Network


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

Hotspot


Dive into the research topics where Pingguo Chen is active.

Publication


Featured researches published by Pingguo Chen.


Blood | 2010

A murine model of severe immune thrombocytopenia is induced by antibody- and CD8+ T cell-mediated responses that are differentially sensitive to therapy

Leola Chow; Rukhsana Aslam; Edwin R. Speck; Michael Kim; Norman Cridland; Michelle Lee Webster; Pingguo Chen; Kim Sahib; Heyu Ni; Alan H. Lazarus; M. Bernadette Garvey; John Freedman; John W. Semple

Immune thrombocytopenia (ITP) is a bleeding disorder characterized by antibody-opsonized platelets being prematurely destroyed in the spleen, although some patients with ITP may have a cell-mediated form of thrombocytopenia. Although several animal models of ITP have been developed, few mimic primary chronic ITP nor have any shown cell-mediated platelet destruction. To create this type of model, splenocytes from CD61 knockout mice immunized against CD61(+) platelets were transferred into severe combined immunodeficient (SCID) (CD61(+)) mouse recipients, and their platelet counts and phenotypes were observed. As few as 5 x 10(4) splenocytes induced a significant thrombocytopenia and bleeding mortality (80%) in recipients within 3 weeks after transfer. Depletion of lymphocyte subsets before transfer showed that the splenocytes ability to induce thrombocytopenia and bleeding completely depended on CD4(+) T helper cells and that both CD19(+) B cell (antibody)- and CD8(+) T cell (cell)-mediated effector mechanisms were responsible. Treatment of the SCID mouse recipients with intravenous gamma-globulins raised platelet counts and completely prevented bleeding mortality induced by antibody-mediated effector mechanisms but did not affect cell-mediated disease. This novel model not only shows both antibody- and cell-mediated ITP and bleeding but also suggests that these 2 effector mechanisms have a differential response to therapy.


Journal of Thrombosis and Haemostasis | 2005

Vitronectin stabilizes thrombi and vessel occlusion but plays a dual role in platelet aggregation

Adili Reheman; Peter L. Gross; Hong Yang; Pingguo Chen; D. Allen; Valery Leytin; Jane E. Freedman; Heyu Ni

Summary.  The role of vitronectin (Vn) in thrombosis is currently controversial; both inhibitory and supportive roles have been reported. To monitor directly the function of Vn in thrombotic events at the site of vascular injury, we studied Vn‐deficient (Vn–/–) and wild‐type (WT) control mice with two real‐time intravital microscopy thrombosis models. In the mesenteric arteriole model, vessel injury was induced by ferric chloride. We observed unstable thrombi and a significantly greater number of emboli in Vn–/– mice. Vessel occlusion was also delayed and frequent vessel re‐opening occurred. In the cremaster muscle arteriole model, vessel injury was induced by a nitrogen dye laser. We observed significantly fewer platelets, lower fibrin content, and unstable fibrin within the thrombi of Vn–/– mice. To define further the role of Vn in thrombus growth, we studied platelet aggregation in vitro. Consistent with our in vivo data, the second wave of thrombin‐induced aggregation of gel‐filtered platelets was abolished at a low concentration of thrombin in Vn–/– platelets. Interestingly, adenosine diphosphate (ADP)‐induced platelet aggregation was significantly increased in Vn–/– platelet‐rich plasma (PRP) and this effect was attenuated by adding purified plasma Vn. We also observed increased platelet aggregation induced by shear stress in Vn–/– whole blood. These data demonstrate that Vn is a thrombus stabilizer. However, in contrast to released platelet granule Vn which enhances platelet aggregation, plasma Vn inhibits platelet aggregation.


Advances in Hematology | 2012

Crosstalk between Platelets and the Immune System: Old Systems with New Discoveries

Conglei Li; June Li; Yan-Yan Li; Sean Lang; Issaka Yougbaré; Guangheng Zhu; Pingguo Chen; Heyu Ni

Platelets are small anucleate cells circulating in the blood. It has been recognized for more than 100 years that platelet adhesion and aggregation at the site of vascular injury are critical events in hemostasis and thrombosis; however, recent studies demonstrated that, in addition to these classic roles, platelets also have important functions in inflammation and the immune response. Platelets contain many proinflammatory molecules and cytokines (e.g., P-selectin, CD40L, IL-1β, etc.), which support leukocyte trafficking, modulate immunoglobulin class switch, and germinal center formation. Platelets express several functional Toll-like receptors (TLRs), such as TLR-2, TLR-4, and TLR-9, which may potentially link innate immunity with thrombosis. Interestingly, platelets also contain multiple anti-inflammatory molecules and cytokines (e.g., transforming growth factor-β and thrombospondin-1). Emerging evidence also suggests that platelets are involved in lymphatic vessel development by directly interacting with lymphatic endothelial cells through C-type lectin-like receptor 2. Besides the active contributions of platelets to the immune system, platelets are passively targeted in several immune-mediated diseases, such as autoimmune thrombocytopenia, infection-associated thrombocytopenia, and fetal and neonatal alloimmune thrombocytopenia. These data suggest that platelets are important immune cells and may contribute to innate and adaptive immunity under both physiological and pathological conditions.


Journal of Thrombosis and Haemostasis | 2006

Fibrinogen and von Willebrand factor-independent platelet aggregation in vitro and in vivo

Hong Yang; Adili Reheman; Pingguo Chen; Guangheng Zhu; Richard O. Hynes; Jane E. Freedman; Denisa D. Wagner; Heyu Ni

Summary.  Background: Fibrinogen (Fg) has been considered essential for platelet aggregation. However, we recently demonstrated formation of occlusive thrombi in Fg‐deficient mice and in mice doubly deficient for Fg and von Willebrand factor (Fg/VWF−/−). Methods and results: Here we studied Fg/VWF‐independent platelet aggregation in vitro and found no aggregation in citrated platelet‐rich plasma of Fg/VWF−/− mice. Surprisingly, in Fg/VWF−/− plasma without anticoagulant, adenosine diphosphate induced robust aggregation of Fg/VWF−/− platelets but not of β3‐integrin‐deficient (β3−/−) platelets. In addition, β3−/− platelets did not significantly incorporate into thrombi in Fg/VWF−/− mice. This Fg/VWF‐independent aggregation was blocked by thrombin inhibitors (heparin, hirudin, PPACK), and thrombin or thrombin receptor activation peptide (AYPGKF‐NH2) induced aggregation of gel‐filtered Fg/VWF−/− platelets in 1 mm Ca2+ PIPES buffer. Notably, aggregation in PIPES buffer was only 50–60% of that observed in Fg/VWF−/− plasma. Consistent with the requirement for thrombin in vitro, hirudin completely inhibited thrombus formation in Fg/VWF−/− mice. These data define a novel pathway of platelet aggregation independent of both Fg and VWF. Although this pathway was not detected in the presence of anticoagulants, it was observed under physiological conditions in vivo and in the presence of Ca2+in vitro. Conclusions: β3 integrin, thrombin, and Ca2+ play critical roles in this Fg/VWF‐independent aggregation, and both plasma and platelet granule proteins contribute to this process.


Blood | 2009

Fibrinogen is required for maintenance of platelet intracellular and cell-surface P-selectin expression

Hong Yang; Sean Lang; Zhimin Zhai; Ling Li; Walter H. A. Kahr; Pingguo Chen; Jelena Brkić; Christopher M. Spring; Matthew J. Flick; Jay L. Degen; John Freedman; Heyu Ni

Platelet P-selectin plays important roles in inflammation and contributes to thrombosis and hemostasis. Although it has been reported that von Willebrand factor (VWF) affects P-selectin expression on endothelial cells, little information is available regarding regulation of platelet P-selectin expression. Here, we first observed that P-selectin expression was significantly decreased on platelets of fibrinogen and VWF double-deficient mice. Subsequently, we identified this was due to fibrinogen deficiency. Impaired P-selectin expression on fibrinogen-deficient platelets was further confirmed in human hypofibrinogenemic patients. We demonstrated that this impairment is unlikely due to excessive P-selectin shedding, deficient fibrinogen-mediated cell surface P-selectin binding, or impaired platelet granule release, but rather is due to decreased platelet P-selectin content. Fibrinogen transfusion completely recovered this impairment in fibrinogen-deficient (Fg(-/-)) mice, and engagement of the C-terminus of the fibrinogen gamma chain with beta3 integrin was required for this process. Furthermore, Fg(-/-) platelets significantly increased P-selectin expression following transfusion into beta3 integrin-deficient mice and when cultured with fibrinogen. These data suggest fibrinogen may play important roles in inflammation, thrombosis, and hemostasis via enhancement of platelet P-selectin expression. Since human fibrinogen levels vary significantly in normal and diseased populations, P-selectin as an activation marker on platelets should be used with caution.


Blood | 2010

Animal model of fetal and neonatal immune thrombocytopenia: role of neonatal Fc receptor in the pathogenesis and therapy

Pingguo Chen; Conglei Li; Sean Lang; Guangheng Zhu; Adili Reheman; Christopher M. Spring; John Freedman; Heyu Ni

Fetal and neonatal immune thrombocytopenia (FNIT) is a severe bleeding disorder in which maternal antibodies cross the placenta and destroy fetal/neonatal platelets. It has been demonstrated that the neonatal Fc receptor (FcRn) regulates immunoglobulin G (IgG) homeostasis and plays an important role in transplacental IgG transport. However, the role of FcRn in the pathogenesis and therapy of FNIT has not been studied. Here, we developed an animal model of FNIT using combined β3 integrin-deficient and FcRn-deficient (β3(-/-)FcRn(-/-)) mice. We found that β3(-/-)FcRn(-/-) mice are immunoresponsive to β3(+/+)FcRn(-/-) platelets. The generated antibodies were β3 integrin specific and were maintained at levels that efficiently induced thrombocytopenia in adult β3(+/+)FcRn(-/-) mice. FNIT was observed when immunized β3(-/-)FcRn(+/+) females were bred with β3(+/+)FcRn(+/+) males, while no FNIT occurred in β3(-/-)FcRn(-/-) females bred with β3(+/+)FcRn(-/-) males, suggesting that FcRn is indispensable for the induction of FNIT. We further demonstrated that fetal FcRn was responsible for the transplacental transport of various IgG isotypes. We found that anti-FcRn antibody and intravenous IgG prevented FNIT, and that intravenous IgG ameliorated FNIT through both FcRn-dependent and -independent pathways. Our data suggest that targeting FcRn may be a potential therapy for human FNIT as well as other maternal pathogenic antibody-mediated diseases.


Journal of Clinical Investigation | 2015

Maternal anti-platelet β3 integrins impair angiogenesis and cause intracranial hemorrhage

Issaka Yougbaré; Sean Lang; Hong Yang; Pingguo Chen; Xu Zhao; Wei-She Tai; Darko Zdravic; Brian Vadasz; Conglei Li; Siavash Piran; Alexandra H. Marshall; Guangheng Zhu; Heidi Tiller; Mette Kjær Killie; Shelley Boyd; Howard Leong-Poi; Xiao-Yan Wen; Bjørn Skogen; S. Lee Adamson; John Freedman; Heyu Ni

Fetal and neonatal alloimmune thrombocytopenia (FNAIT) is a life-threatening disease in which intracranial hemorrhage (ICH) is the major risk. Although thrombocytopenia, which is caused by maternal antibodies against β3 integrin and occasionally by maternal antibodies against other platelet antigens, such as glycoprotein GPIbα, has long been assumed to be the cause of bleeding, the mechanism of ICH has not been adequately explored. Utilizing murine models of FNAIT and a high-frequency ultrasound imaging system, we found that ICH only occurred in fetuses and neonates with anti-β3 integrin-mediated, but not anti-GPIbα-mediated, FNAIT, despite similar thrombocytopenia in both groups. Only anti-β3 integrin-mediated FNAIT reduced brain and retina vessel density, impaired angiogenic signaling, and increased endothelial cell apoptosis, all of which were abrogated by maternal administration of intravenous immunoglobulin (IVIG). ICH and impairment of retinal angiogenesis were further reproduced in neonates by injection of anti-β3 integrin, but not anti-GPIbα antisera. Utilizing cultured human endothelial cells, we found that cell proliferation, network formation, and AKT phosphorylation were inhibited only by murine anti-β3 integrin antisera and human anti-HPA-1a IgG purified from mothers with FNAIT children. Our data suggest that fetal hemostasis is distinct and that impairment of angiogenesis rather than thrombocytopenia likely causes FNAIT-associated ICH. Additionally, our results indicate that maternal IVIG therapy can effectively prevent this devastating disorder.


Transfusion | 2012

Toward a prophylaxis against fetal and neonatal alloimmune thrombocytopenia: induction of antibody‐mediated immune suppression and prevention of severe clinical complications in a murine model

Heidi Tiller; Mette Kjær Killie; Pingguo Chen; Mariana Eksteen; Anne Husebekk; Bjørn Skogen; Jens Kjeldsen-Kragh; Heyu Ni

BACKGROUND: Fetal and neonatal alloimmune thrombocytopenia (FNAIT) is a severe bleeding disorder caused by maternal antibody–mediated destruction of fetal or neonatal platelets (PLTs). Results from our recent large screening study suggest that the pathophysiology of FNAIT is more similar to hemolytic disease of the fetus and newborn (HDFN) than previously thought. Immunization against HPA‐1a might therefore be preventable by a prophylactic regimen of inducing antibody‐mediated immune suppression (AMIS), which has been documented to be a useful prophylaxis against HDFN. This preclinical proof‐of‐concept study investigated whether passive administration of anti‐β3 integrin could induce AMIS and thereby prevent clinical complications of FNAIT.


Cardiovascular and Hematological Disorders - Drug Targets | 2013

Fc-independent phagocytosis: implications for IVIG and other therapies in immune-mediated thrombocytopenia.

June Li; Dianne E. van der Wal; Lingyan Zhu; Brian Vadasz; Elisa K. Simpson; Conglei Li; Michelle Lee Webster; Guangheng Zhu; Sean Lang; Pingguo Chen; Qingshu Zeng; Heyu Ni

Phagocytes were first described by Dr. Metchnikoff in 1873. The roles of phagocytes in innate and adaptive immunity have been well established to date, although the molecular mechanisms involved in initiating phagocytosis (through Fc or other receptors) remain to be further explored. Phagocytes in the reticuloendothelial system, particularly macrophages, have been implicated in the clearance of senescent blood cells. The destruction of these cells may be primarily mediated via an Fc-independent pathway. Fc-independent phagocytosis may also play an important role in platelet clearance, including in autoimmune thrombocytopenia (ITP), and in clearance of platelet-rich emboli detached from sites of vascular injury. In ITP, the two major platelet auto-antigens have been located on glycoprotein (GP)IIbIIIa and the GPIb complex. It has been demonstrated that anti-GPIb antibodies, in contrast to anti-GPIIbIIIa, can induce thrombocytopenia in an Fc-independent manner. We further demonstrated in an animal model that intravenous IgG (IVIG) is unable to ameliorate thrombocytopenia caused by most anti-GPIb antibodies, despite its efficacy in anti- GPIIbIIIa-mediated thrombocytopenia. Our data was supported by subsequent retrospective studies with ITP patients by several independent groups. Most recently, we found that anti-GPIb-mediated ITP was also resistant to steroid therapy and that platelet activation and apoptosis induced by anti-GPIb antibodies may be involved in the Fc-independent platelet clearance. Therefore, identification of antibody specificity in patients, e.g. anti-GPIIbIIIa (Fc-dependent) versus anti-GPIb (Fc-independent), may be important for therapies against ITP, as well as other immune-mediated thrombocytopenias.


Thrombosis Journal | 2016

Platelets and platelet adhesion molecules: novel mechanisms of thrombosis and anti-thrombotic therapies

Xiaohong Ruby Xu; Naadiya Carrim; Miguel Antonio Dias Neves; Thomas McKeown; Tyler W. Stratton; Rodrigo Matos Pinto Coelho; Xi Lei; Pingguo Chen; Jianhua Xu; Xiangrong Dai; Benjamin Xiaoyi Li; Heyu Ni

Platelets are central mediators of thrombosis and hemostasis. At the site of vascular injury, platelet accumulation (i.e. adhesion and aggregation) constitutes the first wave of hemostasis. Blood coagulation, initiated by the coagulation cascades, is the second wave of thrombin generation and enhance phosphatidylserine exposure, can markedly potentiate cell-based thrombin generation and enhance blood coagulation. Recently, deposition of plasma fibronectin and other proteins onto the injured vessel wall has been identified as a new “protein wave of hemostasis” that occurs prior to platelet accumulation (i.e. the classical first wave of hemostasis). These three waves of hemostasis, in the event of atherosclerotic plaque rupture, may turn pathogenic, and cause uncontrolled vessel occlusion and thrombotic disorders (e.g. heart attack and stroke). Current anti-platelet therapies have significantly reduced cardiovascular mortality, however, on-treatment thrombotic events, thrombocytopenia, and bleeding complications are still major concerns that continue to motivate innovation and drive therapeutic advances. Emerging evidence has brought platelet adhesion molecules back into the spotlight as targets for the development of novel anti-thrombotic agents. These potential antiplatelet targets mainly include the platelet receptors glycoprotein (GP) Ib-IX-V complex, β3 integrins (αIIb subunit and PSI domain of β3 subunit) and GPVI. Numerous efforts have been made aiming to balance the efficacy of inhibiting thrombosis without compromising hemostasis. This mini-review will update the mechanisms of thrombosis and the current state of antiplatelet therapies, and will focus on platelet adhesion molecules and the novel anti-thrombotic therapies that target them.

Collaboration


Dive into the Pingguo Chen's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heyu Ni

St. Michael's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Heyu Ni

St. Michael's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

June Li

St. Michael's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sean Lang

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge