1
|
Hegde S, Akbar H, Wellendorf AM, Nestheide S, Johnson JF, Zhao X, Setchell KD, Zheng Y, Cancelas JA. Inhibition of RHOA activity preserves the survival and hemostasis function of long-term cold-stored platelets. Blood 2024; 144:1732-1746. [PMID: 39088777 PMCID: PMC11830982 DOI: 10.1182/blood.2023021453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 05/30/2024] [Accepted: 06/28/2024] [Indexed: 08/03/2024] Open
Abstract
ABSTRACT Patients with thrombocytopenia require platelet transfusion to prevent and stop hemorrhage. Cold storage of platelets results in complex molecular lesions, including changes in membrane microdomains that are recognized by host macrophages and hepatocyte counter-receptors, resulting in phagocytosis and clearance upon transfusion. For this reason, platelets are stored at room temperature, a method that confers increased risk of bacterial contamination. By applying signaling analysis and genetic and pharmacological approaches, we identified that cold-induced activation of RAS homolog family, member A (RHOA) GTPase causes the major hallmarks of platelet cold storage lesions. RHOA deficiency renders murine platelets insensitive to cold storage-induced damage, and pharmacological inhibition by a RHOA activation inhibitor, R-G04, can prevent the cold storage-induced lesions. RHOA inhibition prevents myosin activation and clathrin-independent formation and internalization of lipid rafts enriched in active glycosyltransferases as well as abnormal distribution of GPIbα. RHOA inhibition further prevents the metabolic reprogramming of cold storage-induced lesions and allows the maintenance of glycolytic flux and mitochondria-dependent respiration. Importantly, human platelets transfused in mice after cold storage, in the presence of R-G04 or its more potent enantiomer S-G04, can circulate in vivo at similar levels as room temperature-stored platelets while retaining their hemostatic activity in vivo, as assessed by bleeding time correction in aspirin-treated mice. Our studies provide a mechanism-based translational approach to prevent cold storage-induced damage, which is useful for human platelet transfusion in patients with thrombocytopenia.
Collapse
Affiliation(s)
- Shailaja Hegde
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Huzoor Akbar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH
| | - Ashley M. Wellendorf
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Shawnagay Nestheide
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - James F. Johnson
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Xueheng Zhao
- Division of Pathology and Laboratory Medicine, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Kenneth D. Setchell
- Division of Pathology and Laboratory Medicine, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Jose A. Cancelas
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH
- Connell and O’Reilly Families Cell Manipulation Core Facility, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
2
|
O'Donoghue L, Smolenski A. Roles of G proteins and their GTPase-activating proteins in platelets. Biosci Rep 2024; 44:BSR20231420. [PMID: 38808367 PMCID: PMC11139668 DOI: 10.1042/bsr20231420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Platelets are small anucleate blood cells supporting vascular function. They circulate in a quiescent state monitoring the vasculature for injuries. Platelets adhere to injury sites and can be rapidly activated to secrete granules and to form platelet/platelet aggregates. These responses are controlled by signalling networks that include G proteins and their regulatory guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). Recent proteomics studies have revealed the complete spectrum of G proteins, GEFs, and GAPs present in platelets. Some of these proteins are specific for platelets and very few have been characterised in detail. GEFs and GAPs play a major role in setting local levels of active GTP-bound G proteins in response to activating and inhibitory signals encountered by platelets. Thus, GEFs and GAPs are highly regulated themselves and appear to integrate G protein regulation with other cellular processes. This review focuses on GAPs of small G proteins of the Arf, Rab, Ras, and Rho families, as well as of heterotrimeric G proteins found in platelets.
Collapse
Affiliation(s)
- Lorna O'Donoghue
- UCD School of Medicine, University College Dublin, UCD Conway Institute, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green 123, Dublin 2, Ireland
| | - Albert Smolenski
- UCD School of Medicine, University College Dublin, UCD Conway Institute, Belfield, Dublin 4, Ireland
- Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green 123, Dublin 2, Ireland
| |
Collapse
|
3
|
Fernández-Infante C, Hernández-Cano L, Herranz Ó, Berrocal P, Sicilia-Navarro C, González-Porras JR, Bastida JM, Porras A, Guerrero C. Platelet C3G: a key player in vesicle exocytosis, spreading and clot retraction. Cell Mol Life Sci 2024; 81:84. [PMID: 38345631 PMCID: PMC10861696 DOI: 10.1007/s00018-023-05109-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 02/15/2024]
Abstract
C3G is a Rap1 GEF that plays a pivotal role in platelet-mediated processes such as angiogenesis, tumor growth, and metastasis by modulating the platelet secretome. Here, we explore the mechanisms through which C3G governs platelet secretion. For this, we utilized animal models featuring either overexpression or deletion of C3G in platelets, as well as PC12 cell clones expressing C3G mutants. We found that C3G specifically regulates α-granule secretion via PKCδ, but it does not affect δ-granules or lysosomes. C3G activated RalA through a GEF-dependent mechanism, facilitating vesicle docking, while interfering with the formation of the trans-SNARE complex, thereby restricting vesicle fusion. Furthermore, C3G promotes the formation of lamellipodia during platelet spreading on specific substrates by enhancing actin polymerization via Src and Rac1-Arp2/3 pathways, but not Rap1. Consequently, C3G deletion in platelets favored kiss-and-run exocytosis. C3G also controlled granule secretion in PC12 cells, including pore formation. Additionally, C3G-deficient platelets exhibited reduced phosphatidylserine exposure, resulting in decreased thrombin generation, which along with defective actin polymerization and spreading, led to impaired clot retraction. In summary, platelet C3G plays a dual role by facilitating platelet spreading and clot retraction through the promotion of outside-in signaling while concurrently downregulating α-granule secretion by restricting granule fusion.
Collapse
Affiliation(s)
- Cristina Fernández-Infante
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC, Centro de Investigación del Cáncer, Campus Unamuno S/N, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Luis Hernández-Cano
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC, Centro de Investigación del Cáncer, Campus Unamuno S/N, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Óscar Herranz
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC, Centro de Investigación del Cáncer, Campus Unamuno S/N, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Pablo Berrocal
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC, Centro de Investigación del Cáncer, Campus Unamuno S/N, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Carmen Sicilia-Navarro
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC, Centro de Investigación del Cáncer, Campus Unamuno S/N, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - José Ramón González-Porras
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - José María Bastida
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Almudena Porras
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Ciudad Universitaria, Madrid, Spain.
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - Carmen Guerrero
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC, Centro de Investigación del Cáncer, Campus Unamuno S/N, Salamanca, Spain.
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain.
| |
Collapse
|
4
|
Thanasegaran S, Daimon E, Shibukawa Y, Yamazaki N, Okamoto N. Modelling Takenouchi-Kosaki syndrome using disease-specific iPSCs. Stem Cell Res 2023; 73:103221. [PMID: 37918315 DOI: 10.1016/j.scr.2023.103221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Takenouchi-Kosaki Syndrome (TKS) is a congenital multi-organ disorder caused by the de novo missense mutation c.191A > G p. Tyr64Cys (Y64C) in the CDC42 gene. We previously elucidated the functional abnormalities and thrombopoietic effects of Y64C using HEK293 and MEG01 cells. In the present study, we used iPSCs derived from TKS patients to model the disease and successfully recapitulated macrothrombocytopenia, a prominent TKS phenotype. The megakaryopoietic differentiation potential of TKS-iPSCs and platelet production capacity were examined using an efficient platelet production method redesigned from existing protocols. The results obtained showed that TKS-iPSCs produced fewer hematopoietic progenitor cells, exhibited defective megakaryopoiesis, and released platelets with an abnormally low count and giant morphology. We herein report the first analysis of TKS-iPSC-derived megakaryocytes and platelets, and currently utilize this model to perform drug evaluations for TKS. Therefore, our simple yet effective differentiation method, which mimics the disease in a dish, is a feasible strategy for studying hematopoiesis and related diseases.
Collapse
Affiliation(s)
- Suganya Thanasegaran
- Department of Molecular Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Etsuko Daimon
- Department of Molecular Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Yukinao Shibukawa
- Department of Molecular Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Natsuko Yamazaki
- Department of Molecular Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Nobuhiko Okamoto
- Department of Molecular Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan.
| |
Collapse
|
5
|
Structure-Activity Relationship Analysis of Rhosin, a RhoA GTPase Inhibitor, Reveals a New Class of Antiplatelet Agents. Int J Mol Sci 2023; 24:ijms24044167. [PMID: 36835579 PMCID: PMC9961652 DOI: 10.3390/ijms24044167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Current antiplatelet therapies have several clinical complications and are mostly irreversible in terms of suppressing platelet activity; hence, there is a need to develop improved therapeutic agents. Previous studies have implicated RhoA in platelet activation. Here, we further characterized the lead RhoA inhibitor, Rhosin/G04, in platelet function and present structure-activity relationship (SAR) analysis. A screening for Rhosin/G04 analogs in our chemical library by similarity and substructure searches revealed compounds that showed enhanced antiplatelet activity and suppressed RhoA activity and signaling. A screening for Rhosin/G04 analogs in our chemical library using similarity and substructure searches revealed compounds that showed enhanced antiplatelet activity and suppressed RhoA activity and signaling. SAR analysis revealed that the active compounds have a quinoline group optimally attached to the hydrazine at the 4-position and halogen substituents at the 7- or 8-position. Having indole, methylphenyl, or dichloro-phenyl substituents led to better potency. Rhosin/G04 contains a pair of enantiomers, and S-G04 is significantly more potent than R-G04 in inhibiting RhoA activation and platelet aggregation. Furthermore, the inhibitory effect is reversible, and S-G04 is capable of inhibiting diverse-agonist-stimulated platelet activation. This study identified a new generation of small-molecule RhoA inhibitors, including an enantiomer capable of broadly and reversibly modulating platelet activity.
Collapse
|
6
|
Dandamudi A, Akbar H, Cancelas J, Zheng Y. Rho GTPase Signaling in Platelet Regulation and Implication for Antiplatelet Therapies. Int J Mol Sci 2023; 24:ijms24032519. [PMID: 36768837 PMCID: PMC9917354 DOI: 10.3390/ijms24032519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 01/31/2023] Open
Abstract
Platelets play a vital role in regulating hemostasis and thrombosis. Rho GTPases are well known as molecular switches that control various cellular functions via a balanced GTP-binding/GTP-hydrolysis cycle and signaling cascade through downstream effectors. In platelets, Rho GTPases function as critical regulators by mediating signal transduction that drives platelet activation and aggregation. Mostly by gene targeting and pharmacological inhibition approaches, Rho GTPase family members RhoA, Rac1, and Cdc42 have been shown to be indispensable in regulating the actin cytoskeleton dynamics in platelets, affecting platelet shape change, spreading, secretion, and aggregation, leading to thrombus formation. Additionally, studies of Rho GTPase function using platelets as a non-transformed model due to their anucleated nature have revealed valuable information on cell signaling principles. This review provides an updated summary of recent advances in Rho GTPase signaling in platelet regulation. We also highlight pharmacological approaches that effectively inhibited platelet activation to explore their possible development into future antiplatelet therapies.
Collapse
Affiliation(s)
- Akhila Dandamudi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Department of Pathology, University of Cincinnati Graduate School, Cincinnati, OH 45267, USA
| | - Huzoor Akbar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Jose Cancelas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Hoxworth Blood Center, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Department of Pathology, University of Cincinnati Graduate School, Cincinnati, OH 45267, USA
- Correspondence: ; Tel.: +1-513-636-0595
| |
Collapse
|
7
|
Wright JR, Jones S, Parvathy S, Kaczmarek LK, Forsythe I, Farndale RW, Gibbins JM, Mahaut-Smith MP. The voltage-gated K + channel Kv1.3 modulates platelet motility and α 2β 1 integrin-dependent adhesion to collagen. Platelets 2022; 33:451-461. [PMID: 34348571 PMCID: PMC8935947 DOI: 10.1080/09537104.2021.1942818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/03/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022]
Abstract
Kv1.3 is a voltage-gated K+-selective channel with roles in immunity, insulin-sensitivity, neuronal excitability and olfaction. Despite being one of the largest ionic conductances of the platelet surface membrane, its contribution to platelet function is poorly understood. Here we show that Kv1.3-deficient platelets display enhanced ADP-evoked platelet aggregation and secretion, and an increased surface expression of platelet integrin αIIb. In contrast, platelet adhesion and thrombus formation in vitro under arterial shear conditions on surfaces coated with collagen were reduced for samples from Kv1.3-/- compared to wild type mice. Use of collagen-mimetic peptides revealed a specific defect in the engagement with α2β1. Kv1.3-/- platelets developed significantly fewer, and shorter, filopodia than wild type platelets during adhesion to collagen fibrils. Kv1.3-/- mice displayed no significant difference in thrombus formation within cremaster muscle arterioles using a laser-induced injury model, thus other pro-thrombotic pathways compensate in vivo for the adhesion defect observed in vitro. This may include the increased platelet counts of Kv1.3-/- mice, due in part to a prolonged lifespan. The ability of Kv1.3 to modulate integrin-dependent platelet adhesion has important implications for understanding its contribution to normal physiological platelet function in addition to its reported roles in auto-immune diseases and thromboinflammatory models of stroke.
Collapse
Affiliation(s)
- Joy R Wright
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Sarah Jones
- Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Sasikumar Parvathy
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Leonard K Kaczmarek
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, USA
| | - Ian Forsythe
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | | | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | | |
Collapse
|
8
|
Duan X, Perveen R, Dandamudi A, Adili R, Johnson J, Funk K, Berryman M, Davis AK, Holinstat M, Zheng Y, Akbar H. Pharmacologic targeting of Cdc42 GTPase by a small molecule Cdc42 activity-specific inhibitor prevents platelet activation and thrombosis. Sci Rep 2021; 11:13170. [PMID: 34162972 PMCID: PMC8222210 DOI: 10.1038/s41598-021-92654-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 05/27/2021] [Indexed: 01/14/2023] Open
Abstract
Gene targeting of Cdc42 GTPase has been shown to inhibit platelet activation. In this study, we investigated a hypothesis that inhibition of Cdc42 activity by CASIN, a small molecule Cdc42 Activity-Specific INhibitor, may down regulate platelet activation and thrombus formation. We investigated the effects of CASIN on platelet activation in vitro and thrombosis in vivo. In human platelets, CASIN, but not its inactive analog Pirl7, blocked collagen induced activation of Cdc42 and inhibited phosphorylation of its downstream effector, PAK1/2. Moreover, addition of CASIN to washed human platelets inhibited platelet spreading on immobilized fibrinogen. Treatment of human platelets with CASIN inhibited collagen or thrombin induced: (a) ATP secretion and platelet aggregation; and (b) phosphorylation of Akt, ERK and p38-MAPK. Pre-incubation of platelets with Pirl7, an inactive analog of CASIN, failed to inhibit collagen induced aggregation. Washing of human platelets after incubation with CASIN eliminated its inhibitory effect on collagen induced aggregation. Intraperitoneal administration of CASIN to wild type mice inhibited ex vivo aggregation induced by collagen but did not affect the murine tail bleeding times. CASIN administration, prior to laser-induced injury in murine cremaster muscle arterioles, resulted in formation of smaller and unstable thrombi compared to control mice without CASIN treatment. These data suggest that pharmacologic targeting of Cdc42 by specific and reversible inhibitors may lead to the discovery of novel antithrombotic agents.
Collapse
Affiliation(s)
- Xin Duan
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Rehana Perveen
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Akhila Dandamudi
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Reheman Adili
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - James Johnson
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Kevin Funk
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Mark Berryman
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Ashley Kuenzi Davis
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA.
| | - Huzoor Akbar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
9
|
Garcia A, Dunoyer-Geindre S, Nolli S, Strassel C, Reny JL, Fontana P. miR-204-5p and Platelet Function Regulation: Insight into a Mechanism Mediated by CDC42 and GPIIbIIIa. Thromb Haemost 2021; 121:1206-1219. [PMID: 33940656 PMCID: PMC8421094 DOI: 10.1055/a-1497-9649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Several platelet-derived microRNAs are associated with platelet reactivity (PR) and clinical outcome in cardiovascular patients. We previously showed an association between miR-204-5p and PR in stable cardiovascular patients, but data on functional mechanisms are lacking. AIMS To validate miR-204-5p as a regulator of PR in platelet-like structures (PLS) derived from human megakaryocytes and to address mechanistic issues. METHODS Human hematopoietic stem cells were differentiated into megakaryocytes, enabling the transfection of miR-204-5p and the recovery of subsequent PLS. The morphology of transfected megakaryocytes and PLS was characterized using flow cytometry and microscopy. The functional impact of miR-204-5p was assessed using a flow assay, the quantification of the activated form of the GPIIbIIIa receptor, and a fibrinogen-binding assay. Quantitative polymerase chain reaction and western blot were used to evaluate the impact of miR-204-5p on a validated target, CDC42. The impact of CDC42 modulation was investigated using a silencing strategy. RESULTS miR-204-5p transfection induced cytoskeletal changes in megakaryocytes associated with the retracted protrusion of proPLS, but it had no impact on the number of PLS released. Functional assays showed that the PLS produced by megakaryocytes transfected with miR-204-5p were more reactive than controls. This phenotype is mediated by the regulation of GPIIbIIIa expression, a key contributor in platelet-fibrinogen interaction. Similar results were obtained after CDC42 silencing, suggesting that miR-204-5p regulates PR, at least in part, via CDC42 downregulation. CONCLUSION We functionally validated miR-204-5p as a regulator of the PR that occurs through CDC42 downregulation and regulation of fibrinogen receptor expression.
Collapse
Affiliation(s)
- Alix Garcia
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Séverine Nolli
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Jean-Luc Reny
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of General Internal Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Pierre Fontana
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of Angiology and Haemostasis, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
10
|
Deficiency of ARHGAP21 alters megakaryocytic cell lineage responses and enhances platelet hemostatic function. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119012. [PMID: 33727037 DOI: 10.1016/j.bbamcr.2021.119012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/21/2022]
|
11
|
Palace SG, Vitseva O, Proulx MK, Freedman JE, Goguen JD, Koupenova M. Yersinia pestis escapes entrapment in thrombi by targeting platelet function. J Thromb Haemost 2020; 18:3236-3248. [PMID: 33470041 PMCID: PMC8040536 DOI: 10.1111/jth.15065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/15/2020] [Accepted: 07/24/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Platelets are classically recognized for their role in hemostasis and thrombosis. Recent work has demonstrated that platelets can also execute a variety of immune functions. The dual prothrombotic and immunological roles of platelets suggest that they may pose a barrier to the replication or dissemination of extracellular bacteria. However, some bloodborne pathogens, such as the plague bacterium Yersinia pestis, routinely achieve high vascular titers that are necessary for pathogen transmission. OBJECTIVES It is not currently known how or if pathogens circumvent platelet barriers to bacterial dissemination and replication. We sought to determine whether extracellular bloodborne bacterial pathogens actively interfere with platelet function, using Y pestis as a model system. METHODS The interactions and morphological changes of human platelets with various genetically modified Y pestis strains were examined using aggregation assays, immunofluorescence, and scanning electron microscopy. RESULTS Yersinia pestis directly destabilized platelet thrombi, preventing bacterial entrapment in fibrin/platelet clots. This activity was dependent on two well-characterized bacterial virulence factors: the Y pestis plasminogen activator Pla, which stimulates host-mediated fibrinolysis, and the bacterial type III secretion system (T3SS), which delivers bacterial proteins into the cytoplasm of targeted host cells to reduce or prevent effective immunological responses. Platelets intoxicated by the Y pestis T3SS were unable to respond to prothrombotic stimuli, and T3SS expression decreased the formation of neutrophil extracellular traps in platelet thrombi. CONCLUSIONS These findings are the first demonstration of a bacterial pathogen using its T3SS and an endogenous protease to manipulate platelet function and to escape entrapment in platelet thrombi.
Collapse
Affiliation(s)
- Samantha G. Palace
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Olga Vitseva
- Department of Medicine, Division of Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Megan K. Proulx
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jane E. Freedman
- Department of Medicine, Division of Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jon D. Goguen
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Milka Koupenova
- Department of Medicine, Division of Cardiovascular Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
12
|
Hegde S, Wellendorf AM, Zheng Y, Cancelas JA. Antioxidant prevents clearance of hemostatically competent platelets after long-term cold storage. Transfusion 2020; 61:557-567. [PMID: 33247486 DOI: 10.1111/trf.16200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/27/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cold storage of platelets (PLTs) has the potential advantage of prolonging storage time while reducing posttransfusion infection given the decreased likelihood of bacterial outgrowth during storage and possibly beneficial effects in treating bleeding patients. However, cold storage reduces PLT survival through the induction of complex storage lesions, which are more accentuated when storage is prolonged. STUDY DESIGN AND METHODS Whole blood-derived PLT-rich plasma concentrates from seven PLT pools (n = 5 donors per pool). PLT additive solution was added (67%/33% plasma) and the product was split into 50-mL bags. Split units were stored in the presence or absence of 1 mM of N-acetylcysteine (NAC) under agitation for up to 14 days at room temperature or in the cold and were analyzed for PLT activation, fibrinogen-dependent spreading, microparticle formation, mitochondrial respiratory activity, reactive oxygen species (ROS) generation, as well as in vivo survival and bleeding time correction in immunodeficient mice. RESULTS Cold storage of PLTs for 7 days or longer induces significant PLT activation, cytoskeletal damage, impaired fibrinogen spreading, enhances mitochondrial metabolic decoupling and ROS generation, and increases macrophage-dependent phagocytosis and macrophage-independent clearance. Addition of NAC prevents PLT clearance and allows a correction of the prolonged bleeding time in thrombocytopenic, aspirin-treated, immunodeficient mice. CONCLUSIONS Long-term cold storage induces mitochondrial uncoupling and increased proton leak and ROS generation. The resulting ROS is a crucial contributor to the increased macrophage-dependent and -independent clearance of functional PLTs and can be prevented by the antioxidant NAC in a magnesium-containing additive solution.
Collapse
Affiliation(s)
- Shailaja Hegde
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ashley M Wellendorf
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jose A Cancelas
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
13
|
Brait VH, Miró-Mur F, Pérez-de-Puig I, Notario L, Hurtado B, Pedragosa J, Gallizioli M, Jiménez-Altayó F, Arbaizar-Rovirosa M, Otxoa-de-Amezaga A, Monteagudo J, Ferrer-Ferrer M, de la Rosa X, Bonfill-Teixidor E, Salas-Perdomo A, Hernández-Vidal A, Garcia-de-Frutos P, Lauzurica P, Planas AM. CD69 Plays a Beneficial Role in Ischemic Stroke by Dampening Endothelial Activation. Circ Res 2019; 124:279-291. [PMID: 30582456 DOI: 10.1161/circresaha.118.313818] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RATIONALE CD69 is an immunomodulatory molecule induced during lymphocyte activation. Following stroke, T-lymphocytes upregulate CD69 but its function is unknown. OBJECTIVE We investigated whether CD69 was involved in brain damage following an ischemic stroke. METHODS AND RESULTS We used adult male mice on the C57BL/6 or BALB/c backgrounds, including wild-type mice and CD69-/- mice, and CD69+/+ and CD69-/- lymphocyte-deficient Rag2-/- mice, and generated chimeric mice. We induced ischemia by transient or permanent middle cerebral artery occlusion. We measured infarct volume, assessed neurological function, and studied CD69 expression, as well as platelet function, fibrin(ogen) deposition, and VWF (von Willebrand factor) expression in brain vessels and VWF content and activity in plasma, and performed the tail-vein bleeding test and the carotid artery ferric chloride-induced thrombosis model. We also performed primary glial cell cultures and sorted brain CD45-CD11b-CD31+ endothelial cells for mRNA expression studies. We blocked VWF by intravenous administration of anti-VWF antibodies. CD69-/- mice showed larger infarct volumes and worse neurological deficits than the wild-type mice after ischemia. This worsening effect was not attributable to lymphocytes or other hematopoietic cells. CD69 deficiency lowered the time to thrombosis in the carotid artery despite platelet function not being affected. Ischemia upregulated Cd69 mRNA expression in brain endothelial cells. CD69-deficiency increased fibrin(ogen) accumulation in the ischemic tissue, and plasma VWF content and activity, and VWF expression in brain vessels. Blocking VWF reduced infarct volume and reverted the detrimental effect of CD69-/- deficiency. CONCLUSIONS CD69 deficiency promotes a prothrombotic phenotype characterized by increased VWF and worse brain damage after ischemic stroke. The results suggest that CD69 acts as a downregulator of endothelial activation.
Collapse
Affiliation(s)
- Vanessa H Brait
- From the Department of Brain Ischemia and Neurodegeneration (V.H.B., F.M.-M., I.P.-d.-P., J.P., M.G., M.A.-R., A.O.-d.-A., X.d.l.R., E.B.-T., A.M.P.), Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (V.H.B., F.M.-M., J.P., M.G., M.A.-R., A.O.-d.-A., M.F.-F., E.B.-T., A.S.-P., A.H.-V., A.M.P.)
| | - Francesc Miró-Mur
- From the Department of Brain Ischemia and Neurodegeneration (V.H.B., F.M.-M., I.P.-d.-P., J.P., M.G., M.A.-R., A.O.-d.-A., X.d.l.R., E.B.-T., A.M.P.), Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (V.H.B., F.M.-M., J.P., M.G., M.A.-R., A.O.-d.-A., M.F.-F., E.B.-T., A.S.-P., A.H.-V., A.M.P.)
| | - Isabel Pérez-de-Puig
- From the Department of Brain Ischemia and Neurodegeneration (V.H.B., F.M.-M., I.P.-d.-P., J.P., M.G., M.A.-R., A.O.-d.-A., X.d.l.R., E.B.-T., A.M.P.), Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Spain
| | - Laura Notario
- Grupo de Activación Inmunológica, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Madrid, Spain (L.N., P.L.)
| | - Begoña Hurtado
- Department of Cell Death and Proliferation (B.H., P.G.-d.-F.), Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Spain
| | - Jordi Pedragosa
- From the Department of Brain Ischemia and Neurodegeneration (V.H.B., F.M.-M., I.P.-d.-P., J.P., M.G., M.A.-R., A.O.-d.-A., X.d.l.R., E.B.-T., A.M.P.), Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (V.H.B., F.M.-M., J.P., M.G., M.A.-R., A.O.-d.-A., M.F.-F., E.B.-T., A.S.-P., A.H.-V., A.M.P.)
| | - Mattia Gallizioli
- From the Department of Brain Ischemia and Neurodegeneration (V.H.B., F.M.-M., I.P.-d.-P., J.P., M.G., M.A.-R., A.O.-d.-A., X.d.l.R., E.B.-T., A.M.P.), Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (V.H.B., F.M.-M., J.P., M.G., M.A.-R., A.O.-d.-A., M.F.-F., E.B.-T., A.S.-P., A.H.-V., A.M.P.)
| | - Francesc Jiménez-Altayó
- Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain (F.J.A.)
| | - Maria Arbaizar-Rovirosa
- From the Department of Brain Ischemia and Neurodegeneration (V.H.B., F.M.-M., I.P.-d.-P., J.P., M.G., M.A.-R., A.O.-d.-A., X.d.l.R., E.B.-T., A.M.P.), Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (V.H.B., F.M.-M., J.P., M.G., M.A.-R., A.O.-d.-A., M.F.-F., E.B.-T., A.S.-P., A.H.-V., A.M.P.)
| | - Amaia Otxoa-de-Amezaga
- From the Department of Brain Ischemia and Neurodegeneration (V.H.B., F.M.-M., I.P.-d.-P., J.P., M.G., M.A.-R., A.O.-d.-A., X.d.l.R., E.B.-T., A.M.P.), Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (V.H.B., F.M.-M., J.P., M.G., M.A.-R., A.O.-d.-A., M.F.-F., E.B.-T., A.S.-P., A.H.-V., A.M.P.)
| | - Juan Monteagudo
- Hemotherapy and Haemostasis Service, Hospital Clinic, Barcelona, Spain (J.M.)
| | - Maura Ferrer-Ferrer
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (V.H.B., F.M.-M., J.P., M.G., M.A.-R., A.O.-d.-A., M.F.-F., E.B.-T., A.S.-P., A.H.-V., A.M.P.)
| | - Xavier de la Rosa
- From the Department of Brain Ischemia and Neurodegeneration (V.H.B., F.M.-M., I.P.-d.-P., J.P., M.G., M.A.-R., A.O.-d.-A., X.d.l.R., E.B.-T., A.M.P.), Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Spain
| | - Ester Bonfill-Teixidor
- From the Department of Brain Ischemia and Neurodegeneration (V.H.B., F.M.-M., I.P.-d.-P., J.P., M.G., M.A.-R., A.O.-d.-A., X.d.l.R., E.B.-T., A.M.P.), Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (V.H.B., F.M.-M., J.P., M.G., M.A.-R., A.O.-d.-A., M.F.-F., E.B.-T., A.S.-P., A.H.-V., A.M.P.)
| | - Angélica Salas-Perdomo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (V.H.B., F.M.-M., J.P., M.G., M.A.-R., A.O.-d.-A., M.F.-F., E.B.-T., A.S.-P., A.H.-V., A.M.P.)
| | - Alba Hernández-Vidal
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (V.H.B., F.M.-M., J.P., M.G., M.A.-R., A.O.-d.-A., M.F.-F., E.B.-T., A.S.-P., A.H.-V., A.M.P.)
| | - Pablo Garcia-de-Frutos
- Department of Cell Death and Proliferation (B.H., P.G.-d.-F.), Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Spain
| | - Pilar Lauzurica
- Grupo de Activación Inmunológica, Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Madrid, Spain (L.N., P.L.)
| | - Anna M Planas
- From the Department of Brain Ischemia and Neurodegeneration (V.H.B., F.M.-M., I.P.-d.-P., J.P., M.G., M.A.-R., A.O.-d.-A., X.d.l.R., E.B.-T., A.M.P.), Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (V.H.B., F.M.-M., J.P., M.G., M.A.-R., A.O.-d.-A., M.F.-F., E.B.-T., A.S.-P., A.H.-V., A.M.P.)
| |
Collapse
|
14
|
Abstract
The vasculature is a dynamic environment in which blood platelets constantly survey the endothelium for sites of vessel damage. The formation of a mechanically coherent hemostatic plug to prevent blood loss relies on a coordinated series of ligand-receptor interactions governing the recruitment, activation, and aggregation of platelets. The physical biology of each step is distinct in that the recruitment of platelets depends on the mechanosensing of the platelet receptor glycoprotein Ib for the adhesive protein von Willebrand factor, whereas platelet activation and aggregation are responsive to the mechanical forces sensed at adhesive junctions between platelets and at the platelet-matrix interface. Herein we take a biophysical perspective to discuss the current understanding of platelet mechanotransduction as well as the measurement techniques used to quantify the physical biology of platelets in the context of thrombus formation under flow.
Collapse
Affiliation(s)
- Caroline E Hansen
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta/Emory University School of Medicine, Atlanta, Georgia 30332, USA; .,Wallace H. Coulter Department of Biomedical Engineering and Institute for Electronics and Nanotechnology, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| | - Yongzhi Qiu
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta/Emory University School of Medicine, Atlanta, Georgia 30332, USA; .,Wallace H. Coulter Department of Biomedical Engineering and Institute for Electronics and Nanotechnology, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| | - Owen J T McCarty
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon 97239, USA.,Division of Hematology and Medical Oncology and Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Wilbur A Lam
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children's Healthcare of Atlanta/Emory University School of Medicine, Atlanta, Georgia 30332, USA; .,Wallace H. Coulter Department of Biomedical Engineering and Institute for Electronics and Nanotechnology, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30332, USA
| |
Collapse
|
15
|
Senini V, Amara U, Paul M, Kim H. Porphyromonas gingivalis lipopolysaccharide activates platelet Cdc42 and promotes platelet spreading and thrombosis. J Periodontol 2019; 90:1336-1345. [PMID: 31106406 DOI: 10.1002/jper.18-0596] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND Periodontitis confers an increased risk for cardiovascular diseases, including thrombosis. However, the molecular mechanisms that potentially link periodontitis with thrombosis are undefined. Here we test the hypothesis that Gram-negative periodontal infection promotes pathological platelet activation and amplifies shape change. We focus specifically on lipopolysaccharide (LPS) signaling to platelets. METHODS Platelets were isolated from blood samples and allowed to spread on coverslips in the presence or absence of LPS purified from the periodontal pathogen Porphyromonas gingivalis. Platelets were fixed and stained with Alexa-488-phalloidin to label the actin cytoskeleton. The degree of platelet spreading and shape change was quantified by confocal microscopy. In a translational pilot study, blood samples were obtained from human subjects exhibiting generalized severe periodontitis (SP) or healthy periodontium (HP). Rotational thromboelastometry was used to quantify the rate of clot formation via the intrinsic coagulation pathway. RESULTS LPS-treated platelets exhibited significantly (P < 0.05) greater spreading and higher numbers of actin-rich filopodia (cell extensions) than controls. We also found that LPS stimulation of platelets promoted the activation of Cdc42, the small GTPase responsible for filopodia formation. Exposure of whole blood samples to LPS significantly (P < 0.05) reduced clotting times. Blood from SP patients clotted significantly (P < 0.05) more rapidly and exhibited shorter partial thromboplastin times compared with HP controls. CONCLUSIONS This is the first study to suggest a mechanism by which LPS stimulation drives Cdc42 activation and platelet spreading. These data are consistent with the notion that periodontitis promotes accelerated clot formation and an increased risk of thrombosis.
Collapse
Affiliation(s)
- Vincent Senini
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,Faculty of Dentistry, University of British Columbia, Vancouver, BC, Canada
| | - Umme Amara
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,Faculty of Dentistry, University of British Columbia, Vancouver, BC, Canada
| | - Manoj Paul
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,Faculty of Dentistry, University of British Columbia, Vancouver, BC, Canada
| | - Hugh Kim
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada.,Faculty of Dentistry, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
16
|
Huang J, Li X, Shi X, Zhu M, Wang J, Huang S, Huang X, Wang H, Li L, Deng H, Zhou Y, Mao J, Long Z, Ma Z, Ye W, Pan J, Xi X, Jin J. Platelet integrin αIIbβ3: signal transduction, regulation, and its therapeutic targeting. J Hematol Oncol 2019; 12:26. [PMID: 30845955 PMCID: PMC6407232 DOI: 10.1186/s13045-019-0709-6] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 02/21/2019] [Indexed: 12/18/2022] Open
Abstract
Integrins are a family of transmembrane glycoprotein signaling receptors that can transmit bioinformation bidirectionally across the plasma membrane. Integrin αIIbβ3 is expressed at a high level in platelets and their progenitors, where it plays a central role in platelet functions, hemostasis, and arterial thrombosis. Integrin αIIbβ3 also participates in cancer progression, such as tumor cell proliferation and metastasis. In resting platelets, integrin αIIbβ3 adopts an inactive conformation. Upon agonist stimulation, the transduction of inside-out signals leads integrin αIIbβ3 to switch from a low- to high-affinity state for fibrinogen and other ligands. Ligand binding causes integrin clustering and subsequently promotes outside-in signaling, which initiates and amplifies a range of cellular events to drive essential platelet functions such as spreading, aggregation, clot retraction, and thrombus consolidation. Regulation of the bidirectional signaling of integrin αIIbβ3 requires the involvement of numerous interacting proteins, which associate with the cytoplasmic tails of αIIbβ3 in particular. Integrin αIIbβ3 and its signaling pathways are considered promising targets for antithrombotic therapy. This review describes the bidirectional signal transduction of integrin αIIbβ3 in platelets, as well as the proteins responsible for its regulation and therapeutic agents that target integrin αIIbβ3 and its signaling pathways.
Collapse
Affiliation(s)
- Jiansong Huang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xia Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaofeng Shi
- Department of Hematology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mark Zhu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jinghan Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shujuan Huang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xin Huang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huafeng Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, 91010, USA
| | - Ling Li
- Department of Hematological Malignancies Translational Science, Gehr Family Center for Leukemia Research, Hematologic Malignancies and Stem Cell Transplantation Institute, Beckman Research Institute, City of Hope Medical Center, Duarte, CA, 91010, USA
| | - Huan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yulan Zhou
- Department of Hematology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jianhua Mao
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Sino-French Research Centre for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhangbiao Long
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhixin Ma
- Clinical Prenatal Diagnosis Center, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenle Ye
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiajia Pan
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaodong Xi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Collaborative Innovation Center of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Sino-French Research Centre for Life Sciences and Genomics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China. .,Key Laboratory of Hematologic Malignancies, Diagnosis and Treatment, Hangzhou, Zhejiang, China. .,Institute of Hematology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
17
|
Alterations in Platelet Alpha-Granule Secretion and Adhesion on Collagen under Flow in Mice Lacking the Atypical Rho GTPase RhoBTB3. Cells 2019; 8:cells8020149. [PMID: 30754723 PMCID: PMC6406922 DOI: 10.3390/cells8020149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/31/2019] [Accepted: 02/07/2019] [Indexed: 12/23/2022] Open
Abstract
Typical Rho GTPases, such as Rac1, Cdc42, and RhoA, act as molecular switches regulating various aspects of platelet cytoskeleton reorganization. The loss of these enzymes results in reduced platelet functionality. Atypical Rho GTPases of the RhoBTB subfamily are characterized by divergent domain architecture. One family member, RhoBTB3, is expressed in platelets, but its function is unclear. In the present study we examined the role of RhoBTB3 in platelet function using a knockout mouse model. We found the platelet count, size, numbers of both alpha and dense granules, and surface receptor profile in these mice were comparable to wild-type mice. Deletion of Rhobtb3 had no effect on aggregation and dense granule secretion in response to a range of agonists including thrombin, collagen, and adenosine diphosphate (ADP). By contrast, alpha-granule secretion increased in mice lacking RhoBTB3 in response to thrombin, collagen related peptide (CRP) and U46619/ADP. Integrin activation and spreading on fibrinogen and collagen under static conditions were also unimpaired; however, we observed reduced platelet accrual on collagen under flow conditions. These defects did not translate into alterations in tail bleeding time. We conclude that genetic deletion of Rhobtb3 leads to subtle alterations in alpha-granule secretion and adhesion to collagen without significant effects on hemostasis in vivo.
Collapse
|
18
|
Hurtado B, Trakala M, Ximénez-Embún P, El Bakkali A, Partida D, Sanz-Castillo B, Álvarez-Fernández M, Maroto M, Sánchez-Martínez R, Martínez L, Muñoz J, García de Frutos P, Malumbres M. Thrombocytopenia-associated mutations in Ser/Thr kinase MASTL deregulate actin cytoskeletal dynamics in platelets. J Clin Invest 2018; 128:5351-5367. [PMID: 30252678 DOI: 10.1172/jci121876] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 09/18/2018] [Indexed: 12/18/2022] Open
Abstract
MASTL, a Ser/Thr kinase that inhibits PP2A-B55 complexes during mitosis, is mutated in autosomal dominant thrombocytopenia. However, the connections between the cell-cycle machinery and this human disease remain unexplored. We report here that, whereas Mastl ablation in megakaryocytes prevented proper maturation of these cells, mice carrying the thrombocytopenia-associated mutation developed thrombocytopenia as a consequence of aberrant activation and survival of platelets. Activation of mutant platelets was characterized by hyperstabilized pseudopods mimicking the effect of PP2A inhibition and actin polymerization defects. These aberrations were accompanied by abnormal hyperphosphorylation of multiple components of the actin cytoskeleton and were rescued both in vitro and in vivo by inhibiting upstream kinases such as PKA, PKC, or AMPK. These data reveal an unexpected role of Mastl in actin cytoskeletal dynamics in postmitotic cells and suggest that the thrombocytopenia-associated mutation in MASTL is a pathogenic dominant mutation that mimics decreased PP2A activity resulting in altered phosphorylation of cytoskeletal regulatory pathways.
Collapse
Affiliation(s)
- Begoña Hurtado
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Department of Cell Death and Proliferation, Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas- Institut d'Investigacions Biomèdiques August Pi i Sunyer- (IIBB-CSIC-IDIBAPS), Barcelona, Spain
| | - Marianna Trakala
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Pilar Ximénez-Embún
- ProteoRed - Instituto de Salud Carlos III (ISCIII) and Proteomics Unit, CNIO, Madrid, Spain
| | - Aicha El Bakkali
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - David Partida
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Belén Sanz-Castillo
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - María Maroto
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ruth Sánchez-Martínez
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Javier Muñoz
- ProteoRed - Instituto de Salud Carlos III (ISCIII) and Proteomics Unit, CNIO, Madrid, Spain
| | - Pablo García de Frutos
- Department of Cell Death and Proliferation, Institut d'Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas- Institut d'Investigacions Biomèdiques August Pi i Sunyer- (IIBB-CSIC-IDIBAPS), Barcelona, Spain
| | - Marcos Malumbres
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| |
Collapse
|
19
|
Akbar H, Duan X, Piatt R, Saleem S, Davis AK, Tandon NN, Bergmeier W, Zheng Y. Small molecule targeting the Rac1-NOX2 interaction prevents collagen-related peptide and thrombin-induced reactive oxygen species generation and platelet activation. J Thromb Haemost 2018; 16:2083-2096. [PMID: 30007118 PMCID: PMC6472274 DOI: 10.1111/jth.14240] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Indexed: 12/29/2022]
Abstract
Essentials Reactive oxygen species (ROS) generation by NOX2 plays a critical role in platelet activation. Rac1 regulation of NOX2 is important for ROS generation. Small molecule inhibitor of the Rac1-p67phox interaction prevents platelet activation. Pharmacologic targeting of Rac1-NOX2 axis can be a viable approach for antithrombotic therapy. SUMMARY Background Platelets from patients with X-linked chronic granulomatous disease or mice deficient in nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) oxidase isoform NOX2 exhibit diminished reactive oxygen species (ROS) generation and platelet activation. Binding of Rac1 GTPase to p67phox plays a critical role in NOX2 activation by facilitating the assembly of the NOX2 enzyme complex. Objective We tested the hypothesis that Phox-I, a rationally designed small molecule inhibitor of Rac-p67phox interaction, may serve as an antithrombosis agent by suppressing ROS production and platelet activation. Results Collagen-related peptide (CRP) induced ROS generation in a time-dependent manner. Platelets from Rac1-/- mice or human platelets treated with NSC23766, a specific Rac inhibitor, produced significantly less ROS in response to CRP. Treatment of platelets with Phox-I inhibited diverse CRP-induced responses, including: (i) ROS generation; (ii) release of P-selectin; (iii) secretion of ATP; (iv) platelet aggregation; and (v) phosphorylation of Akt. Similarly, incubation of platelets with Phox-I inhibited thrombin-induced: (i) secretion of ATP; (ii) platelet aggregation; (iii) rise in cytosolic calcium; and (iv) phosphorylation of Akt. In mouse models, intraperitoneal administration of Phox-I inhibited: (i) collagen-induced platelet aggregation without affecting the tail bleeding time and (ii) in vivo platelet adhesion/accumulation at the laser injury sites on the saphenous vein without affecting the time for complete cessation of blood loss. Conclusions Small molecule targeting of the Rac1-p67phox interaction may present an antithrombosis regimen by preventing GPVI- and non-GPVI-mediated NOX2 activation, ROS generation and platelet function without affecting the bleeding time.
Collapse
Affiliation(s)
- H Akbar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - X Duan
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - R Piatt
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - S Saleem
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - A K Davis
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | | | - W Bergmeier
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Y Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
20
|
Pleines I, Cherpokova D, Bender M. Rho GTPases and their downstream effectors in megakaryocyte biology. Platelets 2018; 30:9-16. [DOI: 10.1080/09537104.2018.1478071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Irina Pleines
- Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Deya Cherpokova
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Markus Bender
- Institute of Experimental Biomedicine I, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
21
|
Atkinson L, Yusuf MZ, Aburima A, Ahmed Y, Thomas SG, Naseem KM, Calaminus SDJ. Reversal of stress fibre formation by Nitric Oxide mediated RhoA inhibition leads to reduction in the height of preformed thrombi. Sci Rep 2018; 8:3032. [PMID: 29445102 PMCID: PMC5813033 DOI: 10.1038/s41598-018-21167-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/24/2018] [Indexed: 01/10/2023] Open
Abstract
Evidence has emerged to suggest that thrombi are dynamic structures with distinct areas of differing platelet activation and inhibition. We hypothesised that Nitric oxide (NO), a platelet inhibitor, can modulate the actin cytoskeleton reversing platelet spreading, and therefore reduce the capability of thrombi to withstand a high shear environment. Our data demonstrates that GSNO, DEANONOate, and a PKG-activating cGMP analogue reversed stress fibre formation and increased actin nodule formation in adherent platelets. This effect is sGC dependent and independent of ADP and thromboxanes. Stress fibre formation is a RhoA dependent process and NO induced RhoA inhibition, however, it did not phosphorylate RhoA at ser188 in spread platelets. Interestingly NO and PGI2 synergise to reverse stress fibre formation at physiologically relevant concentrations. Analysis of high shear conditions indicated that platelets activated on fibrinogen, induced stress fibre formation, which was reversed by GSNO treatment. Furthermore, preformed thrombi on collagen post perfused with GSNO had a 30% reduction in thrombus height in comparison to the control. This study demonstrates that NO can reverse key platelet functions after their initial activation and identifies a novel mechanism for controlling excessive thrombosis.
Collapse
Affiliation(s)
- L Atkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - M Z Yusuf
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - A Aburima
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - Y Ahmed
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK
| | - S G Thomas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - K M Naseem
- Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - S D J Calaminus
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, HU6 7RX, UK.
| |
Collapse
|
22
|
Wufuer Y, Shan X, Sailike M, Adilaimu K, Ma S, Wang H. GPVI‑Fc‑PEG improves cerebral infarct volume and cerebral thrombosis in mouse model with cerebral thrombosis. Mol Med Rep 2017; 16:7561-7568. [PMID: 28944903 DOI: 10.3892/mmr.2017.7556] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/07/2017] [Indexed: 11/05/2022] Open
Abstract
Cerebral thrombosis is one of the most common causes of cerebral infarction, and anticoagulation therapy is a routine treatment in patients with hemorrhagic cerebral venous thrombosis. The hemostatic function of platelets is important for the anticoagulation therapy of thrombosis. Glycoprotein VI (GPVI) is reported as the major signaling receptor for collagen and is exclusively expressed on platelets and megakaryocytes, initiating platelet recruitment at sites of vascular injury and demonstrating numerous beneficial effects for patients with cerebral thrombosis. In the present study, thrombus formation and platelet adhesion following endothelial injury was monitored in the jugular vein by intra‑vital fluorescence microscopy. The morphological and clinical observations of cerebral thrombosis were investigated and analyzed in a mouse model with cerebral thrombosis. In addition, the present study investigated the effect of fusion protein GPVI modified with Fc and PEG, which is specifically linked to the extracellular domain of GPVI (GPVI‑Fc‑PEG), on thrombus formation following vessel wall injury and on experimental mice with cerebral thrombosis. The maximum tolerated dose (MTD) was identified as 0.18 mg. GPVI‑Fc‑PEG competitively bound to and prevented von Willebrand Factor‑collagen interactions. The results of the present study demonstrated that cerebral thrombosis was greatly relieved and improved functional outcomes treatment with an MTD of GPVI‑Fc‑PEG following endothelial injury, compared with GPVI‑Fc‑treated mice. In addition, cerebral edema and infarct size was improved compared with GPVI‑Fc‑treated mice with ischemic stroke immediately prior to reperfusion. Furthermore, treatment of GPVI‑Fc‑PEG led to increased reperfusion and improved survival following cerebral thrombosis compared with treatment with either single agent alone. Taken together, GPVI‑Fc‑PEG relieved cerebral thrombosis following ischemic stroke and improved prognostic preclinical outcomes without intracranial bleeding, which suggested that GPVI‑Fc‑PEG may be a potential candidate for cerebral thrombosis therapy.
Collapse
Affiliation(s)
- Yimae Wufuer
- Department of Vascular Thyroid Surgery, Gastrointestinal Vascular Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Xuefeng Shan
- The First Department of Pediatric Surgery, Gastrointestinal Vascular Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Magaoweiya Sailike
- Department of Vascular Thyroid Surgery, Gastrointestinal Vascular Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Kamile Adilaimu
- Department of Vascular Thyroid Surgery, Gastrointestinal Vascular Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Songfeng Ma
- The First Department of Pediatric Surgery, Gastrointestinal Vascular Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Huguo Wang
- Department of Vascular Thyroid Surgery, Gastrointestinal Vascular Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| |
Collapse
|
23
|
Abstract
Integrin αIIbβ3 is a highly abundant heterodimeric platelet receptor that can transmit information bidirectionally across the plasma membrane, and plays a critical role in hemostasis and thrombosis. Upon platelet activation, inside-out signaling pathways increase the affinity of αIIbβ3 for fibrinogen and other ligands. Ligand binding and integrin clustering subsequently stimulate outside-in signaling, which initiates and amplifies a range of cellular events driving essential platelet processes such as spreading, thrombus consolidation, and clot retraction. Integrin αIIbβ3 has served as an excellent model for the study of integrin biology, and it has become clear that integrin outside-in signaling is highly complex and involves a vast array of enzymes, signaling adaptors, and cytoskeletal components. In this review, we provide a concise but comprehensive overview of αIIbβ3 outside-in signaling, focusing on the key players involved, and how they cooperate to orchestrate this critical aspect of platelet biology. We also discuss gaps in the current understanding of αIIbβ3 outside-in signaling and highlight avenues for future investigation.
Collapse
|
24
|
Ngo ATP, Thierheimer MLD, Babur Ö, Rocheleau AD, Huang T, Pang J, Rigg RA, Mitrugno A, Theodorescu D, Burchard J, Nan X, Demir E, McCarty OJT, Aslan JE. Assessment of roles for the Rho-specific guanine nucleotide dissociation inhibitor Ly-GDI in platelet function: a spatial systems approach. Am J Physiol Cell Physiol 2017; 312:C527-C536. [PMID: 28148498 PMCID: PMC5407014 DOI: 10.1152/ajpcell.00274.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 12/29/2022]
Abstract
On activation at sites of vascular injury, platelets undergo morphological alterations essential to hemostasis via cytoskeletal reorganizations driven by the Rho GTPases Rac1, Cdc42, and RhoA. Here we investigate roles for Rho-specific guanine nucleotide dissociation inhibitor proteins (RhoGDIs) in platelet function. We find that platelets express two RhoGDI family members, RhoGDI and Ly-GDI. Whereas RhoGDI localizes throughout platelets in a granule-like manner, Ly-GDI shows an asymmetric, polarized localization that largely overlaps with Rac1 and Cdc42 as well as microtubules and protein kinase C (PKC) in platelets adherent to fibrinogen. Antibody interference and platelet spreading experiments suggest a specific role for Ly-GDI in platelet function. Intracellular signaling studies based on interactome and pathways analyses also support a regulatory role for Ly-GDI, which is phosphorylated at PKC substrate motifs in a PKC-dependent manner in response to the platelet collagen receptor glycoprotein (GP) VI-specific agonist collagen-related peptide. Additionally, PKC inhibition diffuses the polarized organization of Ly-GDI in spread platelets relative to its colocalization with Rac1 and Cdc42. Together, our results suggest a role for Ly-GDI in the localized regulation of Rho GTPases in platelets and hypothesize a link between the PKC and Rho GTPase signaling systems in platelet function.
Collapse
Affiliation(s)
- Anh T P Ngo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Marisa L D Thierheimer
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon.,School of Chemical, Biological and Environmental Engineering, Oregon State University, Corvallis, Oregon; and
| | - Özgün Babur
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon.,Computational Biology Program, Oregon Health & Science University, Portland, Oregon
| | - Anne D Rocheleau
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Tao Huang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Jiaqing Pang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Rachel A Rigg
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Annachiara Mitrugno
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Dan Theodorescu
- Department of Surgery, Department of Pharmacology, and Comprehensive Cancer Center University of Colorado, Aurora, Colorado
| | - Julja Burchard
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon
| | - Xiaolin Nan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Emek Demir
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon.,Computational Biology Program, Oregon Health & Science University, Portland, Oregon
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon.,Division of Hematology & Medical Oncology, Oregon Health & Science University, Portland, Oregon
| | - Joseph E Aslan
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, Oregon;
| |
Collapse
|
25
|
Menter DG, Davis JS, Tucker SC, Hawk E, Crissman JD, Sood AK, Kopetz S, Honn KV. Platelets: “First Responders” in Cancer Progression and Metastasis. PLATELETS IN THROMBOTIC AND NON-THROMBOTIC DISORDERS 2017:1111-1132. [DOI: 10.1007/978-3-319-47462-5_74] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
26
|
Pdlim7 Regulates Arf6-Dependent Actin Dynamics and Is Required for Platelet-Mediated Thrombosis in Mice. PLoS One 2016; 11:e0164042. [PMID: 27792740 PMCID: PMC5085081 DOI: 10.1371/journal.pone.0164042] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/19/2016] [Indexed: 11/19/2022] Open
Abstract
Upon vessel injury, platelets become activated and rapidly reorganize their actin cytoskeleton to adhere to the site of endothelial damage, triggering the formation of a fibrin-rich plug to prevent further blood loss. Inactivation of Pdlim7 provides the new perspective that regulation of actin cytoskeletal changes in platelets is dependent on the encoded PDZ-LIM protein. Loss-of-function of Pdlim7 triggers hypercoagulopathy and causes significant perinatal lethality in mice. Our in vivo and in vitro studies reveal that Pdlim7 is dynamically distributed along actin fibers, and lack of Pdlim7 leads to a marked inability to rearrange the actin cytoskeleton. Specifically, the absence of Pdlim7 prevents platelets from bundling actin fibers into a concentric ring that defines the round spread shape of activated platelets. Similarly, in mouse embryonic fibroblasts, loss of Pdlim7 abolishes the formation of stress fibers needed to adopt the typical elongated fibroblast shape. In addition to revealing a fundamental cell biological role in actin cytoskeletal organization, we also demonstrate a function of Pdlim7 in regulating the cycling between the GTP/GDP-bound states of Arf6. The small GTPase Arf6 is an essential factor required for actin dynamics, cytoskeletal rearrangements, and platelet activation. Consistent with our findings of significantly elevated initial F-actin ratios and subsequent morphological aberrations, loss of Pdlim7 causes a shift in balance towards an increased Arf6-GTP level in resting platelets. These findings identify a new Pdlim7-Arf6 axis controlling actin dynamics and implicate Pdlim7 as a primary endogenous regulator of platelet-dependent hemostasis.
Collapse
|
27
|
Akbar H, Duan X, Saleem S, Davis AK, Zheng Y. RhoA and Rac1 GTPases Differentially Regulate Agonist-Receptor Mediated Reactive Oxygen Species Generation in Platelets. PLoS One 2016; 11:e0163227. [PMID: 27681226 PMCID: PMC5040254 DOI: 10.1371/journal.pone.0163227] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 09/06/2016] [Indexed: 12/19/2022] Open
Abstract
Agonist induced generation of reactive oxygen species (ROS) by NADPH oxidases (NOX) enhances platelet aggregation and hence the risk of thrombosis. RhoA and Rac1 GTPases are involved in ROS generation by NOX in a variety of cells, but their roles in platelet ROS production remain unclear. In this study we used platelets from RhoA and Rac1 conditional knockout mice as well as human platelets treated with Rhosin and NSC23767, rationally designed small molecule inhibitors of RhoA and Rac GTPases, respectively, to better define the contributions of RhoA and Rac1 signaling to ROS generation and platelet activation. Treatment of platelets with Rhosin inhibited: (a) U46619 induced activation of RhoA; (b) phosphorylation of p47phox, a critical component of NOX; (c) U46619 or thrombin induced ROS generation; (d) phosphorylation of myosin light chain (MLC); (e) platelet shape change; (f) platelet spreading on immobilized fibrinogen; and (g) release of P-selectin, secretion of ATP and aggregation. Conditional deletion of RhoA or Rac1 gene inhibited thrombin induced ROS generation in platelets. Addition of Y27632, a RhoA inhibitor, NSC23766 or Phox-I, an inhibitor of Rac1-p67phox interaction, to human platelets blocked thrombin induced ROS generation. These data suggest that: (a) RhoA/ROCK/p47phox signaling axis promotes ROS production that, at least in part, contributes to platelet activation in conjunction with or independent of the RhoA/ROCK mediated phosphorylation of MLC; and (b) RhoA and Rac1 differentially regulate ROS generation by inhibiting phosphorylation of p47phox and Rac1-p67phox interaction, respectively.
Collapse
Affiliation(s)
- Huzoor Akbar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, United States of America
- * E-mail:
| | - Xin Duan
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, United States of America
| | - Saima Saleem
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, United States of America
| | - Ashley K. Davis
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, United States of America
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, United States of America
| |
Collapse
|
28
|
Antkowiak A, Viaud J, Severin S, Zanoun M, Ceccato L, Chicanne G, Strassel C, Eckly A, Leon C, Gachet C, Payrastre B, Gaits-Iacovoni F. Cdc42-dependent F-actin dynamics drive structuration of the demarcation membrane system in megakaryocytes. J Thromb Haemost 2016; 14:1268-84. [PMID: 26991240 DOI: 10.1111/jth.13318] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 03/03/2016] [Indexed: 02/04/2023]
Abstract
UNLABELLED Essentials Information about the formation of the demarcation membrane system (DMS) is still lacking. We investigated the role of the cytoskeleton in DMS structuration in megakaryocytes. Cdc42/Pak-dependent F-actin remodeling regulates DMS organization for proper megakaryopoiesis. These data highlight the mandatory role of F-actin in platelet biogenesis. SUMMARY Background Blood platelet biogenesis results from the maturation of megakaryocytes (MKs), which involves the development of a complex demarcation membrane system (DMS). Therefore, MK differentiation is an attractive model for studying membrane remodeling. Objectives We sought to investigate the mechanism of DMS structuration in relationship to the cytoskeleton. Results Using three-dimensional (3D) confocal imaging, we have identified consecutive stages of DMS organization that rely on F-actin dynamics to polarize membranes and nuclei territories. Interestingly, microtubules are not involved in this process. We found that the mechanism underlying F-actin-dependent DMS formation required the activation of the guanosine triphosphate hydrolase Cdc42 and its p21-activated kinase effectors (Pak1/2/3). Förster resonance energy transfer demonstrated that active Cdc42 was associated with endomembrane dynamics throughout terminal maturation. Inhibition of Cdc42 or Pak1/2/3 severely destructured the DMS and blocked proplatelet formation. Even though this process does not require containment within the hematopoietic niche, because DMS structuration was observed upon thrombopoietin-treatment in suspension, integrin outside-in signaling was required for Pak activation and probably resulted from secretion of extracellular matrix by MKs. Conclusions These data indicate a functional link, mandatory for MK differentiation, between actin dynamics, regulated by Cdc42/Pak1/2/3, and DMS maturation.
Collapse
Affiliation(s)
- A Antkowiak
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - J Viaud
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - S Severin
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - M Zanoun
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - L Ceccato
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - G Chicanne
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| | - C Strassel
- INSERM, UMR_S949, Université de Strasbourg, Etablissement Français du Sang-Alsace, Toulouse, France
| | - A Eckly
- INSERM, UMR_S949, Université de Strasbourg, Etablissement Français du Sang-Alsace, Toulouse, France
| | - C Leon
- INSERM, UMR_S949, Université de Strasbourg, Etablissement Français du Sang-Alsace, Toulouse, France
| | - C Gachet
- INSERM, UMR_S949, Université de Strasbourg, Etablissement Français du Sang-Alsace, Toulouse, France
| | - B Payrastre
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
- Laboratoire d'Hématologie, CHU de Toulouse, Toulouse, France
| | - F Gaits-Iacovoni
- INSERM, UMR1048, Université Toulouse III, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France
| |
Collapse
|
29
|
Poulter NS, Pollitt AY, Davies A, Malinova D, Nash GB, Hannon MJ, Pikramenou Z, Rappoport JZ, Hartwig JH, Owen DM, Thrasher AJ, Watson SP, Thomas SG. Platelet actin nodules are podosome-like structures dependent on Wiskott-Aldrich syndrome protein and ARP2/3 complex. Nat Commun 2015; 6:7254. [PMID: 26028144 PMCID: PMC4458878 DOI: 10.1038/ncomms8254] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 04/21/2015] [Indexed: 11/09/2022] Open
Abstract
The actin nodule is a novel F-actin structure present in platelets during early spreading. However, only limited detail is known regarding nodule organization and function. Here we use electron microscopy, SIM and dSTORM super-resolution, and live-cell TIRF microscopy to characterize the structural organization and signalling pathways associated with nodule formation. Nodules are composed of up to four actin-rich structures linked together by actin bundles. They are enriched in the adhesion-related proteins talin and vinculin, have a central core of tyrosine phosphorylated proteins and are depleted of integrins at the plasma membrane. Nodule formation is dependent on Wiskott–Aldrich syndrome protein (WASp) and the ARP2/3 complex. WASp−/− mouse blood displays impaired platelet aggregate formation at arteriolar shear rates. We propose actin nodules are platelet podosome-related structures required for platelet–platelet interaction and their absence contributes to the bleeding diathesis of Wiskott–Aldrich syndrome. During early platelet spreading a novel F-actin structure forms, called the actin nodule. Here Poulter et al. demonstrate that actin nodule formation depends on WASp and the Arp2/3 complex, and using super-resolution microscopy they show that nodules bear a structural resemblance to podosomes.
Collapse
Affiliation(s)
- Natalie S Poulter
- Centre for Cardiovascular Sciences, The Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Alice Y Pollitt
- Centre for Cardiovascular Sciences, The Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Amy Davies
- PSIBS doctoral training centre, School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Dessislava Malinova
- Molecular Immunology Unit, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Gerard B Nash
- Centre for Cardiovascular Sciences, The Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Mike J Hannon
- PSIBS doctoral training centre, School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Zoe Pikramenou
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Joshua Z Rappoport
- The Center for Advanced Microscopy and Nikon Imaging Center, Morton 2-681, Northwestern University Feinberg School of Medicine, 303 E. Chicago Avenue, Chicago, Illinois 60611, USA
| | - John H Hartwig
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Dylan M Owen
- Randall Division of Cell and Molecular Biophysics, New Hunt's House, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Adrian J Thrasher
- Molecular Immunology Unit, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Stephen P Watson
- Centre for Cardiovascular Sciences, The Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Steven G Thomas
- Centre for Cardiovascular Sciences, The Medical School, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
30
|
Abstract
Rho GTPases are critical for platelet function. Although the roles of RhoA, Rac and Cdc42 are characterized, platelets express other Rho GTPases, whose activities are less well understood. This review summarizes our understanding of the roles of platelet Rho GTPases and focuses particularly on the functions of Rif and RhoG. In human platelets, Rif interacts with cytoskeleton regulators including formins mDia1 and mDia3, whereas RhoG binds SNARE-complex proteins and cytoskeletal regulators ELMO and DOCK1. Knockout mouse studies suggest that Rif plays no critical functions in platelets, likely due to functional overlap with other Rho GTPases. In contrast, RhoG is essential for normal granule secretion downstream of the collagen receptor GPVI. The central defect in RhoG-/- platelets is reduced dense granule secretion, which impedes integrin activation and aggregation and limits platelet recruitment to growing thrombi under shear, translating into reduced thrombus formation in vivo. Potential avenues for future work on Rho GTPases in platelets are also highlighted, including identification of the key regulator for platelet filopodia formation and investigation of the role of the many Rho GTPase regulators in platelet function in both health and disease.
Collapse
|
31
|
Pak2 restrains endomitosis during megakaryopoiesis and alters cytoskeleton organization. Blood 2015; 125:2995-3005. [PMID: 25824689 DOI: 10.1182/blood-2014-10-604504] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 03/17/2015] [Indexed: 12/13/2022] Open
Abstract
Megakaryocyte maturation and polyploidization are critical for platelet production; abnormalities in these processes are associated with myeloproliferative disorders, including thrombocytopenia. Megakaryocyte maturation signals through cascades that involve p21-activated kinase (Pak) function; however, the specific role for Pak kinases in megakaryocyte biology remains elusive. Here, we identify Pak2 as an essential effector of megakaryocyte maturation, polyploidization, and proplatelet formation. Genetic deletion of Pak2 in murine bone marrow is associated with macrothrombocytopenia, altered megakaryocyte ultrastructure, increased bone marrow megakaryocyte precursors, and an elevation of mature CD41(+) megakaryocytes, as well as an increased number of polyploid cells. In Pak2(-/-) mice, platelet clearance rate was increased, as was production of newly synthesized, reticulated platelets. In vitro, Pak2(-/-) megakaryocytes demonstrate increased polyploidization associated with alterations in β1-tubulin expression and organization, decreased proplatelet extensions, and reduced phosphorylation of the endomitosis regulators LIM domain kinase 1, cofilin, and Aurora A/B/C. Together, these data establish a novel role for Pak2 as an important regulator of megakaryopoiesis, polyploidization, and cytoskeletal dynamics in developing megakaryocytes.
Collapse
|
32
|
Li J, Kim K, Barazia A, Tseng A, Cho J. Platelet-neutrophil interactions under thromboinflammatory conditions. Cell Mol Life Sci 2015; 72:2627-43. [PMID: 25650236 DOI: 10.1007/s00018-015-1845-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Revised: 01/07/2015] [Accepted: 01/26/2015] [Indexed: 12/11/2022]
Abstract
Platelets primarily mediate hemostasis and thrombosis, whereas leukocytes are responsible for immune responses. Since platelets interact with leukocytes at the site of vascular injury, thrombosis and vascular inflammation are closely intertwined and occur consecutively. Recent studies using real-time imaging technology demonstrated that platelet-neutrophil interactions on the activated endothelium are an important determinant of microvascular occlusion during thromboinflammatory disease in which inflammation is coupled to thrombosis. Although the major receptors and counter receptors have been identified, it remains poorly understood how heterotypic platelet-neutrophil interactions are regulated under disease conditions. This review discusses our current understanding of the regulatory mechanisms of platelet-neutrophil interactions in thromboinflammatory disease.
Collapse
Affiliation(s)
- Jing Li
- Department of Pharmacology, University of Illinois College of Medicine, 835 S. Wolcott Ave, E403, Chicago, IL, 60612, USA
| | | | | | | | | |
Collapse
|
33
|
Elvers M. RhoGAPs and Rho GTPases in platelets. Hamostaseologie 2015; 36:168-77. [PMID: 25639730 DOI: 10.5482/hamo-14-09-0046] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/13/2015] [Indexed: 01/03/2023] Open
Abstract
Platelet cytoskeletal reorganization is essential for platelet adhesion and thrombus formation in hemostasis and thrombosis. The Rho GTPases RhoA, Rac1 and Cdc42 are the main players in cytoskeletal dynamics of platelets responsible for the formation of filopodia and lamellipodia to strongly increase the platelet surface upon activation. They are involved in platelet activation and aggregate formation including platelet secretion, integrin activation and arterial thrombus formation. The activity of Rho GTPases is tightly controlled by different proteins such as GTPase-activating proteins (GAPs). GAPs stimulate GTP hydrolysis to terminate Rho signaling. The role and impact of GAPs in platelets is not well-defined and many of the RhoGAPs identified are not known to be present in platelets or to have any function in platelets. The recently identified RhoGAPs Oligophrenin1 (OPHN1) and Nadrin regulate the activity of RhoA, Rac1 and Cdc42 and subsequent platelet cytoskeletal reorganization, platelet activation and thrombus formation. In the last years, the analysis of genetically modified mice helped to gain the understanding of Rho GTPases and their regulators in cytoskeletal rearrangements and other Rho mediated cellular processes in platelets.
Collapse
Affiliation(s)
- Margitta Elvers
- Margitta Elvers, Ph.D., Department of Clinical and Experimental Hemostasis, Hemotherapy and Transfusion Medicine, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany, Tel. +49/(0)211/81-08851, Fax -17498., E-mail:
| |
Collapse
|
34
|
Zhang Q, Dove CG, Hor JL, Murdock HM, Strauss-Albee DM, Garcia JA, Mandl JN, Grodick RA, Jing H, Chandler-Brown DB, Lenardo TE, Crawford G, Matthews HF, Freeman AF, Cornall RJ, Germain RN, Mueller SN, Su HC. DOCK8 regulates lymphocyte shape integrity for skin antiviral immunity. ACTA ACUST UNITED AC 2014; 211:2549-66. [PMID: 25422492 PMCID: PMC4267229 DOI: 10.1084/jem.20141307] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Zhang et al. show that DOCK8-deficient T and NK cells develop cell and nuclear shape abnormalities that do not impair chemotaxis but contribute to a form of cell death they term cytothripsis. Cytothripsis of DOCK8-deficient cells prevents the generation of long-lived skin-resident memory CD8 T cells resulting in impaired immune response to skin infection. DOCK8 mutations result in an inherited combined immunodeficiency characterized by increased susceptibility to skin and other infections. We show that when DOCK8-deficient T and NK cells migrate through confined spaces, they develop cell shape and nuclear deformation abnormalities that do not impair chemotaxis but contribute to a distinct form of catastrophic cell death we term cytothripsis. Such defects arise during lymphocyte migration in collagen-dense tissues when DOCK8, through CDC42 and p21-activated kinase (PAK), is unavailable to coordinate cytoskeletal structures. Cytothripsis of DOCK8-deficient cells prevents the generation of long-lived skin-resident memory CD8 T cells, which in turn impairs control of herpesvirus skin infections. Our results establish that DOCK8-regulated shape integrity of lymphocytes prevents cytothripsis and promotes antiviral immunity in the skin.
Collapse
Affiliation(s)
- Qian Zhang
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Christopher G Dove
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jyh Liang Hor
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, and The ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, and The ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Heardley M Murdock
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Dara M Strauss-Albee
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jordan A Garcia
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Judith N Mandl
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Rachael A Grodick
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Huie Jing
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Devon B Chandler-Brown
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Timothy E Lenardo
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Greg Crawford
- MRC Human Immunology Unit, Nuffield Department of Medicine, Oxford University, Oxford OX3 7BN, England, UK
| | - Helen F Matthews
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Alexandra F Freeman
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Richard J Cornall
- MRC Human Immunology Unit, Nuffield Department of Medicine, Oxford University, Oxford OX3 7BN, England, UK
| | - Ronald N Germain
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Scott N Mueller
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, and The ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, and The ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Helen C Su
- Laboratory of Host Defenses, Laboratory of Systems Biology, Laboratory of Immunology, and Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
35
|
Kassassir H, Siewiera K, Sychowski R, Watała C. Can the antiplatelet effects of cangrelor be reliably studied in mice under in vivo and in vitro conditions using flow cytometry? Pharmacol Rep 2014; 65:870-83. [PMID: 24145081 DOI: 10.1016/s1734-1140(13)71068-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 02/05/2013] [Indexed: 11/17/2022]
Abstract
BACKGROUND The effects of blood platelet inhibitors are often not quite equivalent under in vivo and in vitro conditions. Amongst various models of human pathology using laboratory animals, mice offer several benefits that make them convenient tools for studying the putative therapeutic value of various compounds. However, despite its advantages, the mouse model has methodological limitations concerning the small amount of blood available and technical difficulties with its collection. Among the variety of available methods used to study blood platelet activation and/or reactivity, flow cytometry seems an attractive technique that largely minimizes the constraints of using small rodents and enables outcomes of laboratory research to be transferred successfully to clinical practice. In this study we aimed at a critical evaluation of the optimal discriminative flow cytometric protocol, useful for reliable studies of the effect of cangrelor, a P2Y12 receptor antagonist, on mouse platelets under in vitro and in vivo conditions. METHODS Blood samples were drawn from two-month-old female BALB/c mice. Protocols differing in methods of anesthesia, blood withdrawal, anticoagulation, gating antibodies, blood preparation and fixation were tested to optimize the one best suited to discrimination between resting and activated platelets. The antiplatelet capabilities of cangrelor were tested in vitro (140 μM in whole blood) and in vivo (7.8 mg/kg b.w. administered once, directly into the bloodstream through the vena cava of the anesthetized animal, 15 min prior to blood withdrawal). Expressions of P-selectin, activated α(IIb)β3 complex and GPIba were monitored using two-color flow cytometry. RESULTS "Washed blood" anticoagulated with low molecular weight heparin demonstrated the best discrimination between circulating (resting) platelets and upon their in vitro response to thrombin, collagen or ADP in freshly-stained unfixed cell suspensions. Cangrelor inhibited the expression of the active form of the integrin a(IIb)β3 to approximately the same extent under in vitro and in vivo conditions (84.5 ± 7.7% vs. 75.4 ± 19.5% for the in vitro and in vivo approaches, respectively, n.s.). CONCLUSIONS The agreement between the in vivo and in vitro approaches with respect to cangrelor-inhibited hallmarks of blood platelet activation and reactivity supports our proposal that flow cytometry is useful and reliable for determining the effects of antiplatelet agents on the activation of circulating platelets in the mouse model, as well as the in vitro response of platelets to agonists.
Collapse
Affiliation(s)
- Hassan Kassassir
- Laboratory of Animal Experimental Models, Department of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Veterans' Central Hospital, Żeromskiego 113, PL 90-549 Łódź, Poland.
| | | | | | | |
Collapse
|
36
|
Menter DG, Tucker SC, Kopetz S, Sood AK, Crissman JD, Honn KV. Platelets and cancer: a casual or causal relationship: revisited. Cancer Metastasis Rev 2014; 33:231-69. [PMID: 24696047 PMCID: PMC4186918 DOI: 10.1007/s10555-014-9498-0] [Citation(s) in RCA: 232] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human platelets arise as subcellular fragments of megakaryocytes in bone marrow. The physiologic demand, presence of disease such as cancer, or drug effects can regulate the production circulating platelets. Platelet biology is essential to hemostasis, vascular integrity, angiogenesis, inflammation, innate immunity, wound healing, and cancer biology. The most critical biological platelet response is serving as "First Responders" during the wounding process. The exposure of extracellular matrix proteins and intracellular components occurs after wounding. Numerous platelet receptors recognize matrix proteins that trigger platelet activation, adhesion, aggregation, and stabilization. Once activated, platelets change shape and degranulate to release growth factors and bioactive lipids into the blood stream. This cyclic process recruits and aggregates platelets along with thrombogenesis. This process facilitates wound closure or can recognize circulating pathologic bodies. Cancer cell entry into the blood stream triggers platelet-mediated recognition and is amplified by cell surface receptors, cellular products, extracellular factors, and immune cells. In some cases, these interactions suppress immune recognition and elimination of cancer cells or promote arrest at the endothelium, or entrapment in the microvasculature, and survival. This supports survival and spread of cancer cells and the establishment of secondary lesions to serve as important targets for prevention and therapy.
Collapse
Affiliation(s)
- David G Menter
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | | | | | | | | | | |
Collapse
|
37
|
Jiang P, Lan Y, Luo J, Ren YL, Liu DG, Pang JX, Liu J, Li J, Wang C, Cai JP. Rapamycin promoted thrombosis and platelet adhesion to endothelial cells by inducing membrane remodeling. BMC Cell Biol 2014; 15:7. [PMID: 24564184 PMCID: PMC3936831 DOI: 10.1186/1471-2121-15-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 02/06/2014] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Recently, evidence indicated that the rapamycin-eluting stent which was used worldwide may contribute to an increased risk for thrombosis. On the contrary, other researchers found it was safe. Thus, it is necessary to clarify the effect of rapamycin on thrombosis and the corresponding mechanisms. RESULTS The effects of rapamycin in vivo were evaluated by modified deep vein thrombosis animal model. The platelets were from healthy volunteers and the platelet-endothelium (purchased from ATCC) adhesion in cultured endothelial cells was assessed. Membrane rufflings in endothelial cells were examined by confocal and electron microscope. Thrombus formation increased in rats that were injected with rapamycin. Electron microscope analysis exhibited microvilli on the rapamycin-treated endothelium in rats. Rapamycin enhanced membrane ruffling in human umbilical vein endothelial cells (HUVECs) and adhesion of platelets to HUVECs. The platelet-HUVECs adhesion was attenuated when cells were treated with cytochalacin B. Inhibition of autophagy by 3-methyladenine led to suppression of membrane ruffles in HUVECs and augmentation of platelet-endothelial adhesion. CONCLUSIONS In conclusion, we found that endothelial membrane remodeling induced by rapamycin is crucial for the adhesion of platelets to endothelial cells and thereby for thrombosis in vivo, and that the endothelial membrane remodeling is autophagy dependent.
Collapse
Affiliation(s)
- Ping Jiang
- The Key Laboratory of Geriatrics, Beijing Hospital and Beijing Institute of Geriatrics, Ministry of Health, No,1, DaHua Road, Dong Dan, Beijing 100730, P,R,China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The molecular pathways that regulate megakaryocyte production have historically been identified through multiple candidate gene approaches. Several transcription factors critical for generating megakaryocytes were identified by promoter analysis of megakaryocyte-specific genes, and their biological roles then verified by gene knockout studies; for example, GATA-1, NF-E2, and RUNX1 were identified in this way. In contrast, other transcription factors important for megakaryopoiesis were discovered through a systems approach; for example, c-Myb was found to be critical for the erythroid versus megakaryocyte lineage decision by genome-wide loss-of-function studies. The regulation of the levels of these transcription factors is, for the most part, cell intrinsic, although that assumption has recently been challenged. Epigenetics also impacts megakaryocyte gene expression, mediated by histone acetylation and methylation. Several cytokines have been identified to regulate megakaryocyte survival, proliferation, and differentiation, most prominent of which is thrombopoietin. Upon binding to its receptor, the product of the c-Mpl proto-oncogene, thrombopoietin induces a conformational change that activates a number of secondary messengers that promote cell survival, proliferation, and differentiation, and down-modulate receptor signaling. Among the best studied are the signal transducers and activators of transcription (STAT) proteins; phosphoinositol-3-kinase; mitogen-activated protein kinases; the phosphatases PTEN, SHP1, SHP2, and SHIP1; and the suppressors of cytokine signaling (SOCS) proteins. Additional signals activated by these secondary mediators include mammalian target of rapamycin; β(beta)-catenin; the G proteins Rac1, Rho, and CDC42; several transcription factors, including hypoxia-inducible factor 1α(alpha), the homeobox-containing proteins HOXB4 and HOXA9, and a number of signaling mediators that are reduced, including glycogen synthase kinase 3α(alpha) and the FOXO3 family of forkhead proteins. More recently, systematic interrogation of several aspects of megakaryocyte formation have been conducted, employing genomics, proteomics, and chromatin immunoprecipitation (ChIP) analyses, among others, and have yielded many previously unappreciated signaling mechanisms that regulate megakaryocyte lineage determination, proliferation, and differentiation. This chapter focuses on these pathways in normal and neoplastic megakaryopoiesis, and suggests areas that are ripe for further study.
Collapse
|
39
|
|
40
|
Aslan JE, Baker SM, Loren CP, Haley KM, Itakura A, Pang J, Greenberg DL, David LL, Manser E, Chernoff J, McCarty OJT. The PAK system links Rho GTPase signaling to thrombin-mediated platelet activation. Am J Physiol Cell Physiol 2013; 305:C519-28. [PMID: 23784547 DOI: 10.1152/ajpcell.00418.2012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Regulation of the platelet actin cytoskeleton by the Rho family of small GTPases is essential for the proper maintenance of hemostasis. However, little is known about how intracellular platelet activation from Rho GTPase family members, including Rac, Cdc42, and Rho, translate into changes in platelet actin structures. To better understand how Rho family GTPases coordinate platelet activation, we identified platelet proteins associated with Rac1, a Rho GTPase family member, and actin regulatory protein essential for platelet hemostatic function. Mass spectrometry analysis revealed that upon platelet activation with thrombin, Rac1 associates with a set of effectors of the p21-activated kinases (PAKs), including GIT1, βPIX, and guanine nucleotide exchange factor GEFH1. Platelet activation by thrombin triggered the PAK-dependent phosphorylation of GIT1, GEFH1, and other PAK effectors, including LIMK1 and Merlin. PAK was also required for the thrombin-mediated activation of the MEK/ERK pathway, Akt, calcium signaling, and phosphatidylserine (PS) exposure. Inhibition of PAK signaling prevented thrombin-induced platelet aggregation and blocked platelet focal adhesion and lamellipodia formation in response to thrombin. Together, these results demonstrate that the PAK signaling system is a key orchestrator of platelet actin dynamics, linking Rho GTPase activation downstream of thrombin stimulation to PAK effector function, MAP kinase activation, calcium signaling, and PS exposure in platelets.
Collapse
Affiliation(s)
- Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Aslan JE, Itakura A, Haley KM, Tormoen GW, Loren CP, Baker SM, Pang J, Chernoff J, McCarty OJT. p21 activated kinase signaling coordinates glycoprotein receptor VI-mediated platelet aggregation, lamellipodia formation, and aggregate stability under shear. Arterioscler Thromb Vasc Biol 2013; 33:1544-51. [PMID: 23640496 DOI: 10.1161/atvbaha.112.301165] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Rho GTPase proteins play a central role in regulating the dynamics of the platelet actin cytoskeleton. Yet, little is known regarding how Rho GTPase activation coordinates platelet activation and function. In this study, we aimed to characterize the role of the Rho GTPase effector, p21 activated kinase (PAK), in platelet activation, lamellipodia formation, and aggregate formation under shear. APPROACH AND RESULTS Stimulation of platelets with the glycoprotein receptor VI agonist, collagen-related peptide, rapidly activated PAK in a time course preceding phosphorylation of PAK substrates, LIM domain kinase LIMK1 and the MAPK/ERK kinase MEK, and the subsequent activation of MAPKs and Akt. Pharmacological inhibitors of PAK blocked signaling events downstream of PAK and prevented platelet secretion as well as platelet aggregation in response to collagen-related peptide. PAK inhibitors also prevented PAK activation and platelet spreading on collagen surfaces. PAK was also required for the formation of platelet aggregates and to maintain aggregate stability under physiological shear flow conditions. CONCLUSIONS These results suggest that PAK serves as an orchestrator of platelet functional responses after activation downstream of the platelet collagen receptor, glycoprotein receptor VI.
Collapse
Affiliation(s)
- Joseph E Aslan
- Department of Biomedical Engineering, Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Small GTPase proteins regulate cytoskeletal dynamics to orchestrate diverse cellular functions in organismal physiology, development and disease. The Rho GTPase family member Rac1 is central to actin-driven processes in a number of cell types, particularly platelets, where Rac1 serves as an essential mediator of lamellipodia formation and thrombus stability. Despite the importance of Rac1 to platelet function, little is known about how Rac1 activity is regulated in platelets. We recently defined the tyrosine-kinase based signaling cascade that activates mTOR to regulate Rac1 activation downstream of platelet integrin and glycoprotein receptors. We demonstrated a critical role for the mTOR-Rac1 axis in regulating platelet spreading, aggregation and aggregate stability under shear. These studies suggest that in addition to cancer and transplant medicine, intervention of the mTOR system may have implications for hemostatic and thrombotic processes as well as immunotherapies and intravascular stent design.
Collapse
Affiliation(s)
- Joseph E Aslan
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR USA.
| | | |
Collapse
|
43
|
Goggs R, Savage JS, Mellor H, Poole AW. The small GTPase Rif is dispensable for platelet filopodia generation in mice. PLoS One 2013; 8:e54663. [PMID: 23359340 PMCID: PMC3554654 DOI: 10.1371/journal.pone.0054663] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 12/13/2012] [Indexed: 11/18/2022] Open
Abstract
Background Formation of filopodia and other shape change events are vital for platelet hemostatic function. The mechanisms regulating filopodia formation by platelets are incompletely understood however. In particular the small GTPase responsible for initiating filopodia formation by platelets remains elusive. The canonical pathway involving Cdc42 is not essential for filopodia formation in mouse platelets. The small GTPase Rif (RhoF) provides an alternative route to filopodia generation in other cell types and is expressed in both human and mouse platelets. Hypothesis/Objective We hypothesized that Rif might be responsible for generating filopodia by platelets and generated a novel knockout mouse model to investigate the functional role of Rif in platelets. Methodology/Principal Findings Constitutive RhoF−/− mice are viable and have normal platelet, leukocyte and erythrocyte counts and indices. RhoF−/− platelets form filopodia and spread normally on various agonist surfaces in static conditions and under arterial shear. In addition, RhoF−/− platelets have normal actin dynamics, are able to activate and aggregate normally and secrete from alpha and dense granules in response to collagen related peptide and thrombin stimulation. Conclusions The small GTPase Rif does not appear to be critical for platelet function in mice. Functional overlap between Rif and other small GTPases may be responsible for the non-essential role of Rif in platelets.
Collapse
Affiliation(s)
- Robert Goggs
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom
| | - Joshua S. Savage
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom
| | - Harry Mellor
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Alastair W. Poole
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom
- * E-mail:
| |
Collapse
|
44
|
Abstract
The Rho family of GTP binding proteins, also commonly referred to as the Rho GTPases, are master regulators of the platelet cytoskeleton and platelet function. These low-molecular-weight or 'small' GTPases act as signaling switches in the spatial and temporal transduction, and amplification of signals from platelet cell surface receptors to the intracellular signaling pathways that drive platelet function. The Rho GTPase family members RhoA, Cdc42 and Rac1 have emerged as key regulators in the dynamics of the actin cytoskeleton in platelets and play key roles in platelet aggregation, secretion, spreading and thrombus formation. Rho GTPase regulators, including GEFs and GAPs and downstream effectors, such as the WASPs, formins and PAKs, may also regulate platelet activation and function. In this review, we provide an overview of Rho GTPase signaling in platelet physiology. Previous studies of Rho GTPases and platelets have had a shared history, as platelets have served as an ideal, non-transformed cellular model to characterize Rho function. Likewise, recent studies of the cell biology of Rho GTPase family members have helped to build an understanding of the molecular regulation of platelet function and will continue to do so through the further characterization of Rho GTPases as well as Rho GAPs, GEFs, RhoGDIs and Rho effectors in actin reorganization and other Rho-driven cellular processes.
Collapse
Affiliation(s)
- J E Aslan
- Department of Biomedical Engineering and Cell & Developmental Biology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA.
| | | |
Collapse
|
45
|
Shen B, Delaney MK, Du X. Inside-out, outside-in, and inside-outside-in: G protein signaling in integrin-mediated cell adhesion, spreading, and retraction. Curr Opin Cell Biol 2012; 24:600-6. [PMID: 22980731 DOI: 10.1016/j.ceb.2012.08.011] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 06/06/2012] [Accepted: 08/20/2012] [Indexed: 12/11/2022]
Abstract
The integrin family of cell adhesion receptors mediates bi-directional signaling: 'inside-out' signaling activates the ligand binding function of integrins and 'outside-in' signaling mediates cellular responses induced by ligand binding to integrins leading to cell spreading, retraction, migration, and proliferation. Integrin signaling requires both heterotrimeric G proteins and monomeric small G proteins. This review focuses on recent development in the roles of G proteins in integrin outside-in signaling. The finding of direct interaction between the heterotrimeric G protein subunit Gα13 and integrin β subunits reveals a new mechanism for integrin signaling, and also uncovers a crosstalk between the signaling pathways initiated by G protein-coupled receptors (GPCRs) and integrins. This crosstalk, which may be referred to as 'inside-outside-in' signaling, dynamically regulates contractility and greatly promotes integrin outside-in signaling.
Collapse
Affiliation(s)
- Bo Shen
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612, United States
| | | | | |
Collapse
|
46
|
Rho GTPase function in development: How in vivo models change our view. Exp Cell Res 2012; 318:1779-87. [DOI: 10.1016/j.yexcr.2012.05.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 05/07/2012] [Accepted: 05/10/2012] [Indexed: 12/16/2022]
|
47
|
Schaff M, Receveur N, Bourdon C, Ohlmann P, Lanza F, Gachet C, Mangin PH. β-arrestin-1 participates in thrombosis and regulates integrin aIIbβ3 signalling without affecting P2Y receptors desensitisation and function. Thromb Haemost 2012; 107:735-48. [PMID: 22318609 DOI: 10.1160/th11-06-0430] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 01/05/2012] [Indexed: 01/21/2023]
Abstract
β-arrestin-1 (β-arr1) and β-arrestin-2 (β-arr2) are cytosolic proteins well-known to participate in G protein-coupled receptor desensitisation and signalling. We used genetically-inactivated mice to evaluate the role of β-arr1 or β-arr2 in platelet function, P2Y receptor desensitisation, haemostasis and thrombosis. Platelet aggregation, soluble fibrinogen binding and P-selectin exposure induced by various agonists were near normal in β-arr1-/- and β-arr2-/- platelets. In addition, deficiency in β-arr1 or β-arr2 was not critical for P2Y receptors desensitisation. A functional redundancy between β-arr1 and β-arr2 may explain these unchanged platelet responses. Interestingly, β-arr1-/- but not β-arr2-/- mice were protected against laser- and FeCl3-induced thrombosis. The tail bleeding times, number of rebleeds and volume of blood loss were unchanged in β-arr1-/- and β-arr2-/- mice, suggesting no defect in haemostasis. β-arr1-/- platelet activation upon adhesion to immobilised fibrinogen was inhibited, as attested by a 37 ± 5% (n = 3, p<0.0001) decrease in filopodia extension, suggesting defective signalling through integrin αIIbβ3. β-arr1 appeared to be located downstream of Src family kinases and to regulate αIIbβ3 signalling by increasing Akt phosphorylation. Overall, this study supports a role for β-arr1 in promoting thrombus formation, in part through its participation in αIIbβ3 signalling, and no role of β-arr1 and β-arr2 in agonist-induced platelet activation and P2Y receptors desensitisation.
Collapse
Affiliation(s)
- M Schaff
- INSERM UMR-S949, Etablissement Français du Sang-Alsace (EFS-Alsace), 10, rue Spielmann, BP 36, F-67065 Strasbourg Cedex, France
| | | | | | | | | | | | | |
Collapse
|
48
|
Parguiña AF, Grigorian-Shamagian L, Agra RM, López-Otero D, Rosa I, Alonso J, Teijeira-Fernández E, González-Juanatey JR, García Á. Variations in Platelet Proteins Associated With ST-Elevation Myocardial Infarction. Arterioscler Thromb Vasc Biol 2011; 31:2957-64. [DOI: 10.1161/atvbaha.111.235713] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Our aim in this study was to provide novel information on the molecular mechanisms playing a major role in the unwanted platelet activation associated with ST-elevation myocardial infarction (STEMI).
Methods and Results—
We compared the platelet proteome of 11 STEMI patients to a matched control group of 15 stable chronic ischemic cardiopathy patients. In addition, we did a prospective study to follow the STEMI patients over time. Proteins were separated by high-resolution 2D gel electrophoresis, identified by mass spectrometry, and validated by Western blotting. Platelets from STEMI patients on admission displayed 56 protein spot differences (corresponding to 42 unique genes) compared with the control group. The number of differences decreased with time during the patients' follow-up. Interestingly, the adapter protein CrkL and the active form of Src (phosphorylated in Tyr418) were found to be upregulated in platelets from STEMI patients. Major signaling pathways related to the proteins identified include integrin, integrin-linked kinase, and glycoprotein VI (GPVI) signaling. Interestingly, a study on an independent cohort of patients showed a higher degree of activation of GPVI signaling in STEMI patients.
Conclusion—
CrkL, the active form of Src, and GPVI signaling are upregulated in platelets from STEMI patients.
Collapse
Affiliation(s)
- Andrés F. Parguiña
- From the Department of Pharmacology, Faculty of Pharmacy (A.F.P., I.R., A.G.) and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) (A.F.P., I.R., A.G.), Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Department, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (L.G.-S.); Cardiology Department and Coronary Unit (L.G.-S., R.M.A., D.L.-O., E.T.-F., J.R.G.-J.) and Proteomic Unit, Instituto de Investigaciones Sanitarias (J.A.), Hospital
| | - Lilian Grigorian-Shamagian
- From the Department of Pharmacology, Faculty of Pharmacy (A.F.P., I.R., A.G.) and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) (A.F.P., I.R., A.G.), Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Department, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (L.G.-S.); Cardiology Department and Coronary Unit (L.G.-S., R.M.A., D.L.-O., E.T.-F., J.R.G.-J.) and Proteomic Unit, Instituto de Investigaciones Sanitarias (J.A.), Hospital
| | - Rosa M. Agra
- From the Department of Pharmacology, Faculty of Pharmacy (A.F.P., I.R., A.G.) and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) (A.F.P., I.R., A.G.), Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Department, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (L.G.-S.); Cardiology Department and Coronary Unit (L.G.-S., R.M.A., D.L.-O., E.T.-F., J.R.G.-J.) and Proteomic Unit, Instituto de Investigaciones Sanitarias (J.A.), Hospital
| | - Diego López-Otero
- From the Department of Pharmacology, Faculty of Pharmacy (A.F.P., I.R., A.G.) and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) (A.F.P., I.R., A.G.), Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Department, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (L.G.-S.); Cardiology Department and Coronary Unit (L.G.-S., R.M.A., D.L.-O., E.T.-F., J.R.G.-J.) and Proteomic Unit, Instituto de Investigaciones Sanitarias (J.A.), Hospital
| | - Isaac Rosa
- From the Department of Pharmacology, Faculty of Pharmacy (A.F.P., I.R., A.G.) and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) (A.F.P., I.R., A.G.), Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Department, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (L.G.-S.); Cardiology Department and Coronary Unit (L.G.-S., R.M.A., D.L.-O., E.T.-F., J.R.G.-J.) and Proteomic Unit, Instituto de Investigaciones Sanitarias (J.A.), Hospital
| | - Jana Alonso
- From the Department of Pharmacology, Faculty of Pharmacy (A.F.P., I.R., A.G.) and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) (A.F.P., I.R., A.G.), Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Department, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (L.G.-S.); Cardiology Department and Coronary Unit (L.G.-S., R.M.A., D.L.-O., E.T.-F., J.R.G.-J.) and Proteomic Unit, Instituto de Investigaciones Sanitarias (J.A.), Hospital
| | - Elvis Teijeira-Fernández
- From the Department of Pharmacology, Faculty of Pharmacy (A.F.P., I.R., A.G.) and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) (A.F.P., I.R., A.G.), Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Department, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (L.G.-S.); Cardiology Department and Coronary Unit (L.G.-S., R.M.A., D.L.-O., E.T.-F., J.R.G.-J.) and Proteomic Unit, Instituto de Investigaciones Sanitarias (J.A.), Hospital
| | - José Ramón González-Juanatey
- From the Department of Pharmacology, Faculty of Pharmacy (A.F.P., I.R., A.G.) and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) (A.F.P., I.R., A.G.), Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Department, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (L.G.-S.); Cardiology Department and Coronary Unit (L.G.-S., R.M.A., D.L.-O., E.T.-F., J.R.G.-J.) and Proteomic Unit, Instituto de Investigaciones Sanitarias (J.A.), Hospital
| | - Ángel García
- From the Department of Pharmacology, Faculty of Pharmacy (A.F.P., I.R., A.G.) and Center for Research in Molecular Medicine and Chronic Diseases (CIMUS) (A.F.P., I.R., A.G.), Universidade de Santiago de Compostela, Santiago de Compostela, Spain; Cardiology Department, Complexo Hospitalario Universitario de Vigo, Vigo, Spain (L.G.-S.); Cardiology Department and Coronary Unit (L.G.-S., R.M.A., D.L.-O., E.T.-F., J.R.G.-J.) and Proteomic Unit, Instituto de Investigaciones Sanitarias (J.A.), Hospital
| |
Collapse
|
49
|
A novel small molecule 1,2,3,4,6-penta-O-galloyl-α-D-glucopyranose mimics the antiplatelet actions of insulin. PLoS One 2011; 6:e26238. [PMID: 22073153 PMCID: PMC3206812 DOI: 10.1371/journal.pone.0026238] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 09/22/2011] [Indexed: 11/19/2022] Open
Abstract
Background We have shown that 1,2,3,4,6-penta-O-galloyl-α-D-glucopyranose (α-PGG), an orally effective hypoglycemic small molecule, binds to insulin receptors and activates insulin-mediated glucose transport. Insulin has been shown to bind to its receptors on platelets and inhibit platelet activation. In this study we tested our hypothesis that if insulin possesses anti-platelet properties then insulin mimetic small molecules should mimic antiplatelet actions of insulin. Principal Findings Incubation of human platelets with insulin or α-PGG induced phosphorylation of insulin receptors and IRS-1 and blocked ADP or collagen induced aggregation. Pre-treatment of platelets with α-PGG inhibited thrombin-induced release of P-selectin, secretion of ATP and aggregation. Addition of ADP or thrombin to platelets significantly decreased the basal cyclic AMP levels. Pre-incubation of platelets with α-PGG blocked ADP or thrombin induced decrease in platelet cyclic AMP levels but did not alter the basal or PGE1 induced increase in cAMP levels. Addition of α-PGG to platelets blocked agonist induced rise in platelet cytosolic calcium and phosphorylation of Akt. Administration of α-PGG (20 mg kg−1) to wild type mice blocked ex vivo platelet aggregation induced by ADP or collagen. Conclusions These data suggest that α-PGG inhibits platelet activation, at least in part, by inducing phosphorylation of insulin receptors leading to inhibition of agonist induced: (a) decrease in cyclic AMP; (b) rise in cytosolic calcium; and (c) phosphorylation of Akt. These findings taken together with our earlier reports that α-PGG mimics insulin signaling suggest that inhibition of platelet activation by α-PGG mimics antiplatelet actions of insulin.
Collapse
|