Network


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

Hotspot


Dive into the research topics where Huiying Zhi is active.

Publication


Featured researches published by Huiying Zhi.


Blood | 2016

CRISPR/Cas9-mediated conversion of human platelet alloantigen allotypes

Nanyan Zhang; Huiying Zhi; Brian R. Curtis; Sridhar Rao; Chintan Jobaliya; Mortimer Poncz; Deborah L. French; Peter J. Newman

Human platelet alloantigens (HPAs) reside on functionally important platelet membrane glycoproteins and are caused by single nucleotide polymorphisms in the genes that encode them. Antibodies that form against HPAs are responsible for several clinically important alloimmune bleeding disorders, including fetal and neonatal alloimmune thrombocytopenia and posttransfusion purpura. The HPA-1a/HPA-1b alloantigen system, also known as the Pl(A1)/Pl(A2) polymorphism, is the most frequently implicated HPA among whites, and a single Leu33Pro amino acid polymorphism within the integrin β3 subunit is responsible for generating the HPA-1a/HPA-1b alloantigenic epitopes. HPA-1b/b platelets, like those bearing other low-frequency platelet-specific alloantigens, are relatively rare in the population and difficult to obtain for purposes of transfusion therapy and diagnostic testing. We used CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR associated protein 9) gene-editing technology to transform Leu33 (+) megakaryocytelike DAMI cells and induced pluripotent stem cells (iPSCs) to the Pro33 allotype. CD41(+) megakaryocyte progenitors derived from these cells expressed the HPA-1b (Pl(A2)) alloantigenic epitope, as reported by diagnostic NciI restriction enzyme digestion, DNA sequencing, and western blot analysis using HPA-1b-specific human maternal alloantisera. Application of CRISPR/Cas9 technology to genetically edit this and other clinically-important HPAs holds great potential for production of designer platelets for diagnostic, investigative, and, ultimately, therapeutic use.


PLOS ONE | 2015

Platelet Activation and Thrombus Formation over IgG Immune Complexes Requires Integrin αIIbβ3 and Lyn Kinase

Huiying Zhi; Jing Dai; Junling Liu; Jieqing Zhu; Debra K. Newman; Cunji Gao; Peter J. Newman

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.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Platelets release pathogenic serotonin and return to circulation after immune complex-mediated sequestration

Nathalie Cloutier; Isabelle Allaeys; Geneviève Marcoux; Kellie R. Machlus; Benoit Mailhot; Anne Zufferey; Tania Lévesque; Yann Becker; Nicolas Tessandier; Imene Melki; Huiying Zhi; Guy G. Poirier; Matthew T. Rondina; Joseph E. Italiano; Louis Flamand; Steven E. McKenzie; Francine Côté; Bernhard Nieswandt; Waliul I. Khan; Matthew J. Flick; Peter J. Newman; Steve Lacroix; Paul R. Fortin; Eric Boilard

Significance Immune complexes (ICs) form when antibodies encounter their antigens. ICs are present in blood in multiple pathological conditions. Given the abundance of platelets in blood and that they express a receptor for ICs, called Fcγ receptor IIA (FcγRIIA), we examined the impact of ICs in blood in a mouse model. We found that circulating ICs induced systemic shock, characterized by loss of consciousness, by activating platelet FcγRIIA. Shock was mediated by the liberation of serotonin, a molecule better known for its role in the brain, from platelet granules. During shock, platelets were sequestered in the lungs and brain and returned to the blood circulation after their degranulation. Platelets are thus crucial in response to ICs. There is a growing appreciation for the contribution of platelets to immunity; however, our knowledge mostly relies on platelet functions associated with vascular injury and the prevention of bleeding. Circulating immune complexes (ICs) contribute to both chronic and acute inflammation in a multitude of clinical conditions. Herein, we scrutinized platelet responses to systemic ICs in the absence of tissue and endothelial wall injury. Platelet activation by circulating ICs through a mechanism requiring expression of platelet Fcγ receptor IIA resulted in the induction of systemic shock. IC-driven shock was dependent on release of serotonin from platelet-dense granules secondary to platelet outside-in signaling by αIIbβ3 and its ligand fibrinogen. While activated platelets sequestered in the lungs and leaky vasculature of the blood–brain barrier, platelets also sequestered in the absence of shock in mice lacking peripheral serotonin. Unexpectedly, platelets returned to the blood circulation with emptied granules and were thereby ineffective at promoting subsequent systemic shock, although they still underwent sequestration. We propose that in response to circulating ICs, platelets are a crucial mediator of the inflammatory response highly relevant to sepsis, viremia, and anaphylaxis. In addition, platelets recirculate after degranulation and sequestration, demonstrating that in adaptive immunity implicating antibody responses, activated platelets are longer lived than anticipated and may explain platelet count fluctuations in IC-driven diseases.


PLOS ONE | 2015

Restoration of responsiveness of phospholipase Cγ2-deficient platelets by enforced expression of phospholipase Cγ1.

Yongwei Zheng; Tamara S. Adams; Huiying Zhi; Mei Yu; Renren Wen; Peter J. Newman; Demin Wang; Debra K. Newman

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.


Blood | 2013

Cooperative integrin/ITAM signaling in platelets enhances thrombus formation in vitro and in vivo.

Huiying Zhi; Lubica Rauova; Vincent Hayes; Cunji Gao; Brian Boylan; Debra K. Newman; Steven E. McKenzie; Brian C. Cooley; Mortimer Poncz; Peter J. Newman


Blood | 2015

Crispr/Cas9-Mediated Generation of Mouse Platelets Expressing the Human Platelet Alloantigen, HPA-1a

Huiying Zhi; Nanyan Zhang; Jamie Foeckler; Shawn Kalloway; Sridhar Rao; Brian R. Curtis; Hartmut Weiler; Peter J. Newman


Blood | 2015

Involvement of the EGF Domain in the Formation of the Human Platelet Alloantigen, HPA-1a

Huiying Zhi; Panida Lertkiatmongkol; Brian R. Curtis; Peter J. Newman


Blood | 2014

Designer Platelets: Crispr/Cas-Mediated Conversion of Human Platelet Alloantigen Allotypes

Nanyan Zhang; Huiying Zhi; Sridhar Rao; Peter J. Newman


Archive | 2013

formation in vitro and in vivo Cooperative integrin/ITAM signaling in platelets enhances thrombus

Brian C. Cooley; Mortimer Poncz; Peter J. Newman; Huiying Zhi; Lubica Rauova; Vincent Hayes; Cunji Gao; Brian Boylan; Debra K. Newman; E Steven


Arteriosclerosis, Thrombosis, and Vascular Biology | 2013

Abstract 25: Platelet Activation by GPVI, GPIb-V-IX and aIIbß3 Requires PLC?2 but Not PLC?1

Yongwei Zheng; Huiying Zhi; Tamara S. Adams; Renren Wen; Peter J. Newman; Demin Wang; Debra K. Newman

Collaboration


Dive into the Huiying Zhi's collaboration.

Top Co-Authors

Avatar

Peter J. Newman

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Debra K. Newman

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mortimer Poncz

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Brian Boylan

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Brian C. Cooley

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Brian R. Curtis

Gulf Coast Regional Blood Center

View shared research outputs
Top Co-Authors

Avatar

Lubica Rauova

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Sridhar Rao

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Steven E. McKenzie

Thomas Jefferson University

View shared research outputs
Researchain Logo
Decentralizing Knowledge