1
|
Wadding-Lee CA, Jay M, Shearer SM, Benson TW, Spuzzillo A, Howatt DA, Thompson J, Tourdot BE, Daugherty A, Mackman N, Owens Iii AP. Attenuation of Atherosclerosis With PAR4 Deficiency: A Potential Role of Hematopoietically Expressed PAR4 in Atherosclerosis. Arterioscler Thromb Vasc Biol 2025. [PMID: 40270259 DOI: 10.1161/atvbaha.124.322068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Cardiovascular disease is a significant burden globally and, despite current therapeutics, remains the leading cause of death. PAR (protease-activated receptor) 4 is a receptor highly expressed by hematopoietic cells, strongly activated by thrombin, and plays a vital role in platelet activation and aggregation. However, the role of PAR4 in atherothrombotic disease remains understudied. METHODS Mice on a low-density lipoprotein receptor-deficient (Ldlr-/-) background were bred with Par4-deficient (Par4-/-) mice to create Ldlr-/-/Par4+/+ and Ldlr-/-/Par4-/- cousin lines. Mice were fed high-fat (42%) and high-cholesterol (0.2%) Western diet for 12 weeks for all studies. Bone marrow transplant studies were conducted by irradiating Ldlr-/-/Par4+/+ and Ldlr-/-/Par4-/- mice with 550 rads (2×, 4 hours apart) and then repopulated with Par4+/+ or Par4-/- bone marrow. To determine whether the effects of thrombin were mediated solely by PAR4, the thrombin inhibitor dabigatran was added to the Western diet. RESULTS We observed higher abundance of PAR4 in arteries with atherosclerosis in mouse and human lesions versus healthy controls. Using a global deletion of PAR4, we observed an attenuation in atherosclerosis versus Par4+/+ mice. Bone marrow transplant studies demonstrated these effects were due to hematopoietic cells. When observing whether PAR4 activation via thrombin contributes to atherosclerotic development, Ldlr-/-/Par4-/- mice given dabigatran did not further decrease their atherosclerotic burden. Differences between apoE-deficient (apoE-/-) and Ldlr-/- platelets were assessed for changes in reactivity. PAR4 appeared to be acting independent of PAR1, as there were no changes with the addition of dabigatran to Par4-/- mice. apoE-/- platelets were hyperreactive compared with Ldlr-/- platelets. CONCLUSIONS We conclude that hematopoietic-derived PAR4 plays a vital role in the development and progression of atherosclerosis. More specifically, we observe thrombin-activated PAR4 contributes to the progression. Specifically, targeting of PAR4 may be a potential therapeutic target for cardiovascular disease.
Collapse
Affiliation(s)
- Caris A Wadding-Lee
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, OH. (C.A.W.-L., M.J., S.M.S., T.W.B., A.S., A.P.O.)
- Pathobiology and Molecular Medicine Program, Department of Internal Medicine, University of Cincinnati College of Medicine, OH. (C.A.W.-L., M.J., S.M.S., A.S., A.P.O.)
| | - Megan Jay
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, OH. (C.A.W.-L., M.J., S.M.S., T.W.B., A.S., A.P.O.)
- Pathobiology and Molecular Medicine Program, Department of Internal Medicine, University of Cincinnati College of Medicine, OH. (C.A.W.-L., M.J., S.M.S., A.S., A.P.O.)
| | - Shannon M Shearer
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, OH. (C.A.W.-L., M.J., S.M.S., T.W.B., A.S., A.P.O.)
- Pathobiology and Molecular Medicine Program, Department of Internal Medicine, University of Cincinnati College of Medicine, OH. (C.A.W.-L., M.J., S.M.S., A.S., A.P.O.)
| | - Tyler W Benson
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, OH. (C.A.W.-L., M.J., S.M.S., T.W.B., A.S., A.P.O.)
| | - Anthony Spuzzillo
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, OH. (C.A.W.-L., M.J., S.M.S., T.W.B., A.S., A.P.O.)
- Pathobiology and Molecular Medicine Program, Department of Internal Medicine, University of Cincinnati College of Medicine, OH. (C.A.W.-L., M.J., S.M.S., A.S., A.P.O.)
| | - Deborah A Howatt
- Department of Physiology and Saha Cardiovascular Research Center, University of Kentucky, Lexington. (D.A.H., A.D.)
| | - Joel Thompson
- Division of Endocrinology and Molecular Medicine, Department of Internal Medicine, University of Kentucky, Lexington. (J.T.)
| | - Benjamin E Tourdot
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, OH (B.E.T.)
| | - Alan Daugherty
- Department of Physiology and Saha Cardiovascular Research Center, University of Kentucky, Lexington. (D.A.H., A.D.)
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill (N.M.)
| | - A Phillip Owens Iii
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, OH. (C.A.W.-L., M.J., S.M.S., T.W.B., A.S., A.P.O.)
- Pathobiology and Molecular Medicine Program, Department of Internal Medicine, University of Cincinnati College of Medicine, OH. (C.A.W.-L., M.J., S.M.S., A.S., A.P.O.)
| |
Collapse
|
2
|
Zhang J, Hu F, Zhang J, Xie J, Wang Z, Lv L, Liang H, Liu Q, Chen R, Li H, Su W, Yan R, Chen Z, Wang Z, Tang H, Chang YN, Li J, Chen J, Shen M, Xing G, Chen K. Physical-Matched Nanoplatelets Boost Heterogeneous Thrombi Targeting Through Self-Adaptive Deformation for Thrombolysis and Endothelial Repairing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406262. [PMID: 39428893 DOI: 10.1002/smll.202406262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/14/2024] [Indexed: 10/22/2024]
Abstract
The heterogeneity of thrombi in terms of composition, structure, and blood rheology parameters presents a challenge for effective thrombus-targeting drug delivery. To address this, a self-adaptive nano-delivery system, termed D-PLT, is developed. It consists of platelet membrane-cloaked deformable mesoporous organic silicon dioxide nanocomposite, enabling it to respond to the challenge of the heterogeneity of thrombi in arteries and veins. The system exhibits progressive targeting, with the ability to target arterial and venous thrombosis and damaged blood vessels. D-PLT physically matches the pore structure of the thrombus by undergoing varied deformation, leading to advanced targeting and enrichment of arterial and venous thrombus. When co-loaded with the thrombolytic drug urokinase (UK) and the endothelium-protecting agent atorvastatin calcium (AT), the system improves rapid vascular opening of arterial and venous thrombosis in 90 min and provides up to 7 days of durable thrombolysis and recovery from endothelial dysfunction in vivo. This self-adaptive delivery system offers a promising strategy to overcome thrombus heterogeneity.
Collapse
Affiliation(s)
- Junhui Zhang
- Department of Biochemistry and Molecular Biology, Yanbian University Medical College, Yanji, Jilin, 133002, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Fan Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jing Xie
- State Key Laboratory of Explosion Science and Safety Protection, Institute of Technology Beijing, Beijing, 100081, P. R. China
| | - Zhiyu Wang
- State Key Laboratory of Explosion Science and Safety Protection, Institute of Technology Beijing, Beijing, 100081, P. R. China
| | - Linwen Lv
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Haojun Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiuyang Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Ranran Chen
- Department of Biochemistry and Molecular Biology, Yanbian University Medical College, Yanji, Jilin, 133002, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Wenxi Su
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Ruyu Yan
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Ziteng Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Zhijie Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Hongyu Tang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Ya-Nan Chang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Juan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Jun Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Minghua Shen
- Department of Biochemistry and Molecular Biology, Yanbian University Medical College, Yanji, Jilin, 133002, China
| | - Gengmei Xing
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| | - Kui Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Neutron Scattering Science and Technology, Spallation Neutron Source Science Center, Dongguan, 523803, China
| |
Collapse
|
3
|
Wadding-Lee CA, Jay M, Jones SM, Thompson J, Howatt DA, Daugherty A, Mackman N, Owens AP. Attenuation of Atherosclerosis with PAR4 Deficiency: Differential Platelet Outcomes in apoE -/- vs. Ldlr -/- Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.606266. [PMID: 39211209 PMCID: PMC11361089 DOI: 10.1101/2024.08.01.606266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Objective Cardiovascular disease (CVD) is a significant burden globally and, despite current therapeutics, remains the leading cause of death. Platelet inhibitors are of interest in CVD treatment to reduce thrombus formation post-plaque rupture as well their contribution to inflammation throughout the progression of atherosclerosis. Protease activated receptor 4 (PAR4) is a receptor highly expressed by platelets, strongly activated by thrombin, and plays a vital role in platelet activation and aggregation. However, the role of PAR4. Approach and Results Mice on a low-density lipoprotein receptor-deficient ( Ldlr -/- ) background were bred with Par4 deficient ( Par4 -/- ) mice to create Ldlr -/- /Par4 +/+ and Ldlr -/- /Par4 -/- cousin lines. Mice were fed high fat (42%) and cholesterol (0.2%) 'Western' diet for 12 weeks for all studies. Bone marrow transplant (BMT) studies were conducted by irradiating Ldlr -/- /Par4 +/+ and Ldlr -/- /Par4 -/- mice with 550 rads (2x, 4 hours apart) and then repopulated with Par4 +/+ or Par4 -/- bone marrow. To determine if the effects of thrombin were mediated solely by PAR4, the thrombin inhibitor dabigatran was added to the 'Western' diet. Ldlr -/- /Par4 -/- given dabigatran did not further decrease their atherosclerotic burden. Differences between apolipoprotein E deficient ( apoE -/- ) and Ldlr -/- platelets were assessed for changes in reactivity. We observed higher PAR4 abundance in arteries with atherosclerosis in human and mice versus healthy controls. PAR4 deficiency attenuated atherosclerosis in the aortic sinus and root versus proficient controls. BMT studies demonstrated this effect was due to hematopoietic cells, most likely platelets. PAR4 appeared to be acting independent of PAR1, as there werer no changes with addition of dabigatran to PAR4 deficient mice. apoE -/- platelets are hyperreactive compared to Ldlr -/- platelets. Conclusions Hematopoietic-derived PAR4, most likely platelets, plays a vital role in the development and progression of atherosclerosis. Specific targeting of PAR4 may be a potential therapeutic target for CVD. Highlights Deficiency of protease-activated receptor 4 attenuates the development of diet-induced atherosclerosis in a Ldlr -/- mouse model. PAR4 deficiency in hematopoietic cells is atheroprotective. PAR4 deficiency accounts for the majority of thrombin-induced atherosclerosis in a Ldlr -/- mouse model. The examination of platelet-specific proteins and platelet activation should be carefully considered before using the apoE -/- or Ldlr -/- mouse models of atherosclerosis.
Collapse
|
4
|
Lee SK, Malik RA, Zhou J, Wang W, Gross PL, Weitz JI, Ramachandran R, Trigatti BL. PAR4 Inhibition Reduces Coronary Artery Atherosclerosis and Myocardial Fibrosis in SR-B1/LDLR Double Knockout Mice. Arterioscler Thromb Vasc Biol 2023; 43:2165-2178. [PMID: 37675637 PMCID: PMC10597419 DOI: 10.1161/atvbaha.123.319767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND SR-B1 (scavenger receptor class B type 1)/LDLR (low-density lipoprotein receptor) double knockout mice fed a high-fat, high-cholesterol diet containing cholate exhibit coronary artery disease characterized by occlusive coronary artery atherosclerosis, platelet accumulation in coronary arteries, and myocardial fibrosis. Platelets are involved in atherosclerosis development, and PAR (protease-activated receptor) 4 has a prominent role in platelet function in mice. However, the role of PAR4 on coronary artery disease in mice has not been tested. METHODS We tested the effects of a PAR4 inhibitory pepducin (RAG8) on diet-induced aortic sinus and coronary artery atherosclerosis, platelet accumulation in atherosclerotic coronary arteries, and myocardial fibrosis in SR-B1/LDLR double knockout mice. SR-B1/LDLR double knockout mice were fed a high-fat, high-cholesterol diet containing cholate and injected daily with 20 mg/kg of either the RAG8 pepducin or a control reverse-sequence pepducin (SRQ8) for 20 days. RESULTS Platelets from the RAG8-treated mice exhibited reduced thrombin and PAR4 agonist peptide-mediated activation compared with those from control SRQ8-treated mice when tested ex vivo. Although aortic sinus atherosclerosis levels did not differ, RAG8-treated mice exhibited reduced coronary artery atherosclerosis, reduced platelet accumulation in atherosclerotic coronary arteries, and reduced myocardial fibrosis. These protective effects were not accompanied by changes in circulating lipids, inflammatory cytokines, or immune cells. However, RAG8-treated mice exhibited reduced VCAM-1 (vascular cell adhesion molecule 1) protein levels in nonatherosclerotic coronary artery cross sections and reduced leukocyte accumulation in atherosclerotic coronary artery cross sections compared with those from SRQ8-treated mice. CONCLUSIONS The PAR4 inhibitory RAG8 pepducin reduced coronary artery atherosclerosis and myocardial fibrosis in SR-B1/LDLR double knockout mice fed a high-fat, high-cholesterol diet containing cholate. Furthermore, RAG8 reduced VCAM-1 in nonatherosclerotic coronary arteries and reduced leukocyte and platelet accumulation in atherosclerotic coronary arteries. These findings identify PAR4 as an attractive target in reducing coronary artery disease development, and the use of RAG8 may potentially be beneficial in cardiovascular disease.
Collapse
Affiliation(s)
- Samuel K. Lee
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Ontario, Canada (S.K.L., W.W., J.I.W., B.L.T.)
| | - Rida A. Malik
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Department of Medicine (R.A.M., J.Z., P.L.G., J.I.W.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
| | - Ji Zhou
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Department of Medicine (R.A.M., J.Z., P.L.G., J.I.W.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
| | - Wei Wang
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Ontario, Canada (S.K.L., W.W., J.I.W., B.L.T.)
| | - Peter L. Gross
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Department of Medicine (R.A.M., J.Z., P.L.G., J.I.W.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
| | - Jeffrey I. Weitz
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Department of Medicine (R.A.M., J.Z., P.L.G., J.I.W.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Ontario, Canada (S.K.L., W.W., J.I.W., B.L.T.)
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.R.)
| | - Bernardo L. Trigatti
- Thrombosis and Atherosclerosis Research Institute (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.), McMaster University, Hamilton, Ontario, Canada
- Hamilton Health Sciences, Ontario, Canada (S.K.L., R.A.M., J.Z., W.W., P.L.G., J.I.W., B.L.T.)
- Department of Biochemistry and Biomedical Sciences McMaster University, Hamilton, Ontario, Canada (S.K.L., W.W., J.I.W., B.L.T.)
| |
Collapse
|
5
|
Alenazy FO, Harbi MH, Kavanagh DP, Price J, Brady P, Hargreaves O, Harrison P, Slater A, Tiwari A, Nicolson PLR, Connolly DL, Kirchhof P, Kalia N, Jandrot-Perrus M, Mangin PH, Watson SP, Thomas MR. Amplified inhibition of atherosclerotic plaque-induced platelet activation by glenzocimab with dual antiplatelet therapy. J Thromb Haemost 2023; 21:3236-3251. [PMID: 37541591 DOI: 10.1016/j.jtha.2023.07.018] [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: 01/22/2023] [Revised: 06/23/2023] [Accepted: 07/16/2023] [Indexed: 08/06/2023]
Abstract
BACKGROUND Aspirin and platelet P2Y12 inhibitors, such as ticagrelor, suboptimally inhibit microvascular thrombosis during ST-elevation myocardial infarction. Glycoprotein (GP) IIb/IIIa inhibitors may further inhibit this but cause excessive bleeding. OBJECTIVES We investigated whether combination of glenzocimab, a GPVI inhibitor, with aspirin and ticagrelor provides additional antithrombotic effects, as GPVI has a critical role in atherothrombosis but minimal involvement in hemostasis. METHODS We investigated the effects of glenzocimab (monoclonal antibody Fab fragment) using blood from healthy donors and patients with acute coronary syndrome treated with aspirin and ticagrelor. Platelets were stimulated with multiple agonists, including atherosclerotic plaque, from patients undergoing carotid endarterectomy. RESULTS Aspirin and ticagrelor partially inhibited atherosclerotic plaque-induced platelet aggregation by 48% compared with control (34 ± 3 vs 65 ± 4 U; P < .001). Plaque-induced platelet aggregation, adhesion, secretion, and activation were critically dependent on GPVI activation. Glenzocimab alone reduced plaque-induced aggregation by 75% compared with control (16 ± 4 vs 65 ± 4 U; P < .001) and by >95% when combined with aspirin and ticagrelor (3 ± 1 vs 65 ± 4 U; P < .001). Glenzocimab reduced platelet aggregation, adhesion, and thrombin generation when added to blood of aspirin- and ticagrelor-treated patients with acute coronary syndrome. Glenzocimab shared several antithrombotic effects with the GPIIb/IIIa inhibitor eptifibatide with less effect on general hemostasis assessed by rotational thromboelastometry. In a murine intravital model of ST-elevation myocardial infarction, genetic depletion of GPVI reduced microvascular thrombosis. CONCLUSION Addition of glenzocimab to aspirin and ticagrelor enhances platelet inhibition via multiple mechanisms of atherothrombosis. Compared with a GPIIb/IIIa inhibitor, glenzocimab shares multiple antithrombotic effects, with less inhibition of mechanisms involved in general hemostasis.
Collapse
Affiliation(s)
- Fawaz O Alenazy
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Maan H Harbi
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Pharmacology and Toxicology Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Dean P Kavanagh
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Joshua Price
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Brady
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Department of Cardiology, Sandwell and West Birmingham Hospitals National Health Service (NHS) Trust, Birmingham, United Kingdom
| | - Oscar Hargreaves
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Paul Harrison
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Alexandre Slater
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Alok Tiwari
- Department of Vascular Surgery, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Phillip L R Nicolson
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Derek L Connolly
- Department of Cardiology, Sandwell and West Birmingham Hospitals National Health Service (NHS) Trust, Birmingham, United Kingdom
| | - Paulus Kirchhof
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Department of Cardiology, University Heart and Vascular Center (UKE) Hamburg, Hamburg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Germany
| | - Neena Kalia
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | | | - Pierre H Mangin
- UMR_S1255, INSERM, Etablissement Francais du Sang-Alsace, Strasbourg, France
| | - Steve P Watson
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), The Universities of Birmingham and Nottingham, The Midlands, United Kingdom
| | - Mark R Thomas
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom; Department of Cardiology, Sandwell and West Birmingham Hospitals National Health Service (NHS) Trust, Birmingham, United Kingdom; Department of Cardiology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom.
| |
Collapse
|
6
|
Modulation of Glycoprotein VI and Its Downstream Signaling Pathways as an Antiplatelet Target. Int J Mol Sci 2022; 23:ijms23179882. [PMID: 36077280 PMCID: PMC9456422 DOI: 10.3390/ijms23179882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
Abstract
Antiplatelet therapy aims to reduce the risk of thrombotic events while maintaining hemostasis. A promising current approach is the inhibition of platelet glycoprotein GPVI-mediated adhesion pathways; pathways that do not involve coagulation. GPVI is a signaling receptor integral for collagen-induced platelet activation and participates in the thrombus consolidation process, being a suitable target for thrombosis prevention. Considering this, the blocking or antibody-mediated depletion of GPVI is a promising antiplatelet therapy for the effective and safe treatment of thrombotic diseases without a significant risk of bleeding and impaired hemostatic plug formation. This review describes the current knowledge concerning pharmaceutical approaches to platelet GPVI modulation and its downstream signaling pathways in this context.
Collapse
|
7
|
Lee RH, Kawano T, Grover SP, Bharathi V, Martinez D, Cowley DO, Mackman N, Bergmeier W, Antoniak S. Genetic deletion of platelet PAR4 results in reduced thrombosis and impaired hemostatic plug stability. J Thromb Haemost 2022; 20:422-433. [PMID: 34689407 PMCID: PMC8792346 DOI: 10.1111/jth.15569] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Protease-activated receptor 4 (PAR4) is expressed by a wide variety of cells, including megakaryocytes/platelets, immune cells, cardiomyocytes, and lung epithelial cells. It is the only functional thrombin receptor on murine platelets. A global deficiency of PAR4 is associated with impaired hemostasis and reduced thrombosis. OBJECTIVE We aimed to generate a mouse line with a megakaryocyte/platelet-specific deletion of PAR4 (PAR4fl/fl ;PF4Cre+ ) and use the mouse line to investigate the role of platelet PAR4 in hemostasis and thrombosis in mice. METHODS Platelets from PAR4fl/fl ;PF4Cre+ were characterized in vitro. Arterial and venous thrombosis was analyzed. Hemostatic plug formation was analyzed using a saphenous vein laser injury model in mice with global or megakaryocyte/platelet-specific deletion of PAR4 or wild-type mice treated with thrombin or glycoprotein VI (GPVI) inhibitors. RESULTS PAR4fl/fl ;PF4Cre+ platelets were unresponsive to thrombin or specific PAR4 stimulation but not to other agonists. PAR4-/- and PAR4fl/fl ;PF4Cre+ mice both exhibited a similar reduction in arterial thrombosis compared to their respective controls. More importantly, we show for the first time that platelet PAR4 is critical for venous thrombosis in mice. In addition, PAR4-/- mice and PAR4fl/fl ;PF4Cre+ mice exhibited a similar impairment in hemostatic plug stability in a saphenous vein laser injury model. Inhibition of thrombin in wild-type mice gave a similar phenotype. Combined PAR4 deficiency on platelets with GPVI inhibition did not impair hemostatic plug formation but further reduced plug stability. CONCLUSION We generated a novel PAR4fl/fl ;PF4Cre+ mouse line. We used this mouse line to show that PAR4 signaling in platelets is critical for arterial and venous thrombosis and hemostatic plug stability.
Collapse
Affiliation(s)
- Robert H. Lee
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Tomohiro Kawano
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Steven P. Grover
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Vanthana Bharathi
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - David Martinez
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Dale O. Cowley
- UNC Animal Models Core, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nigel Mackman
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wolfgang Bergmeier
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Silvio Antoniak
- UNC Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
8
|
Marín-Quílez A, García-Tuñón I, Fernández-Infante C, Hernández-Cano L, Palma-Barqueros V, Vuelta E, Sánchez-Martín M, González-Porras JR, Guerrero C, Benito R, Rivera J, Hernández-Rivas JM, Bastida JM. Characterization of the Platelet Phenotype Caused by a Germline RUNX1 Variant in a CRISPR/Cas9-Generated Murine Model. Thromb Haemost 2021; 121:1193-1205. [PMID: 33626581 DOI: 10.1055/s-0041-1723987] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
RUNX1-related disorder (RUNX1-RD) is caused by germline variants affecting the RUNX1 gene. This rare, heterogeneous disorder has no specific clinical or laboratory phenotype, making genetic diagnosis necessary. Although international recommendations have been established to classify the pathogenicity of variants, identifying the causative alteration remains a challenge in RUNX1-RD. Murine models may be useful not only for definitively settling the controversy about the pathogenicity of certain RUNX1 variants, but also for elucidating the mechanisms of molecular pathogenesis. Therefore, we developed a knock-in murine model, using the CRISPR/Cas9 system, carrying the RUNX1 p.Leu43Ser variant (mimicking human p.Leu56Ser) to study its pathogenic potential and mechanisms of platelet dysfunction. A total number of 75 mice were generated; 25 per genotype (RUNX1WT/WT, RUNX1WT/L43S, and RUNX1L43S/L43S). Platelet phenotype was assessed by flow cytometry and confocal microscopy. On average, RUNX1L43S/L43S and RUNX1WT/L43S mice had a significantly longer tail-bleeding time than RUNX1WT/WT mice, indicating the variant's involvement in hemostasis. However, only homozygous mice displayed mild thrombocytopenia. RUNX1L43S/L43S and RUNX1WT/L43S displayed impaired agonist-induced spreading and α-granule release, with no differences in δ-granule secretion. Levels of integrin αIIbβ3 activation, fibrinogen binding, and aggregation were significantly lower in platelets from RUNX1L43S/L43S and RUNX1WT/L43S using phorbol 12-myristate 13-acetate (PMA), adenosine diphosphate (ADP), and high thrombin doses. Lower levels of PKC phosphorylation in RUNX1L43S/L43S and RUNX1WT/L43S suggested that the PKC-signaling pathway was impaired. Overall, we demonstrated the deleterious effect of the RUNX1 p.Leu56Ser variant in mice via the impairment of integrin αIIbβ3 activation, aggregation, α-granule secretion, and platelet spreading, mimicking the phenotype associated with RUNX1 variants in the clinical setting.
Collapse
Affiliation(s)
- Ana Marín-Quílez
- Cancer Research Center - CSIC, Instituto de Investigación Biomédica de Salamanca, University of Salamanca, Salamanca, Spain
| | - Ignacio García-Tuñón
- Cancer Research Center - CSIC, Instituto de Investigación Biomédica de Salamanca, University of Salamanca, Salamanca, Spain
| | - Cristina Fernández-Infante
- Cancer Research Center - CSIC, Instituto de Investigación Biomédica de Salamanca, University of Salamanca, Salamanca, Spain
| | - Luis Hernández-Cano
- Cancer Research Center - CSIC, Instituto de Investigación Biomédica de Salamanca, University of Salamanca, Salamanca, Spain
| | - Verónica Palma-Barqueros
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, University of Murcia, Murcia, Spain
| | - Elena Vuelta
- Cancer Research Center - CSIC, Instituto de Investigación Biomédica de Salamanca, University of Salamanca, Salamanca, Spain
- Transgenic Facility, Nucleus, University of Salamanca, Salamanca, Spain
| | - Manuel Sánchez-Martín
- Cancer Research Center - CSIC, Instituto de Investigación Biomédica de Salamanca, University of Salamanca, Salamanca, Spain
- Transgenic Facility, Nucleus, University of Salamanca, Salamanca, Spain
- Department of Medicine, University of Salamanca, Salamanca, Spain
| | - José Ramón González-Porras
- Department of Medicine, University of Salamanca, Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca - IBSAL, Salamanca, Spain
| | - Carmen Guerrero
- Cancer Research Center - CSIC, Instituto de Investigación Biomédica de Salamanca, University of Salamanca, Salamanca, Spain
- Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Rocío Benito
- Cancer Research Center - CSIC, Instituto de Investigación Biomédica de Salamanca, University of Salamanca, Salamanca, Spain
| | - José Rivera
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, University of Murcia, Murcia, Spain
- On behalf of the "Grupo Español de Alteraciones Plaquetarias Congénitas (GEAPC)", Hemorrhagic Diathesis Working Group, SETH
| | - Jesús María Hernández-Rivas
- Cancer Research Center - CSIC, Instituto de Investigación Biomédica de Salamanca, University of Salamanca, Salamanca, Spain
- Department of Medicine, University of Salamanca, Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca - IBSAL, Salamanca, Spain
| | - José María Bastida
- Department of Medicine, University of Salamanca, Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca - IBSAL, Salamanca, Spain
- On behalf of the "Grupo Español de Alteraciones Plaquetarias Congénitas (GEAPC)", Hemorrhagic Diathesis Working Group, SETH
| |
Collapse
|
9
|
Unsworth AJ, Bye AP, Sage T, Gaspar RS, Eaton N, Drew C, Stainer A, Kriek N, Volberding PJ, Hutchinson JL, Riley R, Jones S, Mundell SJ, Cui W, Falet H, Gibbins JM. Antiplatelet properties of Pim kinase inhibition are mediated through disruption of thromboxane A2 receptor signaling. Haematologica 2021; 106:1968-1978. [PMID: 32467143 PMCID: PMC8252961 DOI: 10.3324/haematol.2019.223529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Indexed: 12/17/2022] Open
Abstract
Pim kinases are upregulated in several forms of cancer, contributing to cell survival and tumor development, but their role in platelet function and thrombotic disease has not been explored. We report for the first time that Pim-1 kinase is expressed in human and mouse platelets. Genetic deletion or pharmacological inhibition of Pim kinase results in reduced thrombus formation but is not associated with impaired hemostasis. Attenuation of thrombus formation was found to be due to inhibition of the thromboxane A2 receptor as effects on platelet function were non-additive to inhibition caused by the cyclo-oxygenase inhibitor indomethacin or the thromboxane A2 receptor antagonist GR32191. Treatment with Pim kinase inhibitors caused reduced surface expression of the thromboxane A2 receptor and resulted in reduced responses to thromboxane A2 receptor agonists, indicating a role for Pim kinase in the regulation of thromboxane A2 receptor function. Our research identifies a novel, Pim kinase-dependent regulatory mechanism for the thromboxane A2 receptor and represents a new targeting strategy that is independent of cyclo-oxygenase-1 inhibition or direct antagonism of the thromboxane A2 receptor that, while attenuating thrombosis, does not increase bleeding.
Collapse
Affiliation(s)
- Amanda J Unsworth
- University of Reading and Dept. of Life Sciences, Manchester Metropolitan University Manchester, UK
| | - Alexander P Bye
- Institute for Cardiovascular, Metabolic Research, University of Reading, Reading, UK
| | - Tanya Sage
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Renato S Gaspar
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Nathan Eaton
- Blood Research Institute and Medical College of Wisconsin, Versiti, Milwaukee, WI, USA
| | - Caleb Drew
- Blood Research Institute, Versiti, Milwaukee, WI, USA
| | - Alexander Stainer
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Neline Kriek
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Peter J Volberding
- Blood Research Institute and Medical College of Wisconsin, Versiti, Milwaukee, WI, USA
| | - James L Hutchinson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Ryan Riley
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Sarah Jones
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Stuart J Mundell
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Weiguo Cui
- Blood Research Institute, Versiti and Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hervé Falet
- Blood Research Institute, Versiti and Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jonathan M Gibbins
- Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| |
Collapse
|
10
|
Mayer K, Hein-Rothweiler R, Schüpke S, Janisch M, Bernlochner I, Ndrepepa G, Sibbing D, Gori T, Borst O, Holdenrieder S, Kupka D, Petzold T, Bradaric C, Okrojek R, Leistner DM, Trippel TD, Münzel T, Landmesser U, Pieske B, Zeiher AM, Gawaz MP, Hapfelmeier A, Laugwitz KL, Schunkert H, Kastrati A, Massberg S. Efficacy and Safety of Revacept, a Novel Lesion-Directed Competitive Antagonist to Platelet Glycoprotein VI, in Patients Undergoing Elective Percutaneous Coronary Intervention for Stable Ischemic Heart Disease: The Randomized, Double-blind, Placebo-Controlled ISAR-PLASTER Phase 2 Trial. JAMA Cardiol 2021; 6:753-761. [PMID: 33787834 DOI: 10.1001/jamacardio.2021.0475] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance The assessment of new antithrombotic agents with a favorable safety profile is clinically relevant. Objective To test the efficacy and safety of revacept, a novel, lesion-directed antithrombotic drug, acting as a competitive antagonist to platelet glycoprotein VI. Design, Setting, and Participants A phase 2 randomized clinical trial; patients were enrolled from 9 centers in Germany from November 20, 2017, to February 27, 2020; follow-up ended on March 27, 2020. The study included patients with stable ischemic heart disease (SIHD) undergoing elective percutaneous coronary intervention (PCI). Interventions Single intravenous infusion of revacept, 160 mg, revacept, 80 mg, or placebo prior to the start of PCI on top of standard antithrombotic therapy. Main Outcomes and Measures The primary end point was the composite of death or myocardial injury, defined as an increase in high-sensitivity cardiac troponin to at least 5 times the upper limit of normal within 48 hours from randomization. The safety end point was bleeding type 2 to 5 according to the Bleeding Academic Research Consortium criteria at 30 days. Results Of 334 participants (median age, 67.4 years; interquartile range, 60-75.1 years; 253 men [75.7%]; and 330 White participants [98.8%]), 120 were allocated to receive the 160-mg dose of revacept, 121 were allocated to receive the 80-mg dose, and 93 received placebo. The primary end point showed no significant differences between the revacept and placebo groups: 24.4%, 25.0%, and 23.3% in the revacept, 160 mg, revacept, 80 mg, and placebo groups, respectively (P = .98). The high dose of revacept was associated with a small but significant reduction of high-concentration collagen-induced platelet aggregation, with a median 26.5 AU × min (interquartile range, 0.5-62.2 AU × min) in the revacept, 160 mg, group; 43.5 AU × min (interquartile range, 22.8-99.5 AU × min) in the revacept, 80 mg, group; and 41.0 AU × min (interquartile range, 31.2-101.0 AU × min) in the placebo group (P = .02), while adenosine 5'-diphosphate-induced aggregation was not affected. Revacept did not increase Bleeding Academic Research Consortium type 2 or higher bleeding at 30 days compared with placebo: 5.0%, 5.9%, and 8.6% in the revacept, 160 mg, revacept, 80 mg, and placebo groups, respectively (P = .36). Conclusions and Relevance Revacept did not reduce myocardial injury in patients with stable ischemic heart disease undergoing percutaneous coronary intervention. There were few bleeding events and no significant differences between treatment arms. Trial Registration ClinicalTrials.gov Identifier: NCT03312855.
Collapse
Affiliation(s)
- Katharina Mayer
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University of Munich, Munich, Germany
| | - Ralph Hein-Rothweiler
- Department of Cardiology, Medizinische Klinik und Poliklinik I, Munich University Clinic, Ludwig-Maximilian University of Munich, Munich, Germany
| | - Stefanie Schüpke
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University of Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Marion Janisch
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University of Munich, Munich, Germany
| | - Isabell Bernlochner
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Medizinische Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Gjin Ndrepepa
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University of Munich, Munich, Germany
| | - Dirk Sibbing
- Department of Cardiology, Medizinische Klinik und Poliklinik I, Munich University Clinic, Ludwig-Maximilian University of Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Privatklinik Lauterbacher Mühle am Ostersee, Iffeldorf, Germany
| | - Tommaso Gori
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Germany
| | - Oliver Borst
- Medizinische Klinik III-Kardiologie und Angiologie, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Stefan Holdenrieder
- Institut für Laboratoriumsmedizin, Deutsches Herzzentrum München, Technical University of Munich, Munich, Germany
| | - Danny Kupka
- Department of Cardiology, Medizinische Klinik und Poliklinik I, Munich University Clinic, Ludwig-Maximilian University of Munich, Munich, Germany
| | - Tobias Petzold
- Department of Cardiology, Medizinische Klinik und Poliklinik I, Munich University Clinic, Ludwig-Maximilian University of Munich, Munich, Germany
| | - Christian Bradaric
- Medizinische Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Rainer Okrojek
- Medizinische Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - David M Leistner
- Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Klinik für Kardiologie, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Tobias D Trippel
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany.,Charité-Universitätsmedizin Berlin, Medizinische Klinik mit Schwerpunkt Kardiologie, Campus Virchow-Klinikum, Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany
| | - Thomas Münzel
- Department of Cardiology, University Medical Center Mainz, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Germany
| | - Ulf Landmesser
- Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Klinik für Kardiologie, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany
| | - Burkert Pieske
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Germany.,Charité-Universitätsmedizin Berlin, Medizinische Klinik mit Schwerpunkt Kardiologie, Campus Virchow-Klinikum, Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany
| | - Andreas M Zeiher
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Germany.,Cardiology Division, Department of Medicine III, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Meinrad P Gawaz
- Medizinische Klinik III-Kardiologie und Angiologie, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Alexander Hapfelmeier
- Institute of Medical Informatics, Statistics and Epidemiology, Technical University of Munich School of Medicine, Munich, Germany.,Institute of General Practice and Health Services Research, Technical University of Munich School of Medicine, Munich, Germany
| | - Karl-Ludwig Laugwitz
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.,Medizinische Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Heribert Schunkert
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University of Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Adnan Kastrati
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, Technical University of Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Steffen Massberg
- Department of Cardiology, Medizinische Klinik und Poliklinik I, Munich University Clinic, Ludwig-Maximilian University of Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
11
|
MacKeigan DT, Ni T, Shen C, Stratton TW, Ma W, Zhu G, Bhoria P, Ni H. Updated Understanding of Platelets in Thrombosis and Hemostasis: The Roles of Integrin PSI Domains and their Potential as Therapeutic Targets. Cardiovasc Hematol Disord Drug Targets 2021; 20:260-273. [PMID: 33001021 DOI: 10.2174/1871529x20666201001144541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/20/2020] [Accepted: 07/26/2020] [Indexed: 11/22/2022]
Abstract
Platelets are small blood cells known primarily for their ability to adhere and aggregate at injured vessels to arrest bleeding. However, when triggered under pathological conditions, the same adaptive mechanism of platelet adhesion and aggregation may cause thrombosis, a primary cause of heart attack and stroke. Over recent decades, research has made considerable progress in uncovering the intricate and dynamic interactions that regulate these processes. Integrins are heterodimeric cell surface receptors expressed on all metazoan cells that facilitate cell adhesion, movement, and signaling, to drive biological and pathological processes such as thrombosis and hemostasis. Recently, our group discovered that the plexin-semaphorin-integrin (PSI) domains of the integrin β subunits exert endogenous thiol isomerase activity derived from their two highly conserved CXXC active site motifs. Given the importance of redox reactions in integrin activation and its location in the knee region, this PSI domain activity may be critically involved in facilitating the interconversions between integrin conformations. Our monoclonal antibodies against the β3 PSI domain inhibited its thiol isomerase activity and proportionally attenuated fibrinogen binding and platelet aggregation. Notably, these antibodies inhibited thrombosis without significantly impairing hemostasis or causing platelet clearance. In this review, we will update mechanisms of thrombosis and hemostasis, including platelet versatilities and immune-mediated thrombocytopenia, discuss critical contributions of the newly discovered PSI domain thiol isomerase activity, and its potential as a novel target for anti-thrombotic therapies and beyond.
Collapse
Affiliation(s)
- Daniel T MacKeigan
- Department of Physiology, University of Toronto, Toronto, ON M5S, Canada
| | - Tiffany Ni
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Chuanbin Shen
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Tyler W Stratton
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Wenjing Ma
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Guangheng Zhu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Preeti Bhoria
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Canada
| | - Heyu Ni
- Department of Physiology, University of Toronto, Toronto, ON M5S, Canada
| |
Collapse
|
12
|
GPR56/ADGRG1 is a platelet collagen-responsive GPCR and hemostatic sensor of shear force. Proc Natl Acad Sci U S A 2020; 117:28275-28286. [PMID: 33097663 PMCID: PMC7668045 DOI: 10.1073/pnas.2008921117] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We identified the known collagen receptor GPR56/ADGRG1 on platelets. GPR56 is an adhesion G protein-coupled receptor that becomes activated following forced dissociation of its N-terminal fragment and C-terminal fragment or seven-transmembrane spanning domain (7TM). Fragment dissociation reveals the cryptic stalk of the 7TM, which acts as a tethered peptide agonist, and for GPR56, this activates platelet G13 signaling. GPR56 pharmacological probes activated platelets to undergo shape change and aggregation, which are critical for the formation of hemostatic plugs. Gpr56−/− mice exhibit prolonged bleeding, defective platelet plug formation in vessel injury assays, and delayed thrombotic vessel occlusion. Shear-force dependency of platelet adhesion to immobilized collagen was found to be GPR56 dependent. Circulating platelets roll along exposed collagen at vessel injury sites and respond with filipodia protrusion, shape change, and surface area expansion to facilitate platelet adhesion and plug formation. Various glycoproteins were considered to be both collagen responders and mediators of platelet adhesion, yet the signaling kinetics emanating from these receptors do not fully account for the rapid platelet cytoskeletal changes that occur in blood flow. We found the free N-terminal fragment of the adhesion G protein-coupled receptor (GPCR) GPR56 in human plasma and report that GPR56 is the platelet receptor that transduces signals from collagen and blood flow-induced shear force to activate G protein 13 signaling for platelet shape change. Gpr56−/− mice have prolonged bleeding, defective platelet plug formation, and delayed thrombotic occlusion. Human and mouse blood perfusion studies demonstrated GPR56 and shear-force dependence of platelet adhesion to immobilized collagen. Our work places GPR56 as an initial collagen responder and shear-force transducer that is essential for platelet shape change during hemostasis.
Collapse
|
13
|
Diacylglycerol kinase ζ is a negative regulator of GPVI-mediated platelet activation. Blood Adv 2020; 3:1154-1166. [PMID: 30967391 DOI: 10.1182/bloodadvances.2018026328] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 03/08/2019] [Indexed: 02/07/2023] Open
Abstract
Diacylglycerol kinases (DGKs) are a family of enzymes that convert diacylglycerol (DAG) into phosphatidic acid (PA). The ζ isoform of DGK (DGKζ) has been reported to inhibit T-cell responsiveness by downregulating intracellular levels of DAG. However, its role in platelet function remains undefined. In this study, we show that DGKζ was expressed at significant levels in both platelets and megakaryocytes and that DGKζ-knockout (DGKζ-KO) mouse platelets were hyperreactive to glycoprotein VI (GPVI) agonists, as assessed by aggregation, spreading, granule secretion, and activation of relevant signal transduction molecules. In contrast, they were less responsive to thrombin. Platelets from DGKζ-KO mice accumulated faster on collagen-coated microfluidic surfaces under conditions of arterial shear and stopped blood flow faster after ferric chloride-induced carotid artery injury. Other measures of hemostasis, as measured by tail bleeding time and rotational thromboelastometry analysis, were normal. Interestingly, DGKζ deficiency led to increased GPVI expression on the platelet and megakaryocyte surfaces without affecting the expression of other platelet surface receptors. These results implicate DGKζ as a novel negative regulator of GPVI-mediated platelet activation that plays an important role in regulating thrombus formation in vivo.
Collapse
|
14
|
Abstract
A confluence of technological advances in genetic manipulation and molecular-based fluorescence imaging has led to the widespread adoption of laser injury models to study hemostasis and thrombosis in mice. In all animal models of hemostasis and thrombosis, detailing the nature of experimentally induced vascular injury is paramount in enabling appropriate interpretation of experimental results. A careful appraisal of the literature shows that direct laser-induced injury can result in variable degrees of vascular damage. This review will compare and contrast models of laser injury utilized in the field, with an emphasis on the mechanism and extent of injury, the use of laser injury in different vascular beds and the molecular mechanisms regulating the response to injury. All of these topics will be discussed in the context of how distinct applications of laser injury models may be viewed as representing thrombosis and/or hemostasis.
Collapse
Affiliation(s)
- Timothy J Stalker
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania , Philadelphia, PA, USA
| |
Collapse
|
15
|
Méndez D, Urra FA, Millas-Vargas JP, Alarcón M, Rodríguez-Lavado J, Palomo I, Trostchansky A, Araya-Maturana R, Fuentes E. Synthesis of antiplatelet ortho-carbonyl hydroquinones with differential action on platelet aggregation stimulated by collagen or TRAP-6. Eur J Med Chem 2020; 192:112187. [PMID: 32155530 DOI: 10.1016/j.ejmech.2020.112187] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/23/2020] [Accepted: 02/24/2020] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases are the leading cause of death in the world. Platelets have a major role in cardiovascular events as they bind to the damaged endothelium activating and forming thrombi. Although some hydroquinone scaffold-containing compounds have known antiplatelet activities, currently there is a lack of evidence on the antiplatelet activity of hydroquinones carrying electron attractor groups. In this work, we evaluate the antiplatelet effect of a series of ortho-carbonyl hydroquinone derivatives on cytotoxicity and function of human platelets, using collagen and thrombin receptor activator peptide 6 (TRAP-6) as agonists. Our structure-activity relationship study shows that gem-diethyl/methyl substitutions and the addition/modifications of the third ring of ortho-carbonyl hydroquinone scaffold influence on the selective index (IC50 TRAP-6/IC50 Collagen) and the inhibitory capacity of platelet aggregation. Compounds 3 and 8 inhibit agonist-induced platelet aggregation in a non-competitive manner with IC50 values of 1.77 ± 2.09 μM (collagen) and 11.88 ± 4.59 μM (TRAP-6), respectively and show no cytotoxicity. Both compounds do not affect intracellular calcium levels and mitochondrial bioenergetics. Consistently, they reduce the expression of P-selectin, activation of glycoprotein IIb/IIIa, and release of adenosine triphosphate and CD63 from platelet. Our findings may be used for further development of new drugs in platelet-related thrombosis diseases.
Collapse
Affiliation(s)
- Diego Méndez
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | - Félix A Urra
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, Chile.
| | - Juan Pablo Millas-Vargas
- Instituto de Química de Recursos Naturales, Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Universidad de Talca, Talca, Chile
| | - Marcelo Alarcón
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | - Julio Rodríguez-Lavado
- Departamento de Química Orgánica y Fisicoquímica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Iván Palomo
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Talca, Chile
| | - Andrés Trostchansky
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Ramiro Araya-Maturana
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Talca, Chile; Instituto de Química de Recursos Naturales, Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Universidad de Talca, Talca, Chile; Network for Snake Venom Research and Drug Discovery, Santiago, Chile.
| | - Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Universidad de Talca, Talca, Chile.
| |
Collapse
|
16
|
Le Chapelain O, Jadoui S, Boulaftali Y, Ho-Tin-Noé B. The reversed passive Arthus reaction as a model for investigating the mechanisms of inflammation-associated hemostasis. Platelets 2020; 31:455-460. [PMID: 32105152 DOI: 10.1080/09537104.2020.1732325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
In recent years, accumulating evidence has indicated that platelets continuously repair vascular damage at sites of inflammation and/or infection. Studies in mouse models of inflammation have highlighted the fact that the mechanisms underlying bleeding prevention by platelets in inflamed organs can substantially differ from those supporting primary hemostasis following tail tip transection or thrombus formation in models of thrombosis. As a consequence, exploration of the hemostatic function of platelets in inflammation, as well as assessment of the risk of inflammation-induced bleeding associated with a platelet deficit and/or the use of anti-thrombotic drugs, require the use of dedicated experimental models. In the present review, we present the pros and cons of the cutaneous reversed passive Arthus reaction, a model of inflammation which has been instrumental in studying how inflammation causes vascular injury and how platelets continuously intervene to repair it. The limitations and common issues encountered when working with mouse models of inflammation for investigating platelet functions in inflammation are also discussed.
Collapse
Affiliation(s)
| | - Soumaya Jadoui
- Université de Paris, LVTS, Inserm U1148, F-75018 Paris, France
| | | | | |
Collapse
|
17
|
Chitooligosaccharide Biguanide Repairs Islet β‐Cell Dysfunction by Activating the IRS‐2/PI3K/Akt Signaling Pathway in Type 2 Diabetic Rats. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201800136] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
18
|
Rayes J, Watson SP, Nieswandt B. Functional significance of the platelet immune receptors GPVI and CLEC-2. J Clin Invest 2019; 129:12-23. [PMID: 30601137 DOI: 10.1172/jci122955] [Citation(s) in RCA: 217] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although platelets are best known for their role in hemostasis, they are also crucial in development, host defense, inflammation, and tissue repair. Many of these roles are regulated by the immune-like receptors glycoprotein VI (GPVI) and C-type lectin receptor 2 (CLEC-2), which signal through an immunoreceptor tyrosine-based activation motif (ITAM). GPVI is activated by collagen in the subendothelial matrix, by fibrin and fibrinogen in the thrombus, and by a remarkable number of other ligands. CLEC-2 is activated by the transmembrane protein podoplanin, which is found outside of the vasculature and is upregulated in development, inflammation, and cancer, but there is also evidence for additional ligands. In this Review, we discuss the physiological and pathological roles of CLEC-2 and GPVI and their potential as targets in thrombosis and thrombo-inflammatory disorders (i.e., disorders in which inflammation plays a critical role in the ensuing thrombosis) relative to current antiplatelet drugs.
Collapse
Affiliation(s)
- Julie Rayes
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Steve P Watson
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, United Kingdom
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| |
Collapse
|
19
|
Gillespie S, Holloway PM, Becker F, Rauzi F, Vital SA, Taylor KA, Stokes KY, Emerson M, Gavins FNE. The isothiocyanate sulforaphane modulates platelet function and protects against cerebral thrombotic dysfunction. Br J Pharmacol 2018; 175:3333-3346. [PMID: 29797311 DOI: 10.1111/bph.14368] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 04/30/2018] [Accepted: 05/04/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Platelet activation provides a critical link between inflammation and thrombosis. Sulforaphane (SFN), a naturally occurring isothiocyanate, has been shown to display both anti-inflammatory and anti-thrombotic actions in the systemic microvasculature. As inflammation promotes thrombosis and vice versa, in this study we investigated whether SFN is able to reduce inflammatory potentiation of thrombotic events, suppress platelet activation and thrombus formation in the cerebral microvasculature. EXPERIMENTAL APPROACH Thrombosis was induced in the murine brain using the light/dye-injury model, in conjunction with LPS treatment, with and without SFN treatment. In vitro and in vivo platelet assays (aggregation, flow and other functional tests) were also employed, using both human and murine platelets. KEY RESULTS SFN was found to reduce LPS-mediated enhancement of thrombus formation in the cerebral microcirculation. In tail-bleed experiments, LPS treatment prolonged bleeding time, and SFN treatment was found to protect against this LPS-induced derangement of platelet function. SFN inhibited collagen-mediated platelet aggregation in vitro and in vivo and the associated adhesion and impaired calcium signalling. Furthermore, glycoprotein VI was shown to be involved in the protective effects observed with SFN treatment. CONCLUSIONS AND IMPLICATIONS The data presented here provide evidence for the use of SFN in preventing stroke in selected high-risk patient cohorts.
Collapse
Affiliation(s)
| | - Paul M Holloway
- Division of Brain Sciences, Imperial College London, London, UK.,Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Felix Becker
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, Muenster, Germany
| | - Francesca Rauzi
- Platelet Biology Group, National Heart and Lung Institute, Imperial College London, London, UK
| | - Shantel A Vital
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Kirk A Taylor
- Platelet Biology Group, National Heart and Lung Institute, Imperial College London, London, UK
| | - Karen Y Stokes
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Michael Emerson
- Platelet Biology Group, National Heart and Lung Institute, Imperial College London, London, UK
| | - Felicity N E Gavins
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA.,Department of Neurology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| |
Collapse
|
20
|
Boulaftali Y, Mawhin M, Jandrot‐Perrus M, Ho‐Tin‐Noé B. Glycoprotein VI in securing vascular integrity in inflamed vessels. Res Pract Thromb Haemost 2018; 2:228-239. [PMID: 30046725 PMCID: PMC5974920 DOI: 10.1002/rth2.12092] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/08/2018] [Indexed: 12/12/2022] Open
Abstract
Glycoprotein VI (GPVI), the main platelet receptor for collagen, has been shown to play a central role in various models of thrombosis, and to be a minor actor of hemostasis at sites of trauma. These observations have made of GPVI a novel target for antithrombotic therapy, as its inhibition would ideally combine efficacy with safety. Nevertheless, recent studies have indicated that GPVI could play an important role in preventing bleeding caused by neutrophils in the inflamed skin and lungs. Remarkably, there is evidence that the GPVI-dependent hemostatic function of platelets at the acute phase of inflammation in these organs does not involve aggregation. From a therapeutic perspective, the vasculoprotective action of GPVI in inflammation suggests that blocking of GPVI might bear some risks of bleeding at sites of neutrophil infiltration. In this review, we summarize recent findings on GPVI functions in inflammation and discuss their possible clinical implications and applications.
Collapse
Affiliation(s)
- Yacine Boulaftali
- Laboratory of Vascular Translational ScienceSorbonne Paris CitéInstitut National de la Santé et de la Recherche Médicale (INSERM)Université Paris DiderotParisFrance
| | - Marie‐Anne Mawhin
- Laboratory of Vascular Translational ScienceSorbonne Paris CitéInstitut National de la Santé et de la Recherche Médicale (INSERM)Université Paris DiderotParisFrance
| | - Martine Jandrot‐Perrus
- Laboratory of Vascular Translational ScienceSorbonne Paris CitéInstitut National de la Santé et de la Recherche Médicale (INSERM)Université Paris DiderotParisFrance
| | - Benoît Ho‐Tin‐Noé
- Laboratory of Vascular Translational ScienceSorbonne Paris CitéInstitut National de la Santé et de la Recherche Médicale (INSERM)Université Paris DiderotParisFrance
| |
Collapse
|
21
|
Gardiner EE. Proteolytic processing of platelet receptors. Res Pract Thromb Haemost 2018; 2:240-250. [PMID: 30046726 PMCID: PMC6055504 DOI: 10.1002/rth2.12096] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 03/01/2018] [Indexed: 12/17/2022] Open
Abstract
Platelets have a major role in hemostasis and an emerging role in biological processes including inflammation and immunity. Many of these processes require platelet adhesion and localization at sites of tissue damage or infection and regulated platelet activation, mediated by platelet adheso-signalling receptors, glycoprotein (GP) Ib-IX-V and GPVI. Work from a number of laboratories has demonstrated that levels of these receptors are closely regulated by metalloproteinases of the A Disintegrin And Metalloproteinase (ADAM) family, primarily ADAM17 and ADAM10. It is becoming increasingly evident that platelets have important roles in innate immunity, inflammation, and in combating infection that extends beyond processes of hemostasis. This overview will examine the molecular events that regulate levels of platelet receptors and then assess ramifications for these events in settings where hemostasis, inflammation, and infection processes are triggered.
Collapse
Affiliation(s)
- Elizabeth E. Gardiner
- ACRF Department of Cancer Biology and TherapeuticsJohn Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| |
Collapse
|
22
|
Bergmeier W, Stefanini L. Platelets at the Vascular Interface. Res Pract Thromb Haemost 2018; 2:27-33. [PMID: 29457148 PMCID: PMC5810953 DOI: 10.1002/rth2.12061] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 10/19/2017] [Indexed: 02/01/2023] Open
Abstract
In this brief review paper, we will summarize the State-of-the-Art on how platelet reactivity is regulated in circulation and at sites of vascular injury. Our review discusses recent and ongoing work, presented at this year's International Society on Thrombosis and Haemostasis (ISTH) meeting, on the role of platelets in (1) classical hemostasis at sites of mechanical injury, and (2) the maintenance of vascular integrity at sites of inflammation.
Collapse
Affiliation(s)
- Wolfgang Bergmeier
- Department of Biochemistry and BiophysicsUniversity of North Carolina at Chapel HillChapel HillNCUSA
- McAllister Heart InstituteUniversity of North Carolina at Chapel HillChapel HillNCUSA
| | - Lucia Stefanini
- Department of Internal Medicine and Medical SpecialtiesSapienza University of RomeRomeItaly
| |
Collapse
|
23
|
Lee MY, Verni CC, Herbig BA, Diamond SL. Soluble fibrin causes an acquired platelet glycoprotein VI signaling defect: implications for coagulopathy. J Thromb Haemost 2017; 15:2396-2407. [PMID: 28981200 PMCID: PMC5716900 DOI: 10.1111/jth.13863] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Indexed: 11/27/2022]
Abstract
Essentials Collagen and thrombin when used simultaneously generate highly activated platelets. The effect of thrombin stimulation on subsequent glycoprotein VI (GPVI) function was observed. Soluble fibrin, but not protease activated receptor (PAR) activation, prevented GPVI activation. Circulating soluble fibrin in coagulopathic blood may cause an acquired GPVI signaling defect. SUMMARY Background In coagulopathic blood, circulating thrombin may drive platelet dysfunction. Methods/Results Using calcium dye-loaded platelets, the effect of thrombin exposure and soluble fibrin generation on subsequent platelet GPVI function was investigated. Exposure of apixaban-treated platelet-rich plasma (12% PRP) to thrombin (1-10 nm), but not ADP or thromboxane mimetic U46619 exposure, dramatically blocked subsequent GPVI activation by convulxin, collagen-related peptide or fibrillar collagen. Consistent with soluble fibrin multimerizing and binding GPVI, the onset of convulxin insensitivity required 200-500 s of thrombin exposure, was not mimicked by exposure to PAR-1/4 activating peptides, was not observed with washed platelets, and was blocked by fibrin polymerization inhibitor (GPRP) or factor XIIIa inhibitor (T101). PAR-1 signaling through Gαq was not required because vorapaxar blocked thrombin-induced calcium mobilization but had no effect on the ability of thrombin to impair GPVI-signaling. Convulxin insensitivity was unaffected by the metalloprotease inhibitor GM6001 or the αIIb β3 antagonist GR144053, indicating negligible roles for GPVI shedding or αIIb β3 binding of fibrin. Thrombin treatment of washed platelets resuspended in purified fibrinogen also produced convulxin insensitivity that was prevented by GPRP. Exposure of apixaban/PPACK-treated whole blood to thrombin-treated fibrinogen resulted in > 50% decrease in platelet deposition in a collagen microfluidic assay that required soluble fibrin assembly. Conclusions Conversion of only 1% plasma fibrinogen in coagulopathic blood would generate 90 nm soluble fibrin, far exceeding ~1 nmGPVI in blood. Soluble fibrin, rather than thrombin-induced platelet activation throuh PAR-1 and PAR-4, downregulated GPVI-signaling in response to stimuli, and may lead to subsequent hypofunction of endogenous or transfused platelets.
Collapse
Affiliation(s)
- Mei Yan Lee
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher C. Verni
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bradley A. Herbig
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott L. Diamond
- Department of Chemical and Biomolecular Engineering, Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
24
|
Jagadeeswaran P, Cooley BC, Gross PL, Mackman N. Animal Models of Thrombosis From Zebrafish to Nonhuman Primates: Use in the Elucidation of New Pathologic Pathways and the Development of Antithrombotic Drugs. Circ Res 2017; 118:1363-79. [PMID: 27126647 DOI: 10.1161/circresaha.115.306823] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/30/2015] [Indexed: 12/23/2022]
Abstract
Thrombosis is a leading cause of morbidity and mortality worldwide. Animal models are used to understand the pathological pathways involved in thrombosis and to test the efficacy and safety of new antithrombotic drugs. In this review, we will first describe the central role a variety of animal models of thrombosis and hemostasis has played in the development of new antiplatelet and anticoagulant drugs. These include the widely used P2Y12 antagonists and the recently developed orally available anticoagulants that directly target factor Xa or thrombin. Next, we will describe the new players, such as polyphosphate, neutrophil extracellular traps, and microparticles, which have been shown to contribute to thrombosis in mouse models, particularly venous thrombosis models. Other mouse studies have demonstrated roles for the factor XIIa and factor XIa in thrombosis. This has spurred the development of strategies to reduce their levels or activities as a new approach for preventing thrombosis. Finally, we will discuss the emergence of zebrafish as a model to study thrombosis and its potential use in the discovery of novel factors involved in thrombosis and hemostasis. Animal models of thrombosis from zebrafish to nonhuman primates are vital in identifying pathological pathways of thrombosis that can be safely targeted with a minimal effect on hemostasis. Future studies should focus on understanding the different triggers of thrombosis and the best drugs to prevent each type of thrombotic event.
Collapse
Affiliation(s)
- Pudur Jagadeeswaran
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.).
| | - Brian C Cooley
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.)
| | - Peter L Gross
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.)
| | - Nigel Mackman
- From the Department of Biological Sciences, University of North Texas, Denton (P.J.); Department of Pathology and Laboratory Medicine (B.C.C.), and Department of Medicine (N.M.), University of North Carolina, Chapel Hill; and Department of Medicine, McMaster University, Hamilton, Ontario, Canada (P.L.G.)
| |
Collapse
|
25
|
Xu XR, Zhang D, Oswald BE, Carrim N, Wang X, Hou Y, Zhang Q, Lavalle C, McKeown T, Marshall AH, Ni H. Platelets are versatile cells: New discoveries in hemostasis, thrombosis, immune responses, tumor metastasis and beyond. Crit Rev Clin Lab Sci 2016; 53:409-30. [PMID: 27282765 DOI: 10.1080/10408363.2016.1200008] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Platelets are small anucleate blood cells generated from megakaryocytes in the bone marrow and cleared in the reticuloendothelial system. At the site of vascular injury, platelet adhesion, activation and aggregation constitute the first wave of hemostasis. Blood coagulation, which is initiated by the intrinsic or extrinsic coagulation cascades, is the second wave of hemostasis. Activated platelets can also provide negatively-charged surfaces that harbor coagulation factors and markedly potentiate cell-based thrombin generation. Recently, deposition of plasma fibronectin, and likely other plasma proteins, onto the injured vessel wall has been identified as a new "protein wave of hemostasis" that may occur even earlier than the first wave of hemostasis, platelet accumulation. Although no experimental evidence currently exists, it is conceivable that platelets may also contribute to this protein wave of hemostasis by releasing their granule fibronectin and other proteins that may facilitate fibronectin self- and non-self-assembly on the vessel wall. Thus, platelets may contribute to all three waves of hemostasis and are central players in this critical physiological process to prevent bleeding. Low platelet counts in blood caused by enhanced platelet clearance and/or impaired platelet production are usually associated with hemorrhage. Auto- and allo-immune thrombocytopenias such as idiopathic thrombocytopenic purpura and fetal and neonatal alloimmune thrombocytopenia may cause life-threatening bleeding such as intracranial hemorrhage. When triggered under pathological conditions such as rupture of an atherosclerotic plaque, excessive platelet activation and aggregation may result in thrombosis and vessel occlusion. This may lead to myocardial infarction or ischemic stroke, the major causes of mortality and morbidity worldwide. Platelets are also involved in deep vein thrombosis and thromboembolism, another leading cause of mortality. Although fibrinogen has been documented for more than half a century as essential for platelet aggregation, recent studies demonstrated that fibrinogen-independent platelet aggregation occurs in both gene deficient animals and human patients under physiological and pathological conditions (non-anti-coagulated blood). This indicates that other unidentified platelet ligands may play important roles in thrombosis and might be novel antithrombotic targets. In addition to their critical roles in hemostasis and thrombosis, emerging evidence indicates that platelets are versatile cells involved in many other pathophysiological processes such as innate and adaptive immune responses, atherosclerosis, angiogenesis, lymphatic vessel development, liver regeneration and tumor metastasis. This review summarizes the current knowledge of platelet biology, highlights recent advances in the understanding of platelet production and clearance, molecular and cellular events of thrombosis and hemostasis, and introduces the emerging roles of platelets in the immune system, vascular biology and tumorigenesis. The clinical implications of these basic science and translational research findings will also be discussed.
Collapse
Affiliation(s)
- Xiaohong Ruby Xu
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada .,b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,c Department of Medicine , Guangzhou University of Chinese Medicine , Guangzhou , Guangdong , P.R. China
| | - Dan Zhang
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,c Department of Medicine , Guangzhou University of Chinese Medicine , Guangzhou , Guangdong , P.R. China
| | - Brigitta Elaine Oswald
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,d Canadian Blood Services , Toronto , ON , Canada .,e Department of Physiology , University of Toronto , Toronto , ON , Canada
| | - Naadiya Carrim
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada .,b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,d Canadian Blood Services , Toronto , ON , Canada
| | - Xiaozhong Wang
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,f The Second Affiliated Hospital of Nanchang University , Nanchang , Jiangxi , P.R. China
| | - Yan Hou
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,g Jilin Provincial Center for Disease Prevention and Control , Changchun , Jilin , P.R. China
| | - Qing Zhang
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,h State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University , Guangzhou , Guangdong , P.R. China , and
| | - Christopher Lavalle
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,e Department of Physiology , University of Toronto , Toronto , ON , Canada
| | - Thomas McKeown
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada
| | - Alexandra H Marshall
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada
| | - Heyu Ni
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada .,b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,d Canadian Blood Services , Toronto , ON , Canada .,e Department of Physiology , University of Toronto , Toronto , ON , Canada .,i Department of Medicine , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
26
|
Covarrubias R, Chepurko E, Reynolds A, Huttinger ZM, Huttinger R, Stanfill K, Wheeler DG, Novitskaya T, Robson SC, Dwyer KM, Cowan PJ, Gumina RJ. Role of the CD39/CD73 Purinergic Pathway in Modulating Arterial Thrombosis in Mice. Arterioscler Thromb Vasc Biol 2016; 36:1809-20. [PMID: 27417582 DOI: 10.1161/atvbaha.116.307374] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/29/2016] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Circulating blood cells and endothelial cells express ectonucleoside triphosphate diphosphohydrolase-1 (CD39) and ecto-5'-nucleotidase (CD73). CD39 hydrolyzes extracellular ATP or ADP to AMP. CD73 hydrolyzes AMP to adenosine. The goal of this study was to examine the interplay between CD39 and CD73 cascade in arterial thrombosis. APPROACH AND RESULTS To determine how CD73 activity influences in vivo thrombosis, the time to ferric chloride-induced arterial thrombosis was measured in CD73-null mice. In response to 5% FeCl3, but not to 10% FeCl3, there was a significant decrease in the time to thrombosis in CD73-null mice compared with wild-type mice. In mice overexpressing CD39, ablation of CD73 did not inhibit the prolongation in the time to thrombosis conveyed by CD39 overexpression. However, the CD73 inhibitor α-β-methylene-ADP nullified the prolongation in the time to thrombosis in human CD39 transgenic (hC39-Tg)/CD73-null mice. To determine whether hematopoietic-derived cells or endothelial cell CD39 activity regulates in vivo arterial thrombus, bone marrow transplant studies were conducted. FeCl3-induced arterial thrombosis in chimeric mice revealed a significant prolongation in the time to thrombosis in hCD39-Tg reconstituted wild-type mice, but not on wild-type reconstituted hCD39-Tg mice. Monocyte depletion with clodronate-loaded liposomes normalized the time to thrombosis in hCD39-Tg mice compared with hCD39-Tg mice treated with control liposomes, demonstrating that increased CD39 expression on monocytes protects against thrombosis. CONCLUSIONS These data demonstrate that ablation of CD73 minimally effects in vivo thrombosis, but increased CD39 expression on hematopoietic-derived cells, especially monocytes, attenuates in vivo arterial thrombosis.
Collapse
Affiliation(s)
- Roman Covarrubias
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Elena Chepurko
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Adam Reynolds
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Zachary M Huttinger
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Ryan Huttinger
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Katherine Stanfill
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Debra G Wheeler
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Tatiana Novitskaya
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Simon C Robson
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Karen M Dwyer
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Peter J Cowan
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.)
| | - Richard J Gumina
- From the Division of Cardiovascular Medicine, Department of Medicine (R.C., E.C., T.N., R.J.G.), Department of Pharmacology (R.J.G.), and Department of Pathology Microbiology and Immunology (R.J.G.), Vanderbilt University, Nashville, TN; Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus (A.R., Z.M.H., R.H., K.S., D.G.W.); Transplant Institute, Department of Medicine, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA (S.C.R.); School of Medicine, Deakin University (K.M.D., P.J.C.); Immunology Research Centre, St. Vincent's Hospital (K.M.D.); and Department of Medicine, University of Melbourne, Victoria, Australia (K.M.D., P.J.C.).
| |
Collapse
|
27
|
|
28
|
Abstract
Collagens mediate essential hemostasis by maintaining the integrity and stability of the vascular wall. Imbalanced turnover of collagens by uncontrolled formation and/or degradation may result in pathologic conditions such as fibrosis. Thickening of the vessel wall because of accumulation of collagens may lead to arterial occlusion or thrombosis. Thinning of the wall because of collagen degradation or deficiency may lead to rupture of the vessel wall or aneurysm. Preventing excessive hemorrhage or thrombosis relies on collagen-mediated actions. Von Willebrand factor, integrins and glycoprotein VI, as well as clotting factors, can bind collagen to restore normal hemostasis after trauma. This review outlines the essential roles of collagens in mediating hemostasis, with a focus on collagens types I, III, IV, VI, XV, and XVIII.
Collapse
Affiliation(s)
| | - N G Kjeld
- Nordic Bioscience A/S, Herlev, Denmark
| | | |
Collapse
|
29
|
Ivanciu L, Stalker TJ. Spatiotemporal regulation of coagulation and platelet activation during the hemostatic response in vivo. J Thromb Haemost 2015; 13:1949-59. [PMID: 26386264 PMCID: PMC5847271 DOI: 10.1111/jth.13145] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 08/29/2015] [Indexed: 12/17/2022]
Abstract
The hemostatic response requires the tightly regulated interaction of the coagulation system, platelets, other blood cells and components of the vessel wall at a site of vascular injury. The dysregulation of this response may result in excessive bleeding if the response is impaired, and pathologic thrombosis with vessel occlusion and tissue ischemia if the response is overly robust. Extensive studies over the past decade have sought to unravel the regulatory mechanisms that coordinate the multiple biochemical and cellular responses in time and space to ensure that an optimal response to vascular damage is achieved. These studies have relied in part on advances in in vivo imaging techniques in animal models, allowing for the direct visualization of various molecular and cellular events in real time during the hemostatic response. This review summarizes knowledge gained with these in vivo imaging and other approaches that provides new insights into the spatiotemporal regulation of coagulation and platelet activation at a site of vascular injury.
Collapse
Affiliation(s)
- L Ivanciu
- Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - T J Stalker
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
30
|
Westrick R, Fredman G. Platelets: Context-Dependent Vascular Protectors or Mediators of Disease. Arterioscler Thromb Vasc Biol 2015; 35:e25-9. [PMID: 26109740 DOI: 10.1161/atvbaha.115.305898] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Randal Westrick
- From the Department of Biological Sciences, Oakland University, Rochester, MI (R.W.); and Department of Medicine, Columbia University, New York, NY (G.F.)
| | - Gabrielle Fredman
- From the Department of Biological Sciences, Oakland University, Rochester, MI (R.W.); and Department of Medicine, Columbia University, New York, NY (G.F.).
| | | |
Collapse
|
31
|
Fibrin activates GPVI in human and mouse platelets. Blood 2015; 126:1601-8. [PMID: 26282541 DOI: 10.1182/blood-2015-04-641654] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/11/2015] [Indexed: 01/09/2023] Open
Abstract
The glycoprotein VI (GPVI)-Fc receptor γ (FcRγ) chain is the major platelet signaling receptor for collagen. Paradoxically, in a FeCl3 injury model, occlusion, but not initiation of thrombus formation, is delayed in GPVI-deficient and GPVI-depleted mice. In this study, we demonstrate that GPVI is a receptor for fibrin and speculate that this contributes to development of an occlusive thrombus. We observed a marked increase in tyrosine phosphorylation, including the FcRγ chain and Syk, in human and mouse platelets induced by thrombin in the presence of fibrinogen and the αIIbβ3 blocker eptifibatide. This was not seen in platelets stimulated by a protease activated receptor (PAR)-4 peptide, which is unable to generate fibrin from fibrinogen. The pattern of tyrosine phosphorylation was similar to that induced by activation of GPVI. Consistent with this, thrombin did not induce tyrosine phosphorylation of Syk and the FcRγ chain in GPVI-deficient mouse platelets. Mouse platelets underwent full spreading on fibrin but not fibrinogen, which was blocked in the presence of a Src kinase inhibitor or in the absence of GPVI. Spreading on fibrin was associated with phosphatidylserine exposure (procoagulant activity), and this too was blocked in GPVI-deficient platelets. The ectodomain of GPVI was shown to bind to immobilized monomeric and polymerized fibrin. A marked increase in embolization was seen following FeCl3 injury in GPVI-deficient mice, likely contributing to the delay in occlusion in this model. These results demonstrate that GPVI is a receptor for fibrin and provide evidence that this interaction contributes to thrombus growth and stability.
Collapse
|