogether, the available data support a functional role for platelet FcγRIIA in health and disease, and emphasize the need for further investigation of this receptor.
Collapse
Affiliation(s)
- M Arman
- Centre for Cardiovascular Sciences, Institute of Biomedical Research, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - K Krauel
- Institut für Immunologie und Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
54
|
Abstract
Multiple studies have now shown that various species of bacteria can stimulate platelets; many in a strain and donor-dependent manner. The signalling pathways underlying this platelet activation has been the subject of scrutiny for the last decade. The best-delineated pathway is that in response to Streptococcal species, such as Streptococcus sanguinis (S. sanguinis), Streptococcus gordonii (S. gordonii) and Streptococcus oralis (S. oralis), where a pathway is initiated by the engagement of the low affinity IgG receptor, FcγRIIA. This leads to and involves the tyrosine kinase Syk, the adaptor protein Linker of Activated T Cells (LAT) and subsequently both phospholipase Cγ2 (PLCγ2) and phosphatidylinositol-3-kinase (PI-3-K). Finally, this leads to the expression of the αIIbβ3 integrin, the synthesis and release of thromboxane A2 (T × A2) and the exocytosis of PF4, each of which plays a crucial role in secondary signalling and full platelet activation. Roles for other signalling pathways in Streptococcal-induced platelet activation are less clear, although an ADP-mediated inhibition of adenylyl cyclase, a glycoprotein Ib/IX/V-mediated pathway and perhaps a complement-induced pathway have each been proposed. Platelet activation by Porphyromonas gingivalis (P. gingivalis) at least partially shares the FcγRIIA/Syk/PLCγ2/PI-3-K mechanism utilised by Streptococcal species. However, it has also been suggested that P. gingivalis activates platelets by two additional methods; stimulation of the protease-activated receptors leading to activation of phospholipase Cβ (PLCβ), and the engagement of Toll-like receptors 2 and 4 by released lipopolysaccharide leading to an ill-defined pathway which may involve PI-3-K. Consequently, it appears that bacteria can stimulate platelets by eliciting multiple signalling pathways some of which are common, and some unique, to individual species.
Collapse
|
55
|
Restoration of responsiveness of phospholipase Cγ2-deficient platelets by enforced expression of phospholipase Cγ1. PLoS One 2015; 10:e0119739. [PMID: 25793864 PMCID: PMC4368822 DOI: 10.1371/journal.pone.0119739] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 01/15/2015] [Indexed: 01/21/2023] Open
Abstract
Receptor-mediated platelet activation requires phospholipase C (PLC) activity to elevate intracellular calcium and induce actin cytoskeleton reorganization. PLCs are classified into structurally distinct β, γ, δ, ε, ζ, and η isoforms. There are two PLCγ isoforms (PLCγ1, PLCγ2), which are critical for activation by tyrosine kinase-dependent receptors. Platelets express both PLCγ1 and PLCγ2. Although PLCγ2 has been shown to play a dominant role in platelet activation, the extent to which PLCγ1 contributes has not been evaluated. To ascertain the relative contributions of PLCγ1 and PLCγ2 to platelet activation, we generated conditionally PLCγ1-deficient, wild-type (WT), PLCγ2-deficient, and PLCγ1/PLCγ2 double-deficient mice and measured the ability of platelets to respond to different agonists. We found that PLCγ2 deficiency abrogated αIIbβ3-dependent platelet spreading, GPVI-dependent platelet aggregation, and thrombus formation on collagen-coated surfaces under shear conditions, which is dependent on both GPVI and αIIbβ3. Addition of exogenous ADP overcame defective spreading of PLCγ2-deficient platelets on immobilized fibrinogen, suggesting that PLCγ2 is required for granule secretion in response to αIIbβ3 ligation. Consistently, αIIbβ3-mediated release of granule contents was impaired in the absence of PLCγ2. In contrast, PLCγ1-deficient platelets spread and released granule contents normally on fibrinogen, exhibited normal levels of GPVI-dependent aggregation, and formed thrombi normally on collagen-coated surfaces. Interestingly, enforced expression of PLCγ1 fully restored GPVI-dependent aggregation and αIIbβ3-dependent spreading of PLCγ2-deficient platelets. We conclude that platelet activation through GPVI and αIIbβ3 utilizes PLCγ2 because PLCγ1 levels are insufficient to support responsiveness, but that PLCγ1 can restore responsiveness if expressed at levels normally achieved by PLCγ2.
Collapse
|
56
|
Abstract
PURPOSE OF REVIEW To review the recent developments in understanding the pathophysiology of heparin-induced thrombocytopenia (HIT) and in applying this knowledge to the treatment of patients with suspected and proven HIT. RECENT FINDINGS HIT pathophysiology is dynamic and complex. HIT pathophysiology is initiated by four essential components--heparin (Hep), platelet factor 4 (PF4), IgG antibodies against the Hep-PF4 complex, and platelet FcγRIIa. HIT is propagated by activated platelets, monocytes, endothelial cells, and coagulation proteins. Insights into the unique HIT antibody response continue to emerge, but without consensus as to the relative roles of B cells, T cells, and antigen-presenting cells. Platelet activation via FcγRIIa, the sine qua non of HIT, has become much better appreciated. Therapy remains challenging for several reasons. Suspected HIT is more frequent than proven HIT, because of the widespread use of Hep and the inadequacies of current diagnostic tests and scoring systems. In proven HIT, approved treatments reduce but do not eliminate thrombosis, and have substantial bleeding risk. Rational novel therapeutic strategies, directed at the initiating steps in HIT pathophysiology and with potential combinations staged over time, are in various phases of development. SUMMARY Progress continues in understanding the breadth of molecular and cellular players in HIT. Translation to improved diagnosis and treatment is needed.
Collapse
|
57
|
Moroi AJ, Watson SP. Impact of the PI3-kinase/Akt pathway on ITAM and hemITAM receptors: haemostasis, platelet activation and antithrombotic therapy. Biochem Pharmacol 2015; 94:186-94. [PMID: 25698506 DOI: 10.1016/j.bcp.2015.02.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 02/09/2015] [Accepted: 02/09/2015] [Indexed: 01/16/2023]
Abstract
Phosphoinositide 3-kinases (PI3Ks) are a family of lipid kinases that are activated in response to various stimulants, and they regulate many processes including inflammation; the stress response; gene transcription; and cell proliferation, differentiation, and death. Increasing reports have shown that the PI3Ks and their downstream effector Akt are activated by several platelet receptors that regulate platelet activation and haemostasis. Platelets express two immunoreceptor tyrosine based activation motif (ITAM) receptors, collagen receptor glycoprotein VI (GPVI) and Fcγ receptor IIA (FcγRIIA), which are characterized by two YxxL sequences separated by 6-12 amino acids. Activation of an ITAM receptor initiates a reaction cascade via its YxxL sequence in which signaling molecules such as spleen tyrosine kinase (Syk), linker for activation of T cells (LAT) and phospholipase C γ2 (PLCγ2) become activated, leading to platelet activation. Platelets also express another receptor, C-type lectin 2 (CLEC-2), which has a single YxxL sequence, so it is appropriately called a hemITAM receptor. ITAM receptors and the hemITAM receptor share many signaling features. Here we will summarize our current knowledge about how the PI3K/Akt pathway regulates (hem)ITAM receptor-mediated platelet activation and haemostasis and discuss the possible benefits of targeting PI3K/Akt as an antithrombotic therapy.
Collapse
Affiliation(s)
- Alyssa J Moroi
- Centre for Cardiovascular Sciences, Institute for Biomedical Research, The College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom.
| | - Steve P Watson
- Centre for Cardiovascular Sciences, Institute for Biomedical Research, The College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
58
|
Increased risk of thrombosis in FcγRIIA 131RR patients with HIT due to defective control of platelet activation by plasma IgG2. Blood 2015; 125:2397-404. [PMID: 25680756 DOI: 10.1182/blood-2014-09-594515] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 02/04/2015] [Indexed: 01/14/2023] Open
Abstract
Thrombosis results in heparin-induced thrombocytopenia (HIT) from cellular activation involving Fc receptors. In this study, the FcγRIIA 131RR genotype was found to increase the risk of thrombosis in HIT patients (odds ratio: 5.9; 95% confidence interval: 1.7-20). When platelet aggregation tests (PATs) were performed with platelet-rich plasma (PRP), a shorter lag time was measured in 131RR donors compared to individuals with the HR and HH genotypes in response to HIT plasma or 5B9, a recently developed humanized monoclonal antibody to PF4/heparin. Importantly, this difference was no longer detectable when PATs were performed with washed platelets or immunoglobulin (Ig)G-depleted PRP. Moreover, polyclonal IgG or monoclonal IgG1 added to IgG-depleted PRP increased the lag time in response to 5B9. HH platelets were also sensitive to IgG2, which in contrast, failed to inhibit the response of 131RR platelets to 5B9. Finally, higher tissue factor messenger RNA levels were measured in the whole blood of 131RR donors after activation by HIT antibodies, with increased phospholipid procoagulant activity. These results demonstrate that HIT patients homozygous for the FcγRIIA 131R allele have a higher risk of thrombosis, probably due to increased cell activation by antibodies to PF4/heparin, with a lower inhibitory effect of endogenous IgG, especially from the IgG2 subclass.
Collapse
|
59
|
Hao HZ, He AD, Wang DC, Yin Z, Zhou YJ, Liu G, Liang ML, Da XW, Yao GQ, Xie W, Xiang JZ, Ming ZY. Antiplatelet activity of loureirin A by attenuating Akt phosphorylation: In vitro studies. Eur J Pharmacol 2015; 746:63-9. [PMID: 25445049 DOI: 10.1016/j.ejphar.2014.10.059] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 10/31/2014] [Accepted: 10/31/2014] [Indexed: 02/05/2023]
Abstract
Loureirin A is a flavonoid extracted from Dragon׳s Blood that has been used to promote blood circulation and remove stasis in Chinese traditional medicine. However, the mechanisms of these effects are not fully understood. We explored the anti-platelet activity and underlying mechanism of loureirin A in vitro. Our results indicated that loureirin A negatively affected agonist-induced platelet aggregation such as collagen, collagen-related peptide (CRP), ADP and thrombin. Loureirin A inhibited collagen-induced platelet ATP secretion and thrombin-stimulated P-selectin expression in a dose-dependent manner. Platelet spreading on immobilized fibrinogen was significantly impaired in the presence of loureirin A. Immunoblotting analysis indicated that 100μM of loureirin A almost completely eliminated collagen-induced Akt phosphorylation at Ser473. Interestingly, a submaximal dose (50μM) of loureirin A had an additive inhibitory effect with the phosphoinositide 3-kinase (PI3K) inhibitor Ly294002 on collage-induced Akt phosphorylation in platelets. Taken together, loureirin A had an inhibitory effect on platelet activation, perhaps through an impairment of PI3K/Akt signaling.
Collapse
Affiliation(s)
- Hong-Zhen Hao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China; Department of Pharmacy, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Ao-Di He
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - Dao-Chun Wang
- Department of Traditional Chinese Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhao Yin
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - Ya-Jun Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - Gang Liu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - Ming-Lu Liang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - Xing-Wen Da
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - Guang-Qiang Yao
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - Wen Xie
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - Ji-Zhou Xiang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China
| | - Zhang-Yin Ming
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China; The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan 430030, China.
| |
Collapse
|
60
|
Uppal H, Doudement E, Mahapatra K, Darbonne WC, Bumbaca D, Shen BQ, Du X, Saad O, Bowles K, Olsen S, Lewis Phillips GD, Hartley D, Sliwkowski MX, Girish S, Dambach D, Ramakrishnan V. Potential mechanisms for thrombocytopenia development with trastuzumab emtansine (T-DM1). Clin Cancer Res 2014; 21:123-33. [PMID: 25370470 DOI: 10.1158/1078-0432.ccr-14-2093] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Trastuzumab-emtansine (T-DM1) is an antibody-drug conjugate (ADC) comprising the cytotoxic agent DM1 conjugated to trastuzumab with a stable linker. Thrombocytopenia was the dose-limiting toxicity in the phase I study, and grade ≥3 thrombocytopenia occurred in up to 13% of patients receiving T-DM1 in phase III studies. We investigated the mechanism of T-DM1-induced thrombocytopenia. EXPERIMENTAL DESIGN The effect of T-DM1 on platelet function was measured by aggregometry, and by flow cytometry to detect the markers of activation. The effect of T-DM1 on differentiation and maturation of megakaryocytes (MK) from human hematopoietic stem cells was assessed by flow cytometry and microscopy. Binding, uptake, and catabolism of T-DM1 in MKs, were assessed by various techniques including fluorescence microscopy, scintigraphy to detect T-[H(3)]-DM1 and (125)I-T-DM1, and mass spectrometry. The role of FcγRIIa was assessed using blocking antibodies and mutant constructs of trastuzumab that do not bind FcγR. RESULTS T-DM1 had no direct effect on platelet activation and aggregation, but it did markedly inhibit MK differentiation via a cytotoxic effect. Inhibition occurred with DM1-containing ADCs but not with trastuzumab demonstrating a role for DM1. MKs internalized these ADCs in a HER2-independent, FcγRIIa-dependent manner, resulting in intracellular release of DM1. Binding and internalization of T-DM1 diminished as MKs matured; however, prolonged exposure of mature MKs to T-DM1 resulted in a disrupted cytoskeletal structure. CONCLUSIONS These data support the hypothesis that T-DM1-induced thrombocytopenia is mediated in large part by DM1-induced impairment of MK differentiation, with a less pronounced effect on mature MKs.
Collapse
Affiliation(s)
- Hirdesh Uppal
- Department of Safety Assessment, Genentech, Inc, South San Francisco, California
| | - Estelle Doudement
- Department of Safety Assessment, Genentech, Inc, South San Francisco, California
| | - Kaushiki Mahapatra
- Department of Safety Assessment, Genentech, Inc, South San Francisco, California
| | - Walter C Darbonne
- Department of Oncology Biomarker Development, Development Sciences, gRED, Genentech, Inc, South San Francisco, California
| | - Daniela Bumbaca
- Department of Preclinical and Translational Pharmacokinetics and Pharmodynamics, Genentech, Inc, South San Francisco, California
| | - Ben-Quan Shen
- Department of Preclinical and Translational Pharmacokinetics and Pharmodynamics, Genentech, Inc, South San Francisco, California
| | - Xiaoyan Du
- Department of Oncology Biomarker Development, Development Sciences, gRED, Genentech, Inc, South San Francisco, California
| | - Ola Saad
- Department of Bioanalytical Sciences, Genentech, Inc, South San Francisco, California
| | - Kristin Bowles
- Department of Protein Chemistry, Genentech, Inc, South San Francisco, California
| | - Steve Olsen
- Department of Product Development, Genentech, Inc, South San Francisco, California
| | | | - Dylan Hartley
- Department of Safety Assessment, Genentech, Inc, South San Francisco, California
| | - Mark X Sliwkowski
- Department of Molecular Oncology, Genentech, Inc, South San Francisco, California
| | - Sandhya Girish
- Department of Development Sciences, Genentech, Inc, South San Francisco, California
| | - Donna Dambach
- Department of Small Molecule and Investigative Toxicology, Genentech, Inc, South San Francisco, California
| | - Vanitha Ramakrishnan
- Department of Project Management and Operations, Genentech, Inc, South San Francisco, California.
| |
Collapse
|
61
|
The N-terminal SH2 domain of Syk is required for (hem)ITAM, but not integrin, signaling in mouse platelets. Blood 2014; 125:144-54. [PMID: 25352128 DOI: 10.1182/blood-2014-05-579375] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We have used a novel knockin mouse to investigate the effect of disruption of phosphotyrosine binding of the N-terminal SH2 domain of Syk on platelet activation by GPVI, CLEC-2, and integrin αIIbβ3. The Syk(R41Afl/fl) mouse was crossed to a PF4-Cre(+) mouse to induce expression of the Syk mutant in the megakaryocyte/platelet lineage. Syk(R41Afl/fl;PF4-Cre) mice are born at approximately 50% of the expected frequency and have a similar phenotype to Syk(fl/fl;PF4-Cre) mice, including blood-lymphatic mixing and chyloascites. Anastomosis of the venous and lymphatic vasculatures can be seen in the mesenteric circulation accounting for rapid and continuous mixing of the 2 vasculatures. Platelet activation by CLEC-2 and GPVI is abolished in Syk(R41Afl/fl;PF4-Cre) platelets. Syk phosphorylation on Tyr519/20 is blocked in CLEC-2-stimulated platelets, suggesting a model in which binding of Syk via its N-terminal SH2 domain regulates autophosphorylation. In contrast, outside-in signaling by integrin αIIbβ3 is not altered, but it is inhibited in the presence of inhibitors of Src and Syk tyrosine kinases. These results demonstrate that αIIbβ3 regulates Syk through an ITAM-independent pathway in mice and provide novel insight into the course of events underlying Syk activation and hemITAM phosphorylation by CLEC-2.
Collapse
|
62
|
Abstract
Platelets are essential in maintaining hemostasis following inflammation or injury to the vasculature. Dysregulated platelet activity often results in thrombotic complications leading to myocardial infarction and stroke. Activation of the FcγRIIa receptor leads to immune-mediated thrombosis, which is often life threatening in patients undergoing heparin-induced thrombocytopenia or sepsis. Inhibiting FcγRIIa-mediated activation in platelets has been shown to limit thrombosis and is the principal target for prevention of immune-mediated platelet activation. In this study, we show for the first time that platelet 12(S)-lipoxygenase (12-LOX), a highly expressed oxylipin-producing enzyme in the human platelet, is an essential component of FcγRIIa-mediated thrombosis. Pharmacologic inhibition of 12-LOX in human platelets resulted in significant attenuation of FcγRIIa-mediated aggregation. Platelet 12-LOX was shown to be essential for FcγRIIa-induced phospholipase Cγ2 activity leading to activation of calcium mobilization, Rap1 and protein kinase C activation, and subsequent activation of the integrin αIIbβ3. Additionally, platelets from transgenic mice expressing human FcγRIIa but deficient in platelet 12-LOX, failed to form normal platelet aggregates and exhibited deficiencies in Rap1 and αIIbβ3 activation. These results support an essential role for 12-LOX in regulating FcγRIIa-mediated platelet function and identifies 12-LOX as a potential therapeutic target to limit immune-mediated thrombosis.
Collapse
|
63
|
Xu Z, Chen X, Zhi H, Gao J, Bialkowska K, Byzova TV, Pluskota E, White GC, Liu J, Plow EF, Ma YQ. Direct interaction of kindlin-3 with integrin αIIbβ3 in platelets is required for supporting arterial thrombosis in mice. Arterioscler Thromb Vasc Biol 2014; 34:1961-7. [PMID: 24969775 DOI: 10.1161/atvbaha.114.303851] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Kindlin-3 is a critical supporter of integrin function in platelets. Lack of expression of kindlin-3 protein in patients impairs integrin αIIbβ3-mediated platelet aggregation. Although kindlin-3 has been categorized as an integrin-binding partner, the functional significance of the direct interaction of kindlin-3 with integrin αIIbβ3 in platelets has not been established. Here, we evaluated the significance of the binding of kindlin-3 to integrin αIIbβ3 in platelets in supporting integrin αIIbβ3-mediated platelet functions. APPROACH AND RESULTS We generated a strain of kindlin-3 knockin (K3KI) mice that express a kindlin-3 mutant that carries an integrin-interaction defective substitution. K3KI mice could survive normally and express integrin αIIbβ3 on platelets similar to their wild-type counterparts. Functional analysis revealed that K3KI mice exhibited defective platelet function, including impaired integrin αIIbβ3 activation, suppressed platelet spreading and platelet aggregation, prolonged tail bleeding time, and absence of platelet-mediated clot retraction. In addition, whole blood drawn from K3KI mice showed resistance to in vitro thrombus formation and, as a consequence, K3KI mice were protected from in vivo arterial thrombosis. CONCLUSIONS These observations demonstrate that the direct binding of kindlin-3 to integrin αIIbβ3 is involved in supporting integrin αIIbβ3 activation and integrin αIIbβ3-dependent responses of platelets and consequently contributes significantly to arterial thrombus formation.
Collapse
Affiliation(s)
- Zhen Xu
- From the Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China (Z.X., J.G., E.F.P., Y.-Q.M.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (Z.X., H.Z., G.C.W., Y.-Q.M.); Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao-Tong University School of Medicine, Shanghai, China (X.C., J.L.); and Department of Molecular Cardiology, Cleveland Clinic, OH (K.B., T.V.B., E.P., E.F.P.)
| | - Xue Chen
- From the Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China (Z.X., J.G., E.F.P., Y.-Q.M.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (Z.X., H.Z., G.C.W., Y.-Q.M.); Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao-Tong University School of Medicine, Shanghai, China (X.C., J.L.); and Department of Molecular Cardiology, Cleveland Clinic, OH (K.B., T.V.B., E.P., E.F.P.)
| | - Huiying Zhi
- From the Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China (Z.X., J.G., E.F.P., Y.-Q.M.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (Z.X., H.Z., G.C.W., Y.-Q.M.); Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao-Tong University School of Medicine, Shanghai, China (X.C., J.L.); and Department of Molecular Cardiology, Cleveland Clinic, OH (K.B., T.V.B., E.P., E.F.P.)
| | - Juan Gao
- From the Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China (Z.X., J.G., E.F.P., Y.-Q.M.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (Z.X., H.Z., G.C.W., Y.-Q.M.); Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao-Tong University School of Medicine, Shanghai, China (X.C., J.L.); and Department of Molecular Cardiology, Cleveland Clinic, OH (K.B., T.V.B., E.P., E.F.P.)
| | - Katarzyna Bialkowska
- From the Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China (Z.X., J.G., E.F.P., Y.-Q.M.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (Z.X., H.Z., G.C.W., Y.-Q.M.); Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao-Tong University School of Medicine, Shanghai, China (X.C., J.L.); and Department of Molecular Cardiology, Cleveland Clinic, OH (K.B., T.V.B., E.P., E.F.P.)
| | - Tatiana V Byzova
- From the Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China (Z.X., J.G., E.F.P., Y.-Q.M.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (Z.X., H.Z., G.C.W., Y.-Q.M.); Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao-Tong University School of Medicine, Shanghai, China (X.C., J.L.); and Department of Molecular Cardiology, Cleveland Clinic, OH (K.B., T.V.B., E.P., E.F.P.)
| | - Elzbieta Pluskota
- From the Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China (Z.X., J.G., E.F.P., Y.-Q.M.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (Z.X., H.Z., G.C.W., Y.-Q.M.); Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao-Tong University School of Medicine, Shanghai, China (X.C., J.L.); and Department of Molecular Cardiology, Cleveland Clinic, OH (K.B., T.V.B., E.P., E.F.P.)
| | - Gilbert C White
- From the Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China (Z.X., J.G., E.F.P., Y.-Q.M.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (Z.X., H.Z., G.C.W., Y.-Q.M.); Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao-Tong University School of Medicine, Shanghai, China (X.C., J.L.); and Department of Molecular Cardiology, Cleveland Clinic, OH (K.B., T.V.B., E.P., E.F.P.)
| | - Junling Liu
- From the Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China (Z.X., J.G., E.F.P., Y.-Q.M.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (Z.X., H.Z., G.C.W., Y.-Q.M.); Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao-Tong University School of Medicine, Shanghai, China (X.C., J.L.); and Department of Molecular Cardiology, Cleveland Clinic, OH (K.B., T.V.B., E.P., E.F.P.)
| | - Edward F Plow
- From the Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China (Z.X., J.G., E.F.P., Y.-Q.M.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (Z.X., H.Z., G.C.W., Y.-Q.M.); Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao-Tong University School of Medicine, Shanghai, China (X.C., J.L.); and Department of Molecular Cardiology, Cleveland Clinic, OH (K.B., T.V.B., E.P., E.F.P.)
| | - Yan-Qing Ma
- From the Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China (Z.X., J.G., E.F.P., Y.-Q.M.); Blood Research Institute, Blood Center of Wisconsin, Milwaukee (Z.X., H.Z., G.C.W., Y.-Q.M.); Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao-Tong University School of Medicine, Shanghai, China (X.C., J.L.); and Department of Molecular Cardiology, Cleveland Clinic, OH (K.B., T.V.B., E.P., E.F.P.).
| |
Collapse
|
64
|
Stegner D, Haining EJ, Nieswandt B. Targeting glycoprotein VI and the immunoreceptor tyrosine-based activation motif signaling pathway. Arterioscler Thromb Vasc Biol 2014; 34:1615-20. [PMID: 24925975 DOI: 10.1161/atvbaha.114.303408] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Coronary artery thrombosis and ischemic stroke are often initiated by the disruption of an atherosclerotic plaque and consequent intravascular platelet activation. Thus, antiplatelet drugs are central in the treatment and prevention of the initial, and subsequent, vascular events. However, novel pharmacological targets for platelet inhibition remain an important goal of cardiovascular research because of the negative effect of existing antiplatelet drugs on primary hemostasis. One promising target is the platelet collagen receptor glycoprotein VI. Blockade or antibody-mediated depletion of this receptor in circulating platelets is beneficial in experimental models of thrombosis and thrombo-inflammatory diseases, such as stroke, without impairing hemostasis. In this review, we summarize the importance of glycoprotein VI and (hem)immunoreceptor tyrosine-based activation motif signaling in hemostasis, thrombosis, and thrombo-inflammatory processes and discuss the targeting strategies currently under development for inhibiting glycoprotein VI and its signaling.
Collapse
Affiliation(s)
- David Stegner
- From the Department of Experimental Biomedicine, University Hospital Würzburg and Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Elizabeth J Haining
- From the Department of Experimental Biomedicine, University Hospital Würzburg and Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Bernhard Nieswandt
- From the Department of Experimental Biomedicine, University Hospital Würzburg and Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
65
|
Gillis C, Gouel-Chéron A, Jönsson F, Bruhns P. Contribution of Human FcγRs to Disease with Evidence from Human Polymorphisms and Transgenic Animal Studies. Front Immunol 2014; 5:254. [PMID: 24910634 PMCID: PMC4038777 DOI: 10.3389/fimmu.2014.00254] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 05/14/2014] [Indexed: 11/13/2022] Open
Abstract
The biological activities of human IgG antibodies predominantly rely on a family of receptors for the Fc portion of IgG, FcγRs: FcγRI, FcγRIIA, FcγRIIB, FcγRIIC, FcγRIIIA, FcγRIIIB, FcRL5, FcRn, and TRIM21. All FcγRs bind IgG at the cell surface, except FcRn and TRIM21 that bind IgG once internalized. The affinity of FcγRs for IgG is determined by polymorphisms of human FcγRs and ranges from 2 × 104 to 8 × 107 M−1. The biological functions of FcγRs extend from cellular activation or inhibition, IgG-internalization/endocytosis/phagocytosis to IgG transport and recycling. This review focuses on human FcγRs and intends to present an overview of the current understanding of how these receptors may contribute to various pathologies. It will define FcγRs and their polymorphic variants, their affinity for human IgG subclasses, and review the associations found between FcγR polymorphisms and human pathologies. It will also describe the human FcγR-transgenic mice that have been used to study the role of these receptors in autoimmune, inflammatory, and allergic disease models.
Collapse
Affiliation(s)
- Caitlin Gillis
- Laboratoire Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur , Paris , France ; U760, INSERM , Paris , France
| | - Aurélie Gouel-Chéron
- Laboratoire Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur , Paris , France ; U760, INSERM , Paris , France ; Department of Anesthesia and Intensive Care, Hospital of Bichat-Claude Bernard, Public Assistance-Hospitals of Paris , Paris , France
| | - Friederike Jönsson
- Laboratoire Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur , Paris , France ; U760, INSERM , Paris , France
| | - Pierre Bruhns
- Laboratoire Anticorps en Thérapie et Pathologie, Département d'Immunologie, Institut Pasteur , Paris , France ; U760, INSERM , Paris , France
| |
Collapse
|
66
|
Boulaftali Y, Hess PR, Kahn ML, Bergmeier W. Platelet immunoreceptor tyrosine-based activation motif (ITAM) signaling and vascular integrity. Circ Res 2014; 114:1174-84. [PMID: 24677237 PMCID: PMC4000726 DOI: 10.1161/circresaha.114.301611] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 02/18/2014] [Indexed: 01/27/2023]
Abstract
Platelets are well-known for their critical role in hemostasis, that is, the prevention of blood loss at sites of mechanical vessel injury. Inappropriate platelet activation and adhesion, however, can lead to thrombotic complications, such as myocardial infarction and stroke. To fulfill its role in hemostasis, the platelet is equipped with various G protein-coupled receptors that mediate the response to soluble agonists such as thrombin, ADP, and thromboxane A2. In addition to G protein-coupled receptors, platelets express 3 glycoproteins that belong to the family of immunoreceptor tyrosine-based activation motif receptors: Fc receptor γ chain, which is noncovalently associated with the glycoprotein VI collagen receptor, C-type lectin 2, the receptor for podoplanin, and Fc receptor γII A, a low-affinity receptor for immune complexes. Although both genetic and chemical approaches have documented a critical role for platelet G protein-coupled receptors in hemostasis, the contribution of immunoreceptor tyrosine-based activation motif receptors to this process is less defined. Studies performed during the past decade, however, have identified new roles for platelet immunoreceptor tyrosine-based activation motif signaling in vascular integrity in utero and at sites of inflammation. The purpose of this review is to summarize recent findings on how platelet immunoreceptor tyrosine-based activation motif signaling controls vascular integrity, both in the presence and absence of mechanical injury.
Collapse
Affiliation(s)
- Yacine Boulaftali
- From the McAllister Heart Institute (Y.B., W.B.) and Department of Biochemistry and Biophysics (W.B.), University of North Carolina, Chapel Hill; and Department of Medicine and Division of Cardiology, University of Pennsylvania, Philadelphia (P.R.H., M.L.K.)
| | | | | | | |
Collapse
|
67
|
Abstract
Platelets play crucial functions in hemostasis and the prevention of bleeding. During H1N1 influenza A virus infection, platelets display activation markers. The platelet activation triggers during H1N1 infection remain elusive. We observed that H1N1 induces surface receptor activation, lipid mediator synthesis, and release of microparticles from platelets. These activation processes require the presence of serum/plasma, pointing to the contribution of soluble factor(s). Considering that immune complexes in the H1N1 pandemic were reported to play a pathogenic role, we assessed their contribution in H1N1-induced platelet activation. In influenza-immunized subjects, we observed that the virus scaffolds with immunoglobulin G (IgG) to form immune complexes that promote platelet activation. Mechanistically, this activation occurs through stimulation of low-affinity type 2 receptor for Fc portion of IgG (FcγRIIA), a receptor for immune complexes, independently of thrombin. Using a combination of in vitro and in vivo approaches, we found that the antibodies from H3N2-immunized mice activate transgenic mouse platelets that express FcγRIIA when put in the presence of H1N1, suggesting that cross-reacting influenza antibodies suffice. Alternatively, H1N1 can activate platelets via thrombin formation, independently of complement and FcγRIIA. These observations identify both the adaptive immune response and the innate response against pathogens as 2 intertwined processes that activate platelets during influenza infections.
Collapse
|
68
|
Amplification of bacteria-induced platelet activation is triggered by FcγRIIA, integrin αIIbβ3, and platelet factor 4. Blood 2014; 123:3166-74. [PMID: 24642751 DOI: 10.1182/blood-2013-11-540526] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bacterial adhesion to platelets is mediated via a range of strain-specific bacterial surface proteins that bind to a variety of platelet receptors. It is unclear how these interactions lead to platelet activation. We demonstrate a critical role for the immune receptor FcγRIIA, αIIbβ3, and Src and Syk tyrosine kinases in platelet activation by Staphylococcus aureus, Streptococcus sanguinis, Streptococcus gordonii, Streptococcus oralis, and Streptococcus pneumoniae. FcγRIIA activation is dependent on immunoglobulin G (IgG) and αIIbβ3 engagement. Moreover, feedback agonists adenosine 5'-diphosphate and thromboxane A2 are mandatory for platelet aggregation. Additionally, platelet factor 4 (PF4) binds to bacteria and reduces the lag time for aggregation, and gray platelet syndrome α-granule-deficient platelets do not aggregate to 4 of 5 bacterial strains. We propose that FcγRIIA-mediated activation is a common response mechanism used against a wide range of bacteria, and that release of secondary mediators and PF4 serve as a positive feedback mechanism for activation through an IgG-dependent pathway.
Collapse
|
69
|
Abstract
The importance of CLEC-2, a natural ligand/receptor for Gp38/Podoplanin, in the formation of the lymphatic vasculature has recently been demonstrated. As the development and maintenance of lymph nodes (LNs) is dependent on the formation of the lymphatic vasculature and the differentiation of Gp38/Podoplanin(+) stromal cells, we investigated the role of CLEC-2 in lymphoneogenesis and LN homeostasis. Using constitutive Clec1b(-/-) mice, we showed that while CLEC-2 was not necessary for initiation of the LN anlage, it was required at late stages of development. Constitutive deletion of CLEC-2 induced a profound defect in lymphatic endothelial cell proliferation, resulting in lack of LNs at birth. In contrast, conditional deletion of CLEC-2 in the megakaryocyte/platelet lineage in Clec1b(fl/fl)PF4-Cre mice led to the development of blood-filled LNs and fibrosis, in absence of a proliferative defect of the lymphatic endothelial compartment. This phenotype was also observed in chimeric mice reconstituted with Clec1b(fl/fl)PF4-Cre bone marrow, indicating that CLEC-2 expression in platelets was required for LN integrity. We demonstrated that LNs of Clec1b(fl/fl)PF4-Cre mice are able to sustain primary immune responses but show a defect in immune cell recirculation after repeated immunizations, thus suggesting CLEC-2 as target in chronic immune response.
Collapse
|
70
|
Dütting S, Vögtle T, Morowski M, Schiessl S, Schäfer CM, Watson SK, Hughes CE, Ackermann JA, Radtke D, Hermanns HM, Watson SP, Nitschke L, Nieswandt B. Growth factor receptor-bound protein 2 contributes to (hem)immunoreceptor tyrosine-based activation motif-mediated signaling in platelets. Circ Res 2013; 114:444-453. [PMID: 24265393 DOI: 10.1161/circresaha.114.302670] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
RATIONALE Platelets are anuclear cell fragments derived from bone marrow megakaryocytes (MKs) that safeguard vascular integrity but may also cause pathological vessel occlusion. One major pathway of platelet activation is triggered by 2 receptors that signal through an (hem)immunoreceptor tyrosine-based activation motif (ITAM), the activating collagen receptor glycoprotein (GP) VI and the C-type lectin-like receptor 2 (CLEC-2). Growth factor receptor-bound protein 2 (Grb2) is a ubiquitously expressed adapter molecule involved in signaling processes of numerous receptors in different cell types, but its function in platelets and MKs is unknown. OBJECTIVE We tested the hypothesis that Grb2 is a crucial adapter protein in (hem)immunoreceptor tyrosine-based activation motif signaling in platelets. METHODS AND RESULTS Here, we show that genetic ablation of Grb2 in MKs and platelets did not interfere with MK differentiation or platelet production. However, Grb2-deficiency severely impaired glycoprotein VI-mediated platelet activation because of defective stabilization of the linker of activated T-cell (LAT) signalosome and activation of downstream signaling proteins that resulted in reduced adhesion, aggregation, and coagulant activity on collagen in vitro. Similarly, CLEC-2-mediated signaling was impaired in Grb2-deficient platelets, whereas the cells responded normally to stimulation of G protein-coupled receptors. In vivo, this selective (hem)immunoreceptor tyrosine-based activation motif signaling defect resulted in prolonged bleeding times but affected arterial thrombus formation only after concomitant treatment with acetylsalicylic acid, indicating that defective glycoprotein VI signaling in the absence of Grb2 can be compensated through thromboxane A2-induced G protein-coupled receptor signaling pathways. CONCLUSIONS These results reveal an important contribution of Grb2 in (hem)immunoreceptor tyrosine-based activation motif signaling in platelets in hemostasis and thrombosis by stabilizing the LAT signalosome.
Collapse
Affiliation(s)
- Sebastian Dütting
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| | - Timo Vögtle
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| | - Martina Morowski
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| | - Sarah Schiessl
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| | - Carmen M Schäfer
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| | - Stephanie K Watson
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| | - Craig E Hughes
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| | - Jochen A Ackermann
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| | - Daniel Radtke
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| | - Heike M Hermanns
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| | - Steve P Watson
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| | - Lars Nitschke
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| | - Bernhard Nieswandt
- Department of Experimental Biomedicine, University Hospital Würzburg (S.D., T.V., M.M., S.S., B.N.) and Rudolf Virchow Center for Experimental Biomedicine (S.D., T.V., C.M.S., H.M.H., B.N.), University of Würzburg, Würzburg, Germany; Centre for Cardiovascular Sciences, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (S.K.W., C.E.H., S.P.W.); and Department of Biology, Division of Genetics, University of Erlangen, Erlangen, Germany (J.A.A., D.R., L.N.)
| |
Collapse
|
71
|
Abstract
Platelets play a role in cancer by acting as a dynamic reservoir of effectors that facilitate tumor vascularization, growth, and metastasis. However, little information is available about the mechanism of tumor cell-induced platelet secretion (TCIPS) or the molecular machinery by which effector molecules are released from platelets. Here we demonstrate that tumor cells directly induce platelet secretion. Preincubation of platelets with human colon cancer (Caco-2), prostate cancer (PC3M-luc), or breast cancer cells (MDA-MB-231;MCF-7) resulted in a marked dose-dependent secretion of dense granules. Importantly, TCIPS preceded aggregation which always displayed a characteristic lag time. We investigated the role of platelet receptors and downstream molecules in TCIPS. The most potent modulators of TCIPS were the pharmacologic antagonists of Syk kinase, phospholipase C and protein kinase C, all downstream mediators of the immunoreceptor tyrosine-based activation motif (ITAM) cascade in platelets. Supporting this, we demonstrated a central role for the immune Fcγ receptor IIa (FcγRIIa) in mediating platelet-tumor cell cross-talk. In conclusion, we demonstrate that cancer cells can promote platelet dense-granule secretion, which is required to augment platelet aggregation. In addition, we show a novel essential role for FcγRIIa in prostate cancer cell-induced platelet activation opening the opportunity to develop novel antimetastatic therapies.
Collapse
|
72
|
Abstract
PURPOSE OF REVIEW G protein-coupled receptors (GPCRs) like PAR1/4 and P2Y12 have long been known for their critical role in hemostasis. In contrast, deficiency in the immunoreceptor tyrosine-based activation motif (ITAM)-coupled receptors glycoprotein (GP)VI or C-type lectin-like receptor (CLEC)-2 is associated with only a mild bleeding diathesis in humans and mice. This review summarizes recent developments on the physiological importance of platelet ITAM signaling as well as the molecular mechanisms facilitating this signaling pathway. RECENT FINDINGS Genetic experiments identified a critical role for platelet CLEC-2 signaling in the formation of lymphatic vessels during development. Similarly, signaling by both GPVI and CLEC-2, but not GPCRs, is required for the maintenance of vascular integrity at sites of inflammation in the adult. The molecular mechanisms underlying ITAM signaling in platelets continue to be refined. SUMMARY Platelet ITAM signaling plays a key role for the maintenance of vascular integrity in development and the adult. This novel form of hemostasis differs from hemostasis at sites of vascular injury in that it does not depend on major platelet adhesion receptors or GPCR signaling.
Collapse
Affiliation(s)
- Wolfgang Bergmeier
- University of North Carolina, Chapel Hill, North Carolina 27599-7035, USA.
| | | |
Collapse
|
73
|
Heger M, van Golen RF, Broekgaarden M, van den Bos RR, Neumann HAM, van Gulik TM, van Gemert MJC. Endovascular laser–tissue interactions and biological responses in relation to endovenous laser therapy. Lasers Med Sci 2013; 29:405-22. [DOI: 10.1007/s10103-013-1490-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/03/2013] [Indexed: 01/11/2023]
|
74
|
CD36 recruits α₅β₁ integrin to promote cytoadherence of P. falciparum-infected erythrocytes. PLoS Pathog 2013; 9:e1003590. [PMID: 24009511 PMCID: PMC3757042 DOI: 10.1371/journal.ppat.1003590] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 07/15/2013] [Indexed: 12/15/2022] Open
Abstract
The adhesion of Plasmodium falciparum-infected erythrocytes (IRBC) to receptors on different host cells plays a divergent yet critical role in determining the progression and outcome of the infection. Based on our ex vivo studies with clinical parasite isolates from adult Thai patients, we have previously proposed a paradigm for IRBC cytoadherence under physiological shear stress that consists of a recruitment cascade mediated largely by P-selectin, ICAM-1 and CD36 on primary human dermal microvascular endothelium (HDMEC). In addition, we detected post-adhesion signaling events involving Src family kinases and the adaptor protein p130CAS in endothelial cells that lead to CD36 clustering and cytoskeletal rearrangement which enhance the magnitude of the adhesive strength, allowing adherent IRBC to withstand shear stress of up to 20 dynes/cm2. In this study, we addressed whether CD36 supports IRBC adhesion as part of an assembly of membrane receptors. Using a combination of flow chamber assay, atomic force and confocal microscopy, we showed for the first time by loss- and gain-of function assays that in the resting state, the integrin α5β1 does not support adhesive interactions between IRBC and HDMEC. Upon IRBC adhesion to CD36, the integrin is recruited either passively as part of a molecular complex with CD36, or actively to the site of IRBC attachment through phosphorylation of Src family kinases, a process that is Ca2+-dependent. Clustering of β1 integrin is associated with an increase in IRBC recruitment as well as in adhesive strength after attachment (∼40% in both cases). The adhesion of IRBC to a multimolecular complex on the surface of endothelial cells could be of critical importance in enabling adherent IRBC to withstand the high shear stress in the microcirculations. Targeting integrins may provide a novel approach to decrease IRBC cytoadherence to microvascular endothelium. Of the several species of malaria parasites that infect humans, disease caused by Plasmodium falciparum is responsible for most of the deaths. The unique pathological finding of this infection is the intense adhesion of infected red blood cells (IRBC) in the microcirculation, resulting in obstruction to blood flow and organ dysfunction. The focus of our research is to identify the molecules on host endothelial cells that support the adhesion as potential therapeutic targets. In this report, we showed for the first time a functional association between CD36, a well studied adhesion molecule for parasite adhesion, and α5β1, a member of the integrin family of adhesion molecules that are important for adhesion of leukocytes to blood vessels and cell adhesion to the extracellular matrix. We found that by itself, α5β1 integrin does not support IRBC adhesion. When IRBC adhere to CD36, the integrin is recruited to the site of adhesion through activation of the Src family kinase signaling pathway. As a result, both the number of adherent IRBC and the strength with which they adhere is greatly increased. These results demonstrate that IRBC adhesion is a dynamic and complex process. We need to identify each of the functional components to design anti-adhesive therapy.
Collapse
|
75
|
Abstract
In this issue of Blood, Zhi et al demonstrate an important role for Fc γreceptor IIa (FcγRIIa) in platelet functions dependent on integrin α(IIb)β(3) outside-in signals.
Collapse
|
76
|
Berndt MC, Andrews RK. Platelet hem-Immunoreceptor Tyrosine–Based Activation Motif Receptors. Arterioscler Thromb Vasc Biol 2013; 33:884-5. [DOI: 10.1161/atvbaha.113.301400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Michael C. Berndt
- From the Faculty of Health Sciences, Curtin University, Perth, Western Australia, Australia (M.C.B.); and Systems Haematology Laboratory, Australian Centre for Blood Diseases, Department of Clinical Haematology, Monash University, Melbourne, Australia (R.K.A.)
| | - Robert K. Andrews
- From the Faculty of Health Sciences, Curtin University, Perth, Western Australia, Australia (M.C.B.); and Systems Haematology Laboratory, Australian Centre for Blood Diseases, Department of Clinical Haematology, Monash University, Melbourne, Australia (R.K.A.)
| |
Collapse
|
77
|
Bender M, May F, Lorenz V, Thielmann I, Hagedorn I, Finney BA, Vögtle T, Remer K, Braun A, Bösl M, Watson SP, Nieswandt B. Combined in vivo depletion of glycoprotein VI and C-type lectin-like receptor 2 severely compromises hemostasis and abrogates arterial thrombosis in mice. Arterioscler Thromb Vasc Biol 2013; 33:926-34. [PMID: 23448972 DOI: 10.1161/atvbaha.112.300672] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Platelet inhibition is a major strategy to prevent acute ischemic cardiovascular and cerebrovascular events, which may, however, be associated with an increased bleeding risk. The (hem)immunoreceptor tyrosine activation motif-bearing platelet receptors, glycoprotein VI (GPVI) and C-type lectin-like receptor 2 (CLEC-2), might be promising antithrombotic targets because they can be depleted from circulating platelets by antibody treatment, leading to sustained antithrombotic protection, but only moderately increased bleeding times in mice. APPROACH AND RESULTS We investigated whether both (hem)immunoreceptor tyrosine activation motif-bearing receptors can be targeted simultaneously and what the in vivo consequences of such a combined therapeutic GPVI/CLEC-2 deficiency are. We demonstrate that isolated targeting of either GPVI or CLEC-2 in vivo does not affect expression or function of the respective other receptor. Moreover, simultaneous treatment with both antibodies resulted in the sustained loss of both GPVI and CLEC-2, while leaving other activation pathways intact. However, GPVI/CLEC-2-depleted mice displayed a dramatic hemostatic defect and profound impairment of arterial thrombus formation. Furthermore, a strongly diminished hemostatic response could also be reproduced in mice genetically lacking GPVI and CLEC-2. CONCLUSIONS These results demonstrate that GPVI and CLEC-2 can be simultaneously downregulated in platelets in vivo and reveal an unexpected functional redundancy of the 2 receptors in hemostasis and thrombosis. These findings may have important implications of the potential use of anti-GPVI and anti-CLEC-2-based agents in the prevention of thrombotic diseases.
Collapse
Affiliation(s)
- Markus Bender
- University Hospital Würzburg and Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|