Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Binggang Xiang is active.

Publication


Featured researches published by Binggang Xiang.


Nature Communications | 2013

Platelets Protect from Septic Shock by Inhibiting Macrophage-Dependent Inflammation via the Cyclooxygenase 1 Signalling Pathway

Binggang Xiang; Guoying Zhang; Ling Guo; Xiang-An Li; Andrew J. Morris; Alan Daugherty; Sidney W. Whiteheart; Susan S. Smyth; Zhenyu Li

Although it has long been known that patients with sepsis often have thrombocytopenia and that septic patients with severe thrombocytopenia have a poor prognosis and higher mortality, the role of platelets in the pathogenesis of sepsis is poorly understood. Here we report a protective role of platelets in septic shock. We show that experimental thrombocytopenia induced by intraperitoneal injection of an anti-glycoprotein Ibα monoclonal antibody increases mortality and aggravates organ failure, whereas transfusion of platelets reduces mortality in lipopolysaccharide-induced endotoxemia and a bacterial infusion mouse sepsis model. Plasma concentrations of proinflammatory cytokines TNF-α and IL-6 are elevated by thrombocytopenia and decreased by platelet transfusion in septic mice. Furthermore, we identify that platelets protect from septic shock by inhibiting macrophage-dependent inflammation via the COX1/PGE₂/EP4-dependent pathway. Thus, these findings demonstrate a previously unappreciated role for platelets in septic shock and suggest that platelet transfusion may be effective in treating severely septic patients.


Journal of Biological Chemistry | 2011

Distinct Roles for Rap1b Protein in Platelet Secretion and Integrin αIIbβ3 Outside-in Signaling*

Guoying Zhang; Binggang Xiang; Shaojing Ye; Magdalena Chrzanowska-Wodnicka; Andrew J. Morris; T. Kent Gartner; Sidney W. Whiteheart; Gilbert C. White; Susan S. Smyth; Zhenyu Li

Background: Rap1b is a small G protein that is a key regulator for platelet activation. Results: Agonist-induced Rap1b activation plays a role in platelet secretion, and integrin outside-in signaling-mediated Rap1b activation is important in platelet spreading on fibrinogen and clot retraction. Conclusion: There are dual activation mechanisms of Rap1 that play distinct roles in platelet function. Significance: Learning two novel functions of Rap1b in platelets. Rap1b is activated by platelet agonists and plays a critical role in integrin αIIbβ3 inside-out signaling and platelet aggregation. Here we show that agonist-induced Rap1b activation plays an important role in stimulating secretion of platelet granules. We also show that αIIbβ3 outside-in signaling can activate Rap1b, and integrin outside-in signaling-mediated Rap1b activation is important in facilitating platelet spreading on fibrinogen and clot retraction. Rap1b-deficient platelets had diminished ATP secretion and P-selectin expression induced by thrombin or collagen. Importantly, addition of low doses of ADP and/or fibrinogen restored aggregation of Rap1b-deficient platelets. Furthermore, we found that Rap1b was activated by platelet spreading on immobilized fibrinogen, a process that was not affected by P2Y12 or TXA2 receptor deficiency, but was inhibited by the selective Src inhibitor PP2, the PKC inhibitor Ro-31-8220, or the calcium chelator demethyl-1,2-bis(2-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid tetrakis. Clot retraction was abolished, and platelet spreading on fibrinogen was diminished in Rap1b-deficient platelets compared with wild-type controls. The defects in clot retraction and spreading on fibrinogen of Rap1b-deficient platelets were not rescued by addition of MnCl2, which elicits αIIbβ3 outside-in signaling in the absence of inside-out signaling. Thus, our results reveal two different activation mechanisms of Rap1b as well as novel functions of Rap1b in platelet secretion and in integrin αIIbβ3 outside-in signaling.


Blood | 2011

Biphasic roles for soluble guanylyl cyclase (sGC) in platelet activation.

Guoying Zhang; Binggang Xiang; Anping Dong; Radek C. Skoda; Alan Daugherty; Susan S. Smyth; Xiaoping Du; Zhenyu Li

Nitric oxide (NO) stimulates cGMP synthesis by activating its intracellular receptor, soluble guanylyl cyclase (sGC). It is a currently prevailing concept that No and cGMP inhibits platelet function. However, the data supporting the inhibitory role of NO/sGC/cGMP in platelets have been obtained either in vitro or using whole body gene deletion that affects vessel wall function. Here we have generated mice with sGC gene deleted only in megakaryocytes and platelets. Using the megakaryocyte- and platelet-specific sGC-deficient mice, we identify a stimulatory role of sGC in platelet activation and in thrombosis in vivo. Deletion of sGC in platelets abolished cGMP production induced by either NO donors or platelet agonists, caused a marked defect in aggregation and attenuated secretion in response to low doses of collagen or thrombin. Importantly, megakaryocyte- and platelet-specific sGC deficient mice showed prolonged tail-bleeding times and impaired FeCl₃-induced carotid artery thrombosis in vivo. Interestingly, the inhibitory effect of the NO donor SNP on platelet activation was sGC-dependent only at micromolar concentrations, but sGC-independent at millimolar concentrations. Together, our data demonstrate important roles of sGC in stimulating platelet activation and in vivo thrombosis and hemostasis, and sGC-dependent and -independent inhibition of platelets by NO donors.


Blood | 2015

Autophagy is induced upon platelet activation and is essential for hemostasis and thrombosis

Madhu M. Ouseph; Yunjie Huang; Meenakshi Banerjee; Smita Joshi; Laura J. MacDonald; Yu Zhong; Huijuan Liu; Xianting Li; Binggang Xiang; Guoying Zhang; Massaki Komatsu; Zhenyu Yue; Zhenyu Li; Brian Storrie; Sidney W. Whiteheart; Qing Jun Wang

Autophagy is important for maintaining cellular homeostasis, and thus its deficiency is implicated in a broad spectrum of human diseases. Its role in platelet function has only recently been examined. Our biochemical and imaging studies demonstrate that the core autophagy machinery exists in platelets, and that autophagy is constitutively active in resting platelets. Moreover, autophagy is induced upon platelet activation, as indicated by agonist-induced loss of the autophagy marker LC3II. Additional experiments, using inhibitors of platelet activation, proteases, and lysosomal acidification, as well as platelets from knockout mouse strains, show that agonist-induced LC3II loss is a consequence of platelet signaling cascades and requires proteases, acidic compartments, and membrane fusion. To assess the physiological role of platelet autophagy, we generated a mouse strain with a megakaryocyte- and platelet-specific deletion of Atg7, an enzyme required for LC3II production. Ex vivo analysis of platelets from these mice shows modest defects in aggregation and granule cargo packaging. Although these mice have normal platelet numbers and size distributions, they exhibit a robust bleeding diathesis in the tail-bleeding assay and a prolonged occlusion time in the FeCl3-induced carotid injury model. Our results demonstrate that autophagy occurs in platelets and is important for hemostasis and thrombosis.


Journal of Thrombosis and Haemostasis | 2010

A Gi‐independent mechanism mediating Akt phosphorylation in platelets

Binggang Xiang; Guoying Zhang; J. Liu; Andrew J. Morris; Susan S. Smyth; T. K. Gartner; Zhenyu Li

Summary.  Background: The serine‐threonine kinase Akt plays an important role in regulating platelet activation. Stimulation of platelets with various agonists results in Akt activation as indicated by Akt phosphorylation. However, the mechanisms of Akt phosphorylation in platelets are not completely understood. Objectives and Methods: We used P2Y12 knockout mice to address the role of P2Y12 in Akt phosphorylation in response to thrombin receptors in platelets. Results: Thrombin or the PAR4 thrombin receptor peptide AYPGKF at high concentrations stimulated substantial phosphorylation of Akt residues Thr308 and Ser473 in P2Y12‐deficient platelets. AYPGKF‐induced Akt phosphorylation is enhanced by expression of recombinant human PAR4 cDNA in Chinese hamster ovary (CHO) cells. P2Y12‐independent Akt phosphorylation was not inhibited by integrin inhibitor peptide RGDS or integrin β3 deficiency. Akt phosphorylation induced by thrombin or AYPGKF in P2Y12‐deficient platelets was inhibited by the calcium chelator dimethyl‐BAPTA, the Src family kinase inhibitor PP2, and PI3K inhibitors, respectively. Conclusions: Our results reveal a novel P2Y12‐independent signaling pathway mediating Akt phosphorylation in response to thrombin receptors.


Journal of Biological Chemistry | 2012

The Src Family Kinases and Protein Kinase C Synergize to Mediate Gq-dependent Platelet Activation

Binggang Xiang; Guoying Zhang; Lucia Stefanini; Wolfgang Bergmeier; T. Kent Gartner; Sidney W. Whiteheart; Zhenyu Li

Background: The role of SFKs in G protein-coupled receptor-mediated platelet activation is not well understood. Results: AYPGKF induced Gq/Ca2+-dependent SFK phosphorylation, and AYPGKF-elicited platelet activation was partially inhibited by PP2 but was completely abolished by PKC inhibitors plus SFK inhibitors. Conclusion: Ca2+/SFKs/PI3K and PKC represent alternative pathways mediating AYPGKF-dependent platelet activation. Significance: This work increases understanding of important SFK functions in platelet activation. The Src family kinases (SFKs) play essential roles in collagen- and von Willebrand factor (VWF)-mediated platelet activation. However, the roles of SFKs in G protein-coupled receptor-mediated platelet activation and the molecular mechanisms whereby SFKs are activated by G protein-coupled receptor stimulation are not fully understood. Here we show that the thrombin receptor protease-activated receptor 4 agonist peptide AYPGKF elicited SFK phosphorylation in P2Y12 deficient platelets but stimulated minimal SFK phosphorylation in platelets lacking Gq. We have previously shown that thrombin-induced SFK phosphorylation was inhibited by the calcium chelator 5,5′-dimethyl-bis-(o-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid (dimethyl-BAPTA). The calcium ionophore A23187 induced SFK phosphorylation in both wild-type and Gq deficient platelets. Together, these results indicate that SFK phosphorylation in response to thrombin receptor stimulation is downstream from Gq/Ca2+ signaling. Moreover, A23187-induced thromboxane A2 synthesis, platelet aggregation, and secretion were inhibited by preincubation of platelets with a selective SFK inhibitor, PP2. AYPGKF-induced thromboxane A2 production in wild-type and P2Y12 deficient platelets was abolished by PP2, and AYPGKF-mediated P-selectin expression, integrin αIIbβ3 activation, and aggregation of P2Y12 deficient platelets were partially inhibited by the PKC inhibitor Ro-31-8220, PP2, dimethyl-BAPTA, or LY294002, but were abolished by Ro-31-8220 plus PP2, dimethyl-BAPTA, or LY294002. These data indicate that Ca2+/SFKs/PI3K and PKC represent two alternative signaling pathways mediating Gq-dependent platelet activation.


Circulation | 2015

Characterization of a Novel Integrin Binding Protein, VPS33B, Which is Important for Platelet Activation and in vivo Thrombosis and Hemostasis

Binggang Xiang; Guoying Zhang; Shaojing Ye; Rui Zhang; Cai Huang; Jun Liu; Min Tao; Ruan C; Susan S. Smyth; Sidney W. Whiteheart; Zhenyu Li

Background— Integrins are heterodimeric (&agr;/&bgr;) membrane proteins that play fundamental roles in many biological processes, for example, cell adhesion and spreading, which are important for platelet function and hemostasis. The molecular mechanism that regulates integrin activation is not completely understood. Methods and Results— Here, we show that VPS33B, a member of the Sec1/Munc18 family, binds directly to the integrin &bgr; subunit. Overexpression of VPS33B in Chinese hamster ovary cells potentiated &agr;IIb&bgr;3 outside-in signaling but not inside-out signaling. Platelets, from megakaryocyte- and platelet-specific VPS33B conditional knockout mice, had normal morphology, yet their spreading on fibrinogen was impaired and they failed to support clot retraction. Platelet aggregation and ATP secretion in response to low-dose agonists were reduced in the VPS33B knockout mice. &agr;IIb&bgr;3-mediated endocytosis of fibrinogen was also defective. Tail bleeding times and times to occlusion in an FeCl3-induced thrombosis model were prolonged in the VPS33B knockout mice. Furthermore, VPS33B acted upstream of the RhoA-ROCK-MLC and Rac1-dependent pathways that lead to clot retraction and cell spreading, respectively. Conclusions— Our work demonstrates that vesicular trafficking complexes, containing VPS33B, are a novel class of modifiers of integrin function. Our data also provide insights into the molecular mechanism and treatment of arthrogryposis, renal dysfunction, and cholestasis syndrome.


PLOS ONE | 2012

The P2Y12 Antagonists, 2MeSAMP and Cangrelor, Inhibit Platelet Activation through P2Y12/Gi-Dependent Mechanism

Binggang Xiang; Guoying Zhang; Hongmei Ren; Manjula Sunkara; Andrew J. Morris; T. Kent Gartner; Susan S. Smyth; Zhenyu Li

Background ADP is an important physiological agonist that induces integrin activation and platelet aggregation through its receptors P2Y1 (Gαq-coupled) and P2Y12 (Gαi-coupled). P2Y12 plays a critical role in platelet activation and thrombosis. Adenosine-based P2Y12 antagonists, 2-methylthioadenosine 5′-monophosphate triethylammonium salt hydrate (2MeSAMP) and Cangrelor (AR-C69931MX) have been widely used to demonstrate the role of P2Y12 in platelet function. Cangrelor is being evaluated in clinical trials of thrombotic diseases. However, a recent study reported that both 2MeSAMP and Cangrelor raise intra-platelet cAMP levels and inhibit platelet aggregation through a P2Y12-independent mechanism. Methodology/Principal Findings The present work, using P2Y12 deficient mice, sought to clarify previous conflicting reports and to elucidate the mechanisms by which 2MeSAMP and Cangrelor inhibit platelet activation and thrombosis. 2MeSAMP and Cangrelor inhibited aggregation and ATP release of wild-type but not P2Y12 deficient platelets. 2MeSAMP and Cangrelor neither raised intracellular cAMP concentrations nor induced phosphorylation of vasodilator-stimulated phosphoprotein (VASP) in washed human or mouse platelets. Furthermore, unlike the activators (PGI2 and forskolin) of the cAMP pathway, 2MeSAMP and Cangrelor failed to inhibit Ca2+ mobilization, Akt phosphorylation, and Rap1b activation in P2Y12 deficient platelets. Importantly, while injection of Cangrelor inhibited thrombus formation in a FeCl3-induced thrombosis model in wild-type mice, it failed to affect thrombus formation in P2Y12 deficient mice. Conclusions These data together demonstrate that 2MeSAMP and Cangrelor inhibit platelet function through the P2Y12-dependent mechanism both in vitro and in vivo.


Scientific Reports | 2017

TRAF3 negatively regulates platelet activation and thrombosis

Rui Zhang; Guoying Zhang; Binggang Xiang; Xiaofeng Chen; Lijang Tang; Shaojun Shi; Yani Liu; Xun Ai; Ping Xie; Zhenyu Li

CD40 ligand (CD40L), a member of the tumor necrosis factor (TNF) superfamily, binds to CD40, leading to many effects depending on target cell type. Platelets express CD40L and are a major source of soluble CD40L. CD40L has been shown to potentiate platelet activation and thrombus formation, involving both CD40-dependent and -independent mechanisms. A family of proteins called TNF receptor associated factors (TRAFs) plays key roles in mediating CD40L-CD40 signaling. Platelets express several TRAFs. It has been shown that TRAF2 plays a role in CD40L-mediated platelet activation. Here we show that platelet also express TRAF3, which plays a negative role in regulating platelet activation. Thrombin- or collagen-induced platelet aggregation and secretion are increased in TRAF3 knockout mice. The expression levels of collagen receptor GPVI and integrin αIIbβ3 in platelets were not affected by deletion of TRAF3, suggesting that increased platelet activation in the TRAF3 knockout mice was not due to increased expression platelet receptors. Time to formation of thrombi in a FeCl3-induced thrombosis model was significantly shortened in the TRAF3 knockout mice. However, mouse tail-bleeding times were not affected by deletion of TRAF3. Thus, TRAF3 plays a negative role in platelet activation and in thrombus formation in vivo.


Blood | 2016

Arf6 controls platelet spreading and clot retraction via integrin αIIbβ3 trafficking

Yunjie Huang; Smita Joshi; Binggang Xiang; Yasunori Kanaho; Zhenyu Li; Beth A. Bouchard; Carole L. Moncman; Sidney W. Whiteheart

Collaboration


Dive into the Binggang Xiang's collaboration.

Top Co-Authors

Avatar

Zhenyu Li

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shaojing Ye

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cai Huang

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge