1
|
Din M, Paul S, Ullah S, Yang H, Xu RG, Abidin NAZ, Sun A, Chen YC, Gao R, Chowdhury B, Zhou F, Rogers S, Miller M, Biswas A, Hu L, Fan Z, Zahner C, Fan J, Chen Z, Berman M, Xue L, Ju LA, Chen Y. Multi-parametric thrombus profiling microfluidics detects intensified biomechanical thrombogenesis associated with hypertension and aging. Nat Commun 2024; 15:9067. [PMID: 39433750 PMCID: PMC11494109 DOI: 10.1038/s41467-024-53069-9] [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: 01/03/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024] Open
Abstract
Arterial thrombosis is a leading cause of death and disability worldwide with no effective bioassay for clinical prediction. As a symbolic feature of arterial thrombosis, severe stenosis in the blood vessel creates a high-shear, high-gradient flow environment that facilitates platelet aggregation towards vessel occlusion. Here, we present a thrombus profiling assay that monitors the multi-dimensional attributes of thrombi forming in such biomechanical conditions. Using this assay, we demonstrate that different receptor-ligand interactions contribute distinctively to the composition and activation status of the thrombus. Our investigation into hypertensive and older individuals reveals intensified biomechanical thrombogenesis and multi-dimensional thrombus profile abnormalities, endorsing the diagnostic potential of the assay. Furthermore, we identify the hyperactivity of GPIbα-integrin αIIbβ3 mechanosensing axis as a molecular mechanism that contributes to hypertension-associated arterial thrombosis. By studying drug-disease interactions and inter-individual variability, our work reveals a need for personalized anti-thrombotic drug selection that accommodates each patient's pathological profile.
Collapse
Affiliation(s)
- Misbahud Din
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Souvik Paul
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Sana Ullah
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Haoyi Yang
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania, PA, 16802, USA
| | - Rong-Guang Xu
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Division of Thoracic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Allan Sun
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Newtown, NSW, 2042, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Yiyao Catherine Chen
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Rui Gao
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Bari Chowdhury
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Fangyuan Zhou
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Stephenie Rogers
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Mariel Miller
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Atreyee Biswas
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Liang Hu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, 06030, USA
| | - Christopher Zahner
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jing Fan
- Department of Mechanical Engineering, The City University of New York - City College, New York, NY, 10031, USA
| | - Zi Chen
- Division of Thoracic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Megan Berman
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Lingzhou Xue
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania, PA, 16802, USA
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Newtown, NSW, 2042, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Yunfeng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
2
|
Lira AL, Kohs TC, Moellmer SA, Shatzel JJ, McCarty OJ, Puy C. Substrates, Cofactors, and Cellular Targets of Coagulation Factor XIa. Semin Thromb Hemost 2024; 50:962-969. [PMID: 36940715 PMCID: PMC11069399 DOI: 10.1055/s-0043-1764469] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023]
Abstract
Coagulation factor XI (FXI) has increasingly been shown to play an integral role in several physiologic and pathological processes. FXI is among several zymogens within the blood coagulation cascade that are activated by proteolytic cleavage, with FXI converting to the active serine protease form (FXIa). The evolutionary origins of FXI trace back to duplication of the gene that transcribes plasma prekallikrein, a key factor in the plasma kallikrein-kinin system, before further genetic divergence led to FXI playing a unique role in blood coagulation. While FXIa is canonically known for activating the intrinsic pathway of coagulation by catalyzing the conversion of FIX into FIXa, it is promiscuous in nature and has been shown to contribute to thrombin generation independent of FIX. In addition to its role in the intrinsic pathway of coagulation, FXI also interacts with platelets, endothelial cells, and mediates the inflammatory response through activation of FXII and cleavage of high-molecular-weight kininogen to generate bradykinin. In this manuscript, we critically review the current body of knowledge surrounding how FXI navigates the interplay of hemostasis, inflammatory processes, and the immune response and highlight future avenues for research. As FXI continues to be clinically explored as a druggable therapeutic target, understanding how this coagulation factor fits into physiological and disease mechanisms becomes increasingly important.
Collapse
Affiliation(s)
- André L. Lira
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Tia C.L. Kohs
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Samantha A. Moellmer
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Joseph J. Shatzel
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Owen J.T. McCarty
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Cristina Puy
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon
- Divison of Hematology and Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
3
|
Puspitasari YM, Ministrini S, Han J, Karch C, Prisco F, Liberale L, Bengs S, Akhmedov A, Montecucco F, Beer JH, Lüscher TF, Bongiovanni D, Camici GG. Hutchinson-Gilford progeria syndrome mice display accelerated arterial thrombus formation and increased platelet reactivity. Thromb Res 2024; 241:109100. [PMID: 39032390 DOI: 10.1016/j.thromres.2024.109100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
INTRODUCTION Hutchinson-Gilford Progeria Syndrome (HGPS) is an ultra-rare premature aging genetic disorder caused by a point mutation in the lamin A gene, LMNA. Children with HGPS display short lifespans and typically die due to myocardial infarction or ischemic stroke, both acute cardiovascular events that are tightly linked to arterial thrombosis. Despite this fact, the effect of the classic HGPS LMNA gene mutation on arterial thrombosis remains unknown. METHODS Heterozygous LmnaG609G knock-in (LmnaG609G/+) mice, yielding an equivalent classic mutation observed in HGPS patients (c.1824C>T; pG608G mutation in the human LMNA gene) and corresponding wild-type (WT) control littermates underwent photochemically laser-induced carotid injury to trigger thrombosis. Coagulation and fibrinolytic factors were measured. Furthermore, platelet activation and reactivity were investigated. RESULTS LmnaG609G/+ mice displayed accelerated arterial thrombus formation, as underlined by shortened time to occlusion compared to WT littermates. Levels of factors involved in the coagulation and fibrinolytic system were comparable between groups, while LmnaG609G/+ animals showed higher plasma levels of thrombin-antithrombin complex and lower levels of antithrombin. Bone marrow analysis showed larger megakaryocytes in progeric mice. Lastly, enhanced platelet activation upon adenosine diphosphate, collagen-related peptide, and thrombin stimulation was observed in LmnaG609G/+ animals compared to the WT group, indicating a higher platelet reactivity in progeric animals. CONCLUSIONS LMNA mutation in HGPS mice accelerates arterial thrombus formation, which is mediated, at least in part, by enhanced platelet reactivity, which consequently augments thrombin generation. Given the wide spectrum of antiplatelet agents available clinically, further investigation is warranted to consider the most suitable antiplatelet regimen for children with HGPS to mitigate disease mortality and morbidity.
Collapse
Affiliation(s)
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Jiaying Han
- Department of Internal Medicine I, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Caroline Karch
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Francesco Prisco
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Susan Bengs
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Cardiology, Royal Brompton & Harefield Hospitals, National Heart & Lung Institute, Imperial College, London, United Kingdom
| | - Dario Bongiovanni
- Department of Internal Medicine I, Cardiology, University Hospital Augsburg, University of Augsburg, Augsburg, Germany; Department of Cardiovascular Medicine, Humanitas Clinical and Research Center IRCCS and Humanitas University, Rozzano, Milan, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland; Department of Research and Education, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
4
|
Li X, Li S, Zhang W, Wang Q, Zou W. Impacts of P4-ATPase Deletion on Membrane Asymmetry and Disease Development. Cell Biochem Funct 2024; 42:e70004. [PMID: 39425455 DOI: 10.1002/cbf.70004] [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: 06/11/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
Phospholipids exhibit an asymmetrical distribution on the cell membrane. P4-ATPases, type IV lipid flippases, are responsible for establishing and maintaining this phospholipid compositional asymmetry. The essential β subunit CDC50 (also known as TMEM30) assists in the transport and proper functioning of P4-ATPases. Deletion of P4-ATPases and its β subunit disrupts the membrane asymmetry, impacting the growth and development and leading to various diseases affecting the nervous, skeletal muscle, digestive, and hematopoietic systems. This review discusses the crucial roles of P4-ATPases and their β subunit in Saccharomyces cerevisiae, Arabidopsis thaliana, Caenorhabditis elegans, and mammals, offering valuable insights for future research.
Collapse
Affiliation(s)
- Xinyu Li
- School of Public Health, Kunming Medical University, Kunming, China
| | - Shuzhen Li
- School of Public Health, Kunming Medical University, Kunming, China
| | - Weipu Zhang
- School of Public Health, Kunming Medical University, Kunming, China
| | - Qi Wang
- School of Public Health, Kunming Medical University, Kunming, China
| | - Wei Zou
- School of Public Health, Kunming Medical University, Kunming, China
| |
Collapse
|
5
|
Din M, Paul S, Ullah S, Yang H, Xu RG, Abidin NAZ, Sun A, Chen YC, Gao R, Chowdhury B, Zhou F, Rogers S, Miller M, Biswas A, Hu L, Fan Z, Zahner C, Fan J, Chen Z, Berman M, Xue L, Ju LA, Chen Y. Multi-parametric thrombus profiling microfluidics detects intensified biomechanical thrombogenesis associated with hypertension and aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598290. [PMID: 38915705 PMCID: PMC11195082 DOI: 10.1101/2024.06.11.598290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Arterial thrombosis, which represents a critical complication of cardiovascular diseases, is a leading cause of death and disability worldwide with no effective bioassay for clinical prediction. As a symbolic feature of arterial thrombosis, severe stenosis in the blood vessel creates a high-shear, high-gradient flow environment that effectively facilitates platelet aggregation towards vessel occlusion even with platelet amplification loops inhibited. However, no approach is currently available to comprehensively characterize the size, composition and platelet activation status of thrombi forming under this biorheological condition. Here, we present a thrombus profiling assay that monitors the multi-dimensional attributes of thrombi forming in conditions mimicking the physiological scenario of arterial thrombosis. Using this platform, we demonstrate that different receptor-ligand interactions contribute distinctively to the composition and activation status of the thrombus. Our investigation into hypertensive and older individuals reveals intensified biomechanical thrombogenesis and multi-dimensional thrombus profile abnormalities, demonstrating a direct contribution of mechanobiology to arterial thrombosis and endorsing the diagnostic potential of the assay. Furthermore, we identify the hyperactivity of GPIbα-integrin αIIbβ3 mechanosensing axis as a molecular mechanism that contributes to hypertension-associated arterial thrombosis. By studying the interactions between anti-thrombotic inhibitors and hypertension, and the inter-individual variability in personal thrombus profiles, our work reveals a critical need for personalized anti-thrombotic drug selection that accommodates each patient's pathological profile.
Collapse
Affiliation(s)
- Misbahud Din
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Souvik Paul
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Sana Ullah
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Haoyi Yang
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Rong-Guang Xu
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Division of Thoracic Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | - Allan Sun
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW 2042, Australia
| | - Yiyao Catherine Chen
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
| | - Rui Gao
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
| | - Bari Chowdhury
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Fangyuan Zhou
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Stephenie Rogers
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Mariel Miller
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Atreyee Biswas
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Liang Hu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut 06030, USA
| | - Christopher Zahner
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Jing Fan
- Department of Mechanical Engineering, The City University of New York - City College, New York, New York 10031, USA
| | - Zi Chen
- Division of Thoracic Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Megan Berman
- Department of Internal Medicine, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Lingzhou Xue
- Department of Statistics, The Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW 2042, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2006, Australia
| | - Yunfeng Chen
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas 77555, USA
| |
Collapse
|
6
|
Guo J, Cui B, Zheng J, Yu C, Zheng X, Yi L, Zhang S, Wang K. Platelet-derived microparticles and their cargos: The past, present and future. Asian J Pharm Sci 2024; 19:100907. [PMID: 38623487 PMCID: PMC11016590 DOI: 10.1016/j.ajps.2024.100907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/05/2024] [Accepted: 01/31/2024] [Indexed: 04/17/2024] Open
Abstract
All eukaryotic cells can secrete extracellular vesicles, which have a double-membrane structure and are important players in the intercellular communication involved in a variety of important biological processes. Platelets form platelet-derived microparticles (PMPs) in response to activation, injury, or apoptosis. This review introduces the origin, pathway, and biological functions of PMPs and their importance in physiological and pathological processes. In addition, we review the potential applications of PMPs in cancer, vascular homeostasis, thrombosis, inflammation, neural regeneration, biomarkers, and drug carriers to achieve targeted drug delivery. In addition, we comprehensively report on the origin, biological functions, and applications of PMPs. The clinical transformation, high heterogeneity, future development direction, and limitations of the current research on PMPs are also discussed in depth. Evidence has revealed that PMPs play an important role in cell-cell communication, providing clues for the development of PMPs as carriers for relevant cell-targeted drugs. The development history and prospects of PMPs and their cargos are explored in this guidebook.
Collapse
Affiliation(s)
- Jingwen Guo
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Bufeng Cui
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Jie Zheng
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Chang Yu
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xuran Zheng
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Lixin Yi
- School of Pharmacy, China Medical University, Shenyang 110122, China
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Simeng Zhang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Keke Wang
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
7
|
Weaver AJ, McIntosh CS, Kelly SG, Barrera GD, Lizarraga S, Hildreth KE, Williams CE, Grantham L, Yoshida T, Omert L, Bynum JA, Meledeo MA, Reddoch-Cardenas KM. Evaluating the effects of hypoxic storage on platelet function and health using a novel storage system. Transfusion 2024; 64:693-704. [PMID: 38511850 DOI: 10.1111/trf.17784] [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: 09/16/2023] [Revised: 01/14/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Thousands of units of whole blood (WB) and blood components are transfused daily to treat trauma patients. Improved methods for blood storage are critical to support trauma-related care. The Hemanext ONE® system offers a unique method for hypoxic storage of WB, with successfully demonstrated storage of clinically viable RBCs. This work evaluated the system for the storage of WB, focusing on platelet health and function. STUDY DESIGN AND METHODS WB was collected from healthy donors and processed through the Hemanext ONE® system. Hemoglobin oxygen saturation (HbSO2) levels of WB were depleted to 10%, 20%, or 30% of total HbSO2 and then stored in PVC bags sealed in oxygen-impermeable bags (except for normoxic control) with samples collected on days 1, 7, and 14 post-processing. Flow cytometry assessed the activation and apoptosis of platelets. Clot dynamics were assessed based on aggregometry and thromboelastography assays, as well as thrombin generation using a calibrated-automated thrombogram method. RESULTS Hypoxic storage conditions were maintained throughout the storage period. Hypoxia triggered increased lactate production, but pH changes were negligible compared to normoxic control. Storage at 10% HbSO2 had a significant impact on platelet function, resulting in increased activation and reduced clot formation and aggregation. These effects were less significant at 20% and 30% HbSO2. DISCUSSION This study indicates that platelets are sensitive to hypoxic storage and suffer significant metabolic and functional deterioration when stored at or below 10% HbSO2.
Collapse
Affiliation(s)
- A J Weaver
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - C S McIntosh
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - S G Kelly
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - G D Barrera
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - S Lizarraga
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - K E Hildreth
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - C E Williams
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - L Grantham
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - T Yoshida
- Hemanext Inc., Lexington, Massachusetts, USA
| | - L Omert
- Hemanext Inc., Lexington, Massachusetts, USA
| | | | - M A Meledeo
- United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | | |
Collapse
|
8
|
Fernández-Infante C, Hernández-Cano L, Herranz Ó, Berrocal P, Sicilia-Navarro C, González-Porras JR, Bastida JM, Porras A, Guerrero C. Platelet C3G: a key player in vesicle exocytosis, spreading and clot retraction. Cell Mol Life Sci 2024; 81:84. [PMID: 38345631 PMCID: PMC10861696 DOI: 10.1007/s00018-023-05109-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 02/15/2024]
Abstract
C3G is a Rap1 GEF that plays a pivotal role in platelet-mediated processes such as angiogenesis, tumor growth, and metastasis by modulating the platelet secretome. Here, we explore the mechanisms through which C3G governs platelet secretion. For this, we utilized animal models featuring either overexpression or deletion of C3G in platelets, as well as PC12 cell clones expressing C3G mutants. We found that C3G specifically regulates α-granule secretion via PKCδ, but it does not affect δ-granules or lysosomes. C3G activated RalA through a GEF-dependent mechanism, facilitating vesicle docking, while interfering with the formation of the trans-SNARE complex, thereby restricting vesicle fusion. Furthermore, C3G promotes the formation of lamellipodia during platelet spreading on specific substrates by enhancing actin polymerization via Src and Rac1-Arp2/3 pathways, but not Rap1. Consequently, C3G deletion in platelets favored kiss-and-run exocytosis. C3G also controlled granule secretion in PC12 cells, including pore formation. Additionally, C3G-deficient platelets exhibited reduced phosphatidylserine exposure, resulting in decreased thrombin generation, which along with defective actin polymerization and spreading, led to impaired clot retraction. In summary, platelet C3G plays a dual role by facilitating platelet spreading and clot retraction through the promotion of outside-in signaling while concurrently downregulating α-granule secretion by restricting granule fusion.
Collapse
Affiliation(s)
- Cristina Fernández-Infante
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC, Centro de Investigación del Cáncer, Campus Unamuno S/N, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Luis Hernández-Cano
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC, Centro de Investigación del Cáncer, Campus Unamuno S/N, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Óscar Herranz
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC, Centro de Investigación del Cáncer, Campus Unamuno S/N, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Pablo Berrocal
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC, Centro de Investigación del Cáncer, Campus Unamuno S/N, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Carmen Sicilia-Navarro
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC, Centro de Investigación del Cáncer, Campus Unamuno S/N, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - José Ramón González-Porras
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - José María Bastida
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Almudena Porras
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Ciudad Universitaria, Madrid, Spain.
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - Carmen Guerrero
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC, Centro de Investigación del Cáncer, Campus Unamuno S/N, Salamanca, Spain.
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain.
| |
Collapse
|
9
|
You W, Knoops K, Boesten I, Berendschot TTJM, van Zandvoort MAMJ, Benedikter BJ, Webers CAB, Reutelingsperger CPM, Gorgels TGMF. A time window for rescuing dying retinal ganglion cells. Cell Commun Signal 2024; 22:88. [PMID: 38297331 PMCID: PMC10832163 DOI: 10.1186/s12964-023-01427-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/08/2023] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Retinal ganglion cell (RGC) degeneration and death cause vision loss in patients with glaucoma. Regulated cell death, once initiated, is generally considered to be an irreversible process. Recently, we showed that, by timely removing the cell death stimulus, stressed neuronal PC12 cells can recover from phosphatidylserine (PS) exposure, nuclear shrinkage, DNA damage, mitochondrial fragmentation, mitochondrial membrane potential loss, and retraction of neurites, all hallmarks of an activated cell death program. Whether the cell death process can be reversed in neurons of the central nervous system, like RGCs, is still unknown. Here, we studied reversibility of the activated cell death program in primary rat RGCs (prRGCs). METHODS prRGCs were exposed to ethanol (5%, vol/vol) to induce cell death. At different stages of the cell death process, ethanol was removed by washing and injured prRGCs were further cultured in fresh medium to see whether they recovered. The dynamics of single cells were monitored by high-resolution live-cell spinning disk microscopy. PS exposure, mitochondrial structure, membrane potential, and intracellular Ca2+ were revealed by annexin A5-FITC, Mito-tracker, TMRM, and Fluo 8-AM staining, respectively. The distribution of cytochrome c was investigated by immunofluorescence. The ultrastructure of mitochondria was studied by electron microscopy. RESULTS Analysis of temporal relationships between mitochondrial changes and PS exposure showed that fragmentation of the mitochondrial network and loss of mitochondrial membrane potential occurred before PS exposure. Mitochondrial changes proceeded caspase-independently, while PS exposure was caspase dependent. Interestingly, prRGCs recovered quickly from these mitochondrial changes but not from PS exposure at the plasma membrane. Correlative light and electron microscopy showed that stress-induced decrease in mitochondrial area, length and cristae number was reversible. Intracellular Ca2+ was elevated during this stage of reversible mitochondrial injury, but there was no sign of mitochondrial cytochrome c release. CONCLUSIONS Our study demonstrates that RGCs with impaired mitochondrial structure and function can fully recover if there is no mitochondrial cytochrome c release yet, and no PS is exposed at the plasma membrane. This finding indicates that there is a time window for rescuing dying or injured RGCs, by simply removing the cell death stimulus. Video Abstract.
Collapse
Affiliation(s)
- Wenting You
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
- Department of Biochemistry, CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, 6229 ER, The Netherlands
- Department of Mental Health and Neuroscience, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Kèvin Knoops
- The Microscopy CORE lab, Maastricht Multimodal Molecular Imaging Institute, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Iris Boesten
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
| | - Tos T J M Berendschot
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
| | - Marc A M J van Zandvoort
- Department of Molecular Cell Biology, CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, 6229 ER, The Netherlands
- Institute of Molecular Cardiovascular Research (IMCAR), Universitätsklinikum Aachen, 52074, Aachen, Germany
| | - Birke J Benedikter
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
| | - Carroll A B Webers
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
| | - Chris P M Reutelingsperger
- Department of Biochemistry, CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, 6229 ER, The Netherlands.
| | - Theo G M F Gorgels
- University Eye Clinic Maastricht UMC+, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands.
| |
Collapse
|
10
|
De Paoli SH, Patel M, Elhelu OK, Tarandovskiy ID, Tegegn TZ, Simak J. Structural analysis of platelet fragments and extracellular vesicles produced by apheresis platelets during storage. Blood Adv 2024; 8:207-218. [PMID: 37967384 PMCID: PMC10787271 DOI: 10.1182/bloodadvances.2023011325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/01/2023] [Accepted: 11/04/2023] [Indexed: 11/17/2023] Open
Abstract
ABSTRACT Platelets (PLTs) for transfusion can be stored for up to 7 days at room temperature (RT). The quality of apheresis PLTs decreases over storage time, which affects PLT hemostatic functions. Here, we characterized the membranous particles produced by PLT storage lesion (PSLPs), including degranulated PLTs, PLT ghosts, membrane fragments, and extracellular membrane vesicles (PEVs). The PSLPs generated in apheresis platelet units were analyzed on days 1, 3, 5, and 7 of RT storage. A differential centrifugation and a sucrose density gradient were used to separate PSLP populations. PSLPs were characterized using scanning and transmission electron microscopy (EM), flow cytometry (FC), and nanoparticle tracking analysis (NTA). PSLPs have different morphologies and a broad size distribution; FC and NTA showed that the concentration of small and large PSLPs increases with storage time. The density gradient separated 3 PSLP populations: (1) degranulated PLTs, PLT ghosts, and large PLT fragments; (2) PEVs originated from PLT activation and organelles released by necrotic PLTs; and (3) PEV ghosts. Most PSLPs expressed phosphatidyl serine and induced thrombin generation in the plasma. PSLPs contained extracellular mitochondria and some had the autophagosome marker LC3. PSLPs encompass degranulated PLTs, PLT ghosts, large PLT fragments, large and dense PEVs, and low-density PEV ghosts. The activation-related PSLPs are released, particularly during early stage of storage (days 1-3), and the release of apoptosis- and necrosis-related PSLPs prevails after that. No elevation of LC3- and TOM20-positive PSLPs indicates that the increase of extracellular mitochondria during later-stage storage is not associated with PLT mitophagy.
Collapse
Affiliation(s)
- Silvia H De Paoli
- Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Mehulkumar Patel
- Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, MD
| | - Oumsalama K Elhelu
- Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Ivan D Tarandovskiy
- Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
- Hemostasis Branch, Office of Therapeutic Products, Center of Biologics Evaluations and Research, US Food and Drug Administration, Silver Spring, MD
| | - Tseday Z Tegegn
- Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Jan Simak
- Laboratory of Cellular Hematology, Office of Blood Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| |
Collapse
|
11
|
Choi JH, Kim K. Polyhexamethylene Guanidine Phosphate Enhanced Procoagulant Activity through Oxidative-Stress-Mediated Phosphatidylserine Exposure in Platelets. TOXICS 2024; 12:50. [PMID: 38251006 PMCID: PMC10820372 DOI: 10.3390/toxics12010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024]
Abstract
Polyhexamethylene guanidine phosphate (PHMG-p) is a common biocidal disinfectant that is widely used in industry and household products. However, PHMG-p was misused as a humidifier disinfectant (HD) in South Korea, which had fatal health effects. Various health problems including cardiovascular diseases were observed in HD-exposed groups. However, the potential underlying mechanism of HD-associated cardiovascular diseases is poorly understood. Here, we examined the procoagulant activity of platelets caused by PHMG-p and clarified the underlying mechanism. PHMG-p enhanced phosphatidylserine (PS) exposure through alteration of phospholipid transporters, scramblase, and flippase. Intracellular calcium elevation, intracellular ATP depletion, and caspase-3 activation appeared to underlie phospholipid transporter dysregulation caused by PHMG-p, which was mediated by oxidative stress and mitochondrial dysfunction. Notably, antioxidant enzyme catalase and calcium chelator EGTA reversed PHMG-p-induced PS exposure and thrombin generation, confirming the contributive role of oxidative stress and intracellular calcium in the procoagulant effects of PHMG-p. These series of events led to procoagulant activation of platelets, which was revealed as enhanced thrombin generation. Collectively, PHMG-p triggered procoagulant activation of platelets, which may promote prothrombotic risks and cardiovascular diseases. These findings improve our understanding of HD-associated cardiovascular diseases.
Collapse
Affiliation(s)
| | - Keunyoung Kim
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea;
| |
Collapse
|
12
|
Tynngård N, Alshamari A, Sandgren P, Kenny D, Vasilache AM, Abedi MR, Ramström S. High fragmentation in platelet concentrates impacts the activation, procoagulant, and aggregatory capacity of platelets. Platelets 2023; 34:2159018. [PMID: 36632714 DOI: 10.1080/09537104.2022.2159018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Platelets are transfused to patients to prevent bleeding. Since both preparation and storage can impact the hemostatic functions of platelets, we studied platelet concentrates (PCs) with different initial composition in regard to platelet fragmentation and its impact on storage-induced changes in activation potential. Ten whole blood derived PCs were assessed over 7 storage days. Using flow cytometry, platelet (CD41+) subpopulations were characterized for activation potential using activation markers (PAC-1, P-selectin, and LAMP-1), phosphatidylserine (Annexin V), and mitochondrial integrity (DiIC1(5)). Aggregation response, coagulation, and soluble activation markers (cytokines and sGPVI) were also measured. Of the CD41+ events, the PCs contained a median of 82% normal-sized platelets, 10% small platelets, and 8% fragments. The small platelets exhibited procoagulant hallmarks (increased P-selectin and Annexin V and reduced DiIC1(5)). Normal-sized platelets responded to activation, whereas activation potential was decreased for small and abolished for fragments. Five PCs contained a high proportion of small platelets and fragments (median of 28% of CD41+ events), which was significantly higher than the other five PCs (median of 9%). A high proportion of small platelets and fragments was associated with procoagulant hallmarks and decreased activation potential, but, although diminished, they still retained some activation potential throughout 7 days storage.
Collapse
Affiliation(s)
- Nahreen Tynngård
- Department of Clinical Chemistry and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Research and Development Unit in Region Östergötland and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Aseel Alshamari
- Department of Clinical Immunology and Transfusion medicine, Faculty of Medicine and Health, Örebro University, Sweden.,Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Per Sandgren
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Dermot Kenny
- Clinical Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ana Maria Vasilache
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Mohammad R Abedi
- Department of Clinical Immunology and Transfusion medicine, Faculty of Medicine and Health, Örebro University, Sweden
| | - Sofia Ramström
- Department of Clinical Chemistry and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
13
|
Lee RH, Rocco DJ, Nieswandt B, Bergmeier W. The CalDAG-GEFI/Rap1/αIIbβ3 axis minimally contributes to accelerated platelet clearance in mice with constitutive store-operated calcium entry. Platelets 2023; 34:2157383. [PMID: 36683325 PMCID: PMC10032033 DOI: 10.1080/09537104.2022.2157383] [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] [Indexed: 01/24/2023]
Abstract
Circulating platelets maintain low cytosolic Ca2+ concentrations. At sites of vascular injury, agonist-induced Ca2+ release from platelet intracellular stores triggers influx of extracellular Ca2+, a process known as store-operated Ca2+ entry (SOCE). Stromal interaction molecule 1 (Stim1) senses reduced Ca2+ stores and triggers SOCE. Gain-of-function (GOF) mutations in Stim1, such as described for Stormorken syndrome patients or mutant mice (Stim1Sax), are associated with marked thrombocytopenia and increased platelet turnover. We hypothesized that reduced platelet survival in Stim1Sax/+ mice is due to increased Rap1/integrin signaling and platelet clearance in the spleen, similar to what we recently described for mice expressing a mutant version of the Rap1-GAP, Rasa3 (Rasa3hlb/hlb). Stim1Sax/+ mice were crossed with mice deficient in CalDAG-GEFI, a critical calcium-regulated Rap1-GEF in platelets. In contrast to Rasa3hlb/hlb x Caldaggef1-/- mice, only a small increase in the peripheral platelet count, but not platelet lifespan, was observed in Stim1Sax/+ x Caldaggef1-/- mice. Similarly, inhibition of αIIbβ3 integrin in vivo only minimally raised the peripheral platelet count in Stim1Sax/+ mice. Compared to controls, Stim1Sax/+ mice exhibited increased platelet accumulation in the lung, but not the spleen or liver. These results suggest that CalDAG-GEFI/Rap1/integrin signaling contributes only minimally to accelerated platelet turnover caused by constitutive SOCE.
Collapse
Affiliation(s)
- Robert H Lee
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill
- UNC Blood Research Center, University of North Carolina at Chapel Hill
| | - David J Rocco
- UNC Blood Research Center, University of North Carolina at Chapel Hill
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany
- Rudolf Virchow Center, University of Würzburg, Würzburg, Germany
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill
- UNC Blood Research Center, University of North Carolina at Chapel Hill
| |
Collapse
|
14
|
Josefsson EC. Platelet intrinsic apoptosis. Thromb Res 2023; 231:206-213. [PMID: 36739256 DOI: 10.1016/j.thromres.2022.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/31/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022]
Abstract
In a healthy individual, the lifespan of most platelets is tightly regulated by intrinsic, or mitochondrial, apoptosis. This is a special form of programmed cell death governed by the BCL-2 family of proteins, where the prosurvival protein BCL-XL maintains platelet viability by restraining the prodeath proteins BAK and BAX. Restriction of platelet lifespan by activation of BAK and BAX mediated intrinsic apoptosis is essential to maintain a functional, haemostatically reactive platelet population. This review focuses on the molecular regulation of intrinsic apoptosis in platelets, reviews conditions linked to enhanced platelet death, discusses ex vivo storage of platelets and describes caveats associated with the assessment of platelet apoptosis.
Collapse
Affiliation(s)
- Emma C Josefsson
- Sahlgrenska University Hospital, Department of Clinical Chemistry, Gothenburg, Sweden; The University of Gothenburg, Department of Laboratory Medicine, Institute of Biomedicine, Gothenburg, Sweden; The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, VIC 3052, Australia; The University of Melbourne, Department of Medical Biology, 1G Royal Parade, VIC 3052, Australia.
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW Platelet mitochondrial dysfunction is both caused by, as well as a source of oxidative stress. Oxidative stress is a key hallmark of metabolic disorders such as dyslipidemia and diabetes, which are known to have higher risks for thrombotic complications. RECENT FINDINGS Increasing evidence supports a critical role for platelet mitochondria beyond energy production and apoptosis. Mitochondria are key regulators of reactive oxygen species and procoagulant platelets, which both contribute to pathological thrombosis. Studies targeting platelet mitochondrial pathways have reported promising results suggesting antithrombotic effects with limited impact on hemostasis in animal models. SUMMARY Targeting platelet mitochondria holds promise for the reduction of thrombotic complications in patients with metabolic disorders. Future studies should aim at validating these preclinical findings and translate them to the clinic.
Collapse
Affiliation(s)
- Abigail Ajanel
- University of Utah Molecular Medicine Program, Salt Lake City, Utah
- Department Pathology, Division of Microbiology and Pathology, University of Utah, Salt Lake City, Utah
| | - Robert A. Campbell
- University of Utah Molecular Medicine Program, Salt Lake City, Utah
- Department Pathology, Division of Microbiology and Pathology, University of Utah, Salt Lake City, Utah
- Department of Internal Medicine, Division of Hematology, University of Utah, Salt Lake City, Utah
| | - Frederik Denorme
- University of Utah Molecular Medicine Program, Salt Lake City, Utah
- Department of Neurology, Division of Vascular Neurology, University of Utah, Salt Lake City, Utah
| |
Collapse
|
16
|
Moon B, Yang S, Moon H, Lee J, Park D. After cell death: the molecular machinery of efferocytosis. Exp Mol Med 2023; 55:1644-1651. [PMID: 37612408 PMCID: PMC10474042 DOI: 10.1038/s12276-023-01070-5] [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: 04/26/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 08/25/2023] Open
Abstract
Cells constituting a multicellular organism die in a variety of ways throughout life, and most of them die via apoptosis under normal conditions. The occurrence of apoptosis is especially prevalent during development and in tissues with a high cellular turnover rate, such as the thymus and bone marrow. Interestingly, although the number of apoptotic cells produced daily is known to be innumerable in a healthy adult human body, apoptotic cells are rarely observed. This absence is due to the existence of a cellular process called efferocytosis that efficiently clears apoptotic cells. Studies over the past decades have focused on how phagocytes are able to remove apoptotic cells specifically, swiftly, and continuously, resulting in defined molecular and cellular events. In this review, we will discuss the current understanding of the clearance of apoptotic cells at the molecular level.
Collapse
Affiliation(s)
- Byeongjin Moon
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
- Cell Mechanobiology Laboratory, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Susumin Yang
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
- Cell Mechanobiology Laboratory, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Hyunji Moon
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
- Cell Mechanobiology Laboratory, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Juyeon Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
- Cell Mechanobiology Laboratory, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea
| | - Daeho Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea.
- Cell Mechanobiology Laboratory, Gwangju Institute of Science and Technology, Gwangju, 61005, Korea.
| |
Collapse
|
17
|
Agbani EO, Chow L, Nicholas J, Skeith L, Schneider P, Gregory A, Mahe E, Yamaura L, Young D, Dufour A, Paul PP, Walker AM, Mukherjee PG, Poole AW, Poon MC, Lee A. Overexpression of facilitative glucose transporter-3 and membrane procoagulation in maternal platelets of preeclamptic pregnancy. J Thromb Haemost 2023; 21:1903-1919. [PMID: 36963633 DOI: 10.1016/j.jtha.2023.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/19/2023] [Accepted: 03/03/2023] [Indexed: 03/26/2023]
Abstract
BACKGROUND Preeclampsia (PE) is a hypertensive disorder during pregnancy that results in significant adverse maternal and neonatal outcomes. Platelet activation is present in PE and contributes to the thrombo-hemorrhagic states of the disorder. However, the mechanisms that initiate and/or sustain platelet activation in PE are ill-defined. OBJECTIVES We aimed to characterise this mechanism and the procoagulant potentials of platelets in PE. METHODS In this quantitative observational study, we analyzed platelet procoagulant membrane dynamics in patients with PE (n = 21) compared with age-matched normotensive pregnancies (n = 20), gestational hypertension (n = 10), and non-pregnant female controls (n = 19). We analyzed fluorescently labeled indicators of platelet activation, bioenergetics, and procoagulation (phosphatidylserine exposure and thrombin generation), coupled with high-resolution imaging and thrombelastography. We then validated our findings using flow cytometry, immunoassays, classical pharmacology, and convolutional neural network analysis. RESULTS PE platelets showed significant ultra-structural remodeling, are more extensively preactivated than in healthy pregnancies and can circulate as microaggregates. Preactivated platelets of PE externalized phosphatidylserine and thrombin formed on the platelet membranes. Platelets' expression of facilitative glucose transporter-1 increased in all pregnant groups. However, PE platelets additionally overexpress glucose transporter-3 to enhance glucose uptake and sustain activation and secretion events. Although preeclampsia platelets exposed to subendothelial collagen showed incremental activation, the absolute hemostatic response to collagen was diminished, and likely contributed to greater blood loss perioperatively. CONCLUSIONS We revealed 2 bioenergetic mediators in the mechanism of sustained platelet procoagulation in preeclampsia. Although glucose transporter-1 and glucose transporter-3 remain elusive antiprocoagulant targets, they may be sensitive monitors of PE onset and progression.
Collapse
Affiliation(s)
- Ejaife O Agbani
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Alberta, Canada; Libin Cardiovascular Institute, Calgary, Alberta, Canada.
| | - Lorraine Chow
- Department of Anaesthesiology, Perioperative and Pain Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Joshua Nicholas
- Department of Anaesthesiology, Perioperative and Pain Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Leslie Skeith
- Libin Cardiovascular Institute, Calgary, Alberta, Canada; Division of Hematology & Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Prism Schneider
- Department of Surgery, Cumming School of Medicine, University of Calgary, Alberta, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Alberta, Canada
| | - Alexander Gregory
- Libin Cardiovascular Institute, Calgary, Alberta, Canada; Department of Anaesthesiology, Perioperative and Pain Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Etienne Mahe
- Division of Hematology & Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada; Department of Pathology & Laboratory Medicine, University of Calgary, Alberta, Canada
| | - Lisa Yamaura
- Department of Surgery, Cumming School of Medicine, University of Calgary, Alberta, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Alberta, Canada
| | - Daniel Young
- McCaig Institute for Bone and Joint Health, University of Calgary, Alberta, Canada
| | - Antoine Dufour
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Alberta, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Alberta, Canada
| | - Padma Polash Paul
- Braintoy Inc Calgary and Computational Neuroscience Lab, University of Oxford, England, United Kingdom
| | - Andrew M Walker
- Department of Anaesthesiology, Perioperative and Pain Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | | | - Alastair W Poole
- School of Physiology, Pharmacology, and Neuroscience, University of Bristol, England, United Kingdom
| | - Man-Chiu Poon
- Division of Hematology & Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada; Arnie Charbonneau Cancer Institute, Calgary, Alberta, Canada
| | - Adrienne Lee
- Division of Hematology & Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada; Division of Hematology, Department of Medicine/Medical Oncology, University of British Columbia, Island Health, Victoria, Canada
| |
Collapse
|
18
|
Zhang P, Jiang H, Yang M, Bi C, Zhang K, Liu D, Wei M, Jiang Z, Lv K, Fang C, Liu J, Zhang T, Xu Y, Zhang J. AGK Potentiates Arterial Thrombosis by Affecting Talin-1 and αIIbβ3-Mediated Bidirectional Signaling Pathway. Arterioscler Thromb Vasc Biol 2023; 43:1015-1030. [PMID: 37051931 DOI: 10.1161/atvbaha.122.318647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 03/22/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND AGK (acylglycerol kinase) was first identified as a mitochondrial transmembrane protein that exhibits a lipid kinase function. Recent studies have established that AGK promotes cancer growth and metastasis, enhances glycolytic metabolism and function fitness of CD8+ T cells, or regulates megakaryocyte differentiation. However, the role of AGK in platelet activation and arterial thrombosis remains to be elaborated. METHODS We performed hematologic analysis using automated hematology analyzer and investigated platelets morphology by transmission electron microscope. We explored the role of AGK in platelet activation and arterial thrombosis utilizing transgenic mice, platelet functional experiments in vitro, and thrombosis models in vivo. We revealed the regulation effect of AGK on Talin-1 by coimmunoprecipitation, mass spectrometry, immunofluorescence, and Western blot. We tested the role of AGK on lipid synthesis of phosphatidic acid/lysophosphatidic acid and thrombin generation by specific Elisa kits. RESULTS In this study, we found that AGK depletion or AGK mutation had no effect on the platelet average volumes, the platelet microstructures, or the expression levels of the major platelet membrane receptors. However, AGK deficiency or AGK mutation conspicuously decreased multiple aspects of platelet activation, including agonists-induced platelet aggregation, granules secretion, JON/A binding, spreading on Fg (fibrinogen), and clot retraction. AGK deficiency or AGK mutation also obviously delayed arterial thrombus formation but had no effect on tail bleeding time and platelet procoagulant function. Mechanistic investigation revealed that AGK may promote Talin-1Ser425 phosphorylation and affect the αIIbβ3-mediated bidirectional signaling pathway. However, AGK does not affect lipid synthesis of phosphatidic acid/lysophosphatidic acid in platelets. CONCLUSIONS AGK, through its kinase activity, potentiates platelet activation and arterial thrombosis by promoting Talin-1 Ser425 phosphorylation and affecting the αIIbβ3-mediated bidirectional signaling pathway.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Haojie Jiang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China (H.J., M.Y., J.L., Y.X.)
| | - Mina Yang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China (H.J., M.Y., J.L., Y.X.)
| | - Changlong Bi
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Kandi Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Dongsheng Liu
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Meng Wei
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Zheyi Jiang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Keyu Lv
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China (K.L., C.F.)
| | - Chao Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China (K.L., C.F.)
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China (H.J., M.Y., J.L., Y.X.)
| | - Tiantian Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| | - Yanyan Xu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China (H.J., M.Y., J.L., Y.X.)
| | - Junfeng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China (P.Z., C.B., K.Z., D.L., M.W., Z.J., T.Z., J.Z.)
| |
Collapse
|
19
|
Sakuragi T, Nagata S. Regulation of phospholipid distribution in the lipid bilayer by flippases and scramblases. Nat Rev Mol Cell Biol 2023:10.1038/s41580-023-00604-z. [PMID: 37106071 PMCID: PMC10134735 DOI: 10.1038/s41580-023-00604-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 04/29/2023]
Abstract
Cellular membranes function as permeability barriers that separate cells from the external environment or partition cells into distinct compartments. These membranes are lipid bilayers composed of glycerophospholipids, sphingolipids and cholesterol, in which proteins are embedded. Glycerophospholipids and sphingolipids freely move laterally, whereas transverse movement between lipid bilayers is limited. Phospholipids are asymmetrically distributed between membrane leaflets but change their location in biological processes, serving as signalling molecules or enzyme activators. Designated proteins - flippases and scramblases - mediate this lipid movement between the bilayers. Flippases mediate the confined localization of specific phospholipids (phosphatidylserine (PtdSer) and phosphatidylethanolamine) to the cytoplasmic leaflet. Scramblases randomly scramble phospholipids between leaflets and facilitate the exposure of PtdSer on the cell surface, which serves as an important signalling molecule and as an 'eat me' signal for phagocytes. Defects in flippases and scramblases cause various human diseases. We herein review the recent research on the structure of flippases and scramblases and their physiological roles. Although still poorly understood, we address the mechanisms by which they translocate phospholipids between lipid bilayers and how defects cause human diseases.
Collapse
Affiliation(s)
- Takaharu Sakuragi
- Biochemistry & Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shigekazu Nagata
- Biochemistry & Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.
| |
Collapse
|
20
|
Furuta Y, Zhou Z. How do necrotic cells expose phosphatidylserine to attract their predators—What’s unique and what’s in common with apoptotic cells. Front Cell Dev Biol 2023; 11:1170551. [PMID: 37091984 PMCID: PMC10113483 DOI: 10.3389/fcell.2023.1170551] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/27/2023] [Indexed: 04/09/2023] Open
Abstract
Phosphatidylserine (PS) is a lipid component of the plasma membrane. It is asymmetrically distributed to the inner leaflet in live cells. In cells undergoing apoptosis, phosphatidylserine is exposed to the outer surfaces. The exposed phosphatidylserine acts as an evolutionarily conserved “eat-me” signal that attracts neighboring engulfing cells in metazoan organisms, including the nematode Caenorhabditis elegans, the fruit fly Drosophila melanogaster, and mammals. During apoptosis, the exposure of phosphatidylserine to the outer surface of a cell is driven by the membrane scramblases and flippases, the activities of which are regulated by caspases. Cells undergoing necrosis, a kind of cell death frequently associated with cellular injuries and morphologically distinct from apoptosis, were initially believed to allow passive exposure of phosphatidylserine through membrane rupture. Later studies revealed that necrotic cells actively expose phosphatidylserine before any rupture occurs. A recent study in C. elegans further reported that the calcium ion (Ca2+) plays an essential role in promoting the exposure of phosphatidylserine on the surfaces of necrotic cells. These findings indicate that necrotic and apoptotic cells, which die through different molecular mechanisms, use common and unique mechanisms for promoting the exposure of the same “eat me” signal. This article will review the mechanisms regulating the exposure of phosphatidylserine on the surfaces of necrotic and apoptotic cells and highlight their similarities and differences.
Collapse
|
21
|
Johnson L, Lei P, Waters L, Padula MP, Marks DC. Identification of platelet subpopulations in cryopreserved platelet components using multi-colour imaging flow cytometry. Sci Rep 2023; 13:1221. [PMID: 36681723 PMCID: PMC9867743 DOI: 10.1038/s41598-023-28352-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/17/2023] [Indexed: 01/22/2023] Open
Abstract
Cryopreservation of platelets, at - 80 °C with 5-6% DMSO, results in externalisation of phosphatidylserine and the formation of extracellular vesicles (EVs), which may mediate their procoagulant function. The phenotypic features of procoagulant platelets overlap with other platelet subpopulations. The aim of this study was to define the phenotype of in vitro generated platelet subpopulations, and subsequently identify the subpopulations present in cryopreserved components. Fresh platelet components (n = 6 in each group) were either unstimulated as a source of resting platelets; or stimulated with thrombin and collagen to generate a mixture of aggregatory and procoagulant platelets; calcium ionophore (A23187) to generate procoagulant platelets; or ABT-737 to generate apoptotic platelets. Platelet components (n = 6) were cryopreserved with DMSO, thawed and resuspended in a unit of thawed plasma. Multi-colour panels of fluorescent antibodies and dyes were used to identify the features of subpopulations by imaging flow cytometry. A combination of annexin-V (AnnV), CD42b, and either PAC1 or CD62P was able to distinguish the four subpopulations. Cryopreserved platelets contained procoagulant platelets (AnnV+/PAC1-/CD42b+/CD62P+) and a novel population (AnnV+/PAC1-/CD42b+/CD62P-) that did not align with the phenotype of aggregatory (AnnV-/PAC1+/CD42b+/CD62P+) or apoptotic (AnnV+/PAC1-/CD42b-/CD62P-) subpopulations. These data suggests that the enhanced haemostatic potential of cryopreserved platelets may be due to the cryo-induced development of procoagulant platelets, and that additional subpopulations may exist.
Collapse
Affiliation(s)
- Lacey Johnson
- Research and Development, Australian Red Cross Lifeblood, Alexandria, NSW, Australia.
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia.
| | - Pearl Lei
- Research and Development, Australian Red Cross Lifeblood, Alexandria, NSW, Australia
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Lauren Waters
- Research and Development, Australian Red Cross Lifeblood, Alexandria, NSW, Australia
| | - Matthew P Padula
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Denese C Marks
- Research and Development, Australian Red Cross Lifeblood, Alexandria, NSW, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
22
|
Sokolovskaya AA, Popov MA, Sergeeva EA, Metelkin AA, Zybin DI, Shumakov DV, Kubatiev AA. Investigation of Platelet Apoptosis in Patients after Surgical Myocardial Revascularization. Biomedicines 2023; 11:251. [PMID: 36830787 PMCID: PMC9952963 DOI: 10.3390/biomedicines11020251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/13/2023] [Accepted: 01/15/2023] [Indexed: 01/19/2023] Open
Abstract
Platelets are one of the main participants in vascular accidents in cases of coronary heart disease (CHD). In this study, we sought to detect platelet apoptosis in patients with coronary artery disease who underwent scheduled myocardial revascularization surgery. To identify apoptotic events, we analyzed phosphatidylserine (PS) expression on the surface of platelets and mitochondrial membrane potential (ΔΨm) by flow cytometry in two groups of 30 patients aged 45-60 years: Group 1-patients before myocardial revascularization surgery and group 2-patients after myocardial revascularization surgery. The control group consisted of 10 healthy volunteers aged 45-60 years. According to our data, the percentage levels of PS expression in patients greatly decreased after surgery. We confirmed platelet apoptosis by recording depolarization of ΔΨm in pre- and postoperative patients. ΔΨm readings were considerably improved after surgery. Our data indicated that the functional parameters of platelets in patients with coronary heart disease differed from the characteristics of platelets in patients who underwent myocardial revascularization, and from those of patients in a control group. Future studies of platelet phenotypic characteristics and platelet apoptosis biomarkers should greatly advance our understanding of the pathophysiology of coronary heart disease, and further promote the development of methods for predicting adverse outcomes after surgery.
Collapse
Affiliation(s)
- Alisa A. Sokolovskaya
- Department of Molecular and Cellular Pathophysiology, Research Institute of General Pathology and Pathophysiology, Baltiyskaya 8, 125315 Moscow, Russia
| | - Mikhail A. Popov
- Department of Cardiosurgery, Vladimirsky Moscow Regional Research Clinical Institute, Shepkina 61/2, 129110 Moscow, Russia
| | - Ekaterina A. Sergeeva
- Department of Molecular and Cellular Pathophysiology, Research Institute of General Pathology and Pathophysiology, Baltiyskaya 8, 125315 Moscow, Russia
| | - Arkadiy A. Metelkin
- Department of Molecular and Cellular Pathophysiology, Research Institute of General Pathology and Pathophysiology, Baltiyskaya 8, 125315 Moscow, Russia
| | - Dmitry I. Zybin
- Department of Cardiosurgery, Vladimirsky Moscow Regional Research Clinical Institute, Shepkina 61/2, 129110 Moscow, Russia
| | - Dmitry V. Shumakov
- Department of Cardiosurgery, Vladimirsky Moscow Regional Research Clinical Institute, Shepkina 61/2, 129110 Moscow, Russia
| | - Aslan A. Kubatiev
- Department of Molecular and Cellular Pathophysiology, Research Institute of General Pathology and Pathophysiology, Baltiyskaya 8, 125315 Moscow, Russia
| |
Collapse
|
23
|
Sex-dependent effects of canagliflozin and dapagliflozin on hemostasis in normoglycemic and hyperglycemic mice. Sci Rep 2023; 13:932. [PMID: 36650229 PMCID: PMC9845220 DOI: 10.1038/s41598-023-28225-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i) are antihyperglycemic drugs that decrease mortality from cardiovascular diseases. However, their effects on hemostasis in the cardioprotective effects have not been evaluated. Therefore, the effects of canagliflozin (CANA, 100 mg/kg, p.o.) and dapagliflozin (DAPA, 10 mg/kg, p.o.) on the parameters of hemostasis were investigated in female and male normoglycemic and streptozotocin (180 mg/kg, i.p.)-induced diabetic mice. CANA and DAPA reduced platelet activity in thrombus in male and female mice both normoglycemic and diabetic. CANA decreased thrombus formation in diabetic male mice, and platelet activation to ADP in diabetic female and male mice. Activation of fibrinolysis was observed in female mice, both normoglycemic and diabetic. DAPA reduced thrombus formation in diabetic male and female mice, and decreased platelet activation to ADP and fibrin formation in diabetic male mice. DAPA increased fibrin formation in normoglycemic female mice and activated fibrinolysis in diabetic female mice. CANA and DAPA exerted sex-specific effects, which were more pronounced in hyperglycemia. The antithrombotic effect of CANA and DAPA was more noticeable in male mice and could be due to platelet inhibition. The effect on coagulation and fibrinolysis was not clear since an increased coagulation and fibrinolysis were observed only in female mice.
Collapse
|
24
|
Platelets from 13-lined ground squirrels are resistant to cold storage lesions. J Comp Physiol B 2023; 193:125-134. [PMID: 36495374 DOI: 10.1007/s00360-022-01469-y] [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: 07/18/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 12/14/2022]
Abstract
During torpor in a 13-lined ground squirrel heart rate and blood flow decrease, increasing the risk of blood clot formation. In response, cells involved in clotting called platelets are sequestered in the liver, stored in the cold for months, and released back into circulation upon arousal. This is in contrast to non-hibernating mammals, including humans, in which chilled platelets undergo cold storage lesions and phagocytosis, leading to rapid clearance from circulation post-transfusion. Because of this, human platelets must be stored at room temperature, limiting their shelf life to 7 days due to the increased risk of microbial contamination at warmer temperatures. Human and ground squirrel platelets were stored at room temperature or 4 °C before being analyzed for cold storage lesions. Human platelets stored at 4 °C displayed progressive increases in phosphatidylserine surface exposure and caspase activation, while ground squirrel platelets showed minimal change. Following cold storage, sialic acid residues on human platelets were cleaved, leading to increased phagocytosis of human platelets by HepG2 cells. Ground squirrel platelets stored in the cold showed no changes in desialylation and phagocytosis, with Taxol-treated ground squirrel platelets showing the lowest phagocytosis rates between both species and all treatments. These results suggest that ground squirrel platelets may be resistant to cold storage lesions seen in human platelets. Although these experiments were done in vitro, they suggest a mechanism by which ground squirrel platelets are adapted to be stored during hibernation and remain functional following arousal. Other hibernating species may employ similar adaptations to retain functional platelets following torpor.
Collapse
|
25
|
De Silva E, Paul M, Kim H. Apoptosis in platelets is independent of the actin cytoskeleton. PLoS One 2022; 17:e0276584. [PMID: 36378629 PMCID: PMC9665360 DOI: 10.1371/journal.pone.0276584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022] Open
Abstract
Homeostasis between platelet production and clearance is essential for human health. A critical facet of the balance that facilitates platelet clearance from the circulation is apoptosis (programmed cell death). The precise cellular mechanisms that underpin platelet apoptosis are not defined. In nucleated cells, reorganization of the actin cytoskeleton is known to regulate platelet apoptosis. However, the role of the actin cytoskeleton in regulating apoptosis in platelets has not been extensively studied as they are anucleate and exhibit a distinctive physiology. Here, apoptosis was induced in washed human platelets using ABT-737, a BH3-mimetic drug. Mitochondrial depolarization was measured using the ratiometric dye JC-1; surface phosphatidylserine (PS) exposure was measured by annexin V binding; caspase-3 activation was measured by Western blotting. All three apoptotic markers were unaffected by the presence of either the actin depolymerizing drug cytochalasin D or the actin polymerizing drug jasplakinolide. Moreover, platelets were isolated from wild-type (WT) mice and mice deficient in gelsolin (Gsn), an actin-binding protein that is essential for normal cytoskeletal remodeling. In response to ABT-737, gelsolin-null (Gsn-/-) platelets initially showed accelerated PS exposure relative to WT platelets, however, both WT and Gsn-/- platelets exhibited similar levels of mitochondrial depolarization and caspase-3 activation in response to ABT-737. We conclude that ABT-737 induces established markers of platelet apoptosis in an actin-independent manner.
Collapse
Affiliation(s)
- Enoli De Silva
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Manoj Paul
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hugh Kim
- Centre for Blood Research, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
26
|
Coenzyme Q10 Attenuates Human Platelet Aggregation Induced by SARS-CoV-2 Spike Protein via Reducing Oxidative Stress In Vitro. Int J Mol Sci 2022; 23:ijms232012345. [PMID: 36293203 PMCID: PMC9604356 DOI: 10.3390/ijms232012345] [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: 09/09/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 12/23/2022] Open
Abstract
Platelet hyperreactivity and oxidative stress are the important causes of thrombotic disorders in patients with COVID-19. Oxidative stress, induced by the excessive generation of reactive oxygen species (ROS), could increase platelet function and the risk of thrombus formation. Coenzyme Q10 (CoQ10), exhibits strong antioxidative activity and anti-platelet effect. However, the effects and mechanisms of CoQ10 on attenuating platelet aggregation induced by spike protein have never been studied. This study aims to investigate whether the SARS-CoV-2 spike protein potentiates human platelet function via ROS signaling and the protective effect of CoQ10 in vitro. Using a series of platelet function assays, we found that spike protein potentiated platelet aggregation and oxidative stress, such as ROS level, mitochondrial membrane potential depolarization, and lipid damage level (MDA and 8-iso-PGF2α) in vitro. Furthermore, CoQ10 attenuated platelet aggregation induced by spike protein. As an anti-platelet mechanism, we showed that CoQ10 significantly decreased the excess production of ROS induced by spike protein. Our findings show that the protective effect of CoQ10 on spike protein-potentiated platelet aggregation is probably associated with its strong antioxidative ability.
Collapse
|
27
|
Kenny M, Stamboroski S, Taher R, Brüggemann D, Schoen I. Nanofiber Topographies Enhance Platelet-Fibrinogen Scaffold Interactions. Adv Healthc Mater 2022; 11:e2200249. [PMID: 35526111 PMCID: PMC11469041 DOI: 10.1002/adhm.202200249] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/05/2022] [Indexed: 11/07/2022]
Abstract
The initial contact with blood and its components, including plasma proteins and platelets, directs the body's response to foreign materials. Natural scaffolds of extracellular matrix or fibrin contain fibrils with nanoscale dimensions, but how platelets specifically respond to the topography and architecture of fibrous materials is still incompletely understood. Here, planar and nanofiber scaffolds are fabricated from native fibrinogen to characterize the morphology of adherent platelets and activation markers for phosphatidylserine exposure and α-granule secretion by confocal fluorescence microscopy and scanning electron microscopy. Different fibrinogen topographies equally support the spreading and α-granule secretion of washed platelets. In contrast, preincubation of the scaffolds with plasma diminishes platelet spreading on planar fibrinogen surfaces but not on nanofibers. The data show that the enhanced interactions of platelets with nanofibers result from a higher locally accessible surface area, effectively increasing the ligand density for integrin-mediated responses. Overall, fibrinogen nanofibers direct platelets toward robust adhesion formation and α-granule secretion while minimizing their procoagulant activity. Similar results on fibrinogen-coated polydimethylsiloxane substrates with micrometer-sized 3D features suggest that surface topography could be used more generally to steer blood-materials interactions on different length scales for enhancing the initial wound healing steps.
Collapse
Affiliation(s)
- Martin Kenny
- School of Pharmacy and Biomolecular SciencesRoyal College of Surgeons in Ireland (RCSI)123 St Stephen's GreenDublinD02 YN77Ireland
- Irish Centre for Vascular BiologyRoyal College of Surgeons in Ireland (RCSI)123 St Stephen's GreenDublinD02 YN77Ireland
| | - Stephani Stamboroski
- Institute for BiophysicsUniversity of BremenOtto‐Hahn‐Allee 1Bremen28359Germany
- Fraunhofer Institute for Manufacturing Technology and Advanced Materials (IFAM)Wiener Strasse 12Bremen28359Germany
| | - Reem Taher
- School of Pharmacy and Biomolecular SciencesRoyal College of Surgeons in Ireland (RCSI)123 St Stephen's GreenDublinD02 YN77Ireland
| | - Dorothea Brüggemann
- Institute for BiophysicsUniversity of BremenOtto‐Hahn‐Allee 1Bremen28359Germany
- MAPEX Center for Materials and ProcessesUniversity of BremenBremen28359Germany
| | - Ingmar Schoen
- School of Pharmacy and Biomolecular SciencesRoyal College of Surgeons in Ireland (RCSI)123 St Stephen's GreenDublinD02 YN77Ireland
- Irish Centre for Vascular BiologyRoyal College of Surgeons in Ireland (RCSI)123 St Stephen's GreenDublinD02 YN77Ireland
| |
Collapse
|
28
|
Procoagulant platelet sentinels prevent inflammatory bleeding through GPIIBIIIA and GPVI. Blood 2022; 140:121-139. [PMID: 35472164 DOI: 10.1182/blood.2021014914] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 04/12/2022] [Indexed: 11/20/2022] Open
Abstract
Impairment of vascular integrity is a hallmark of inflammatory diseases. We recently reported that single immune-responsive platelets migrate and re-position themselves to sites of vascular injury to prevent bleeding. However, it remains unclear how single platelets preserve vascular integrity once encountering endothelial breaches. Here we demonstrate by intravital microscopy combined with genetic mouse models that procoagulant activation (PA) of single platelets and subsequent recruitment of the coagulation cascade are crucial for the prevention of inflammatory bleeding. Using a novel lactadherin-based compound we detect phosphatidylserine (PS)-positive procoagulant platelets in the inflamed vasculature. We identify exposed collagen as the central trigger arresting platelets and initiating subsequent PA in a CypD- and TMEM16F-dependent manner both in vivo and in vitro. Platelet PA promotes binding of the prothrombinase complex to the platelet membrane, greatly enhancing thrombin activity resulting in fibrin formation. PA of migrating platelets is initiated by co-stimulation via integrin αIIbβ3 (GPIIBIIIA)/Gα13-mediated outside-in-signaling and GPVI signaling, leading to an above-threshold intracellular calcium release. This effectively targets the coagulation cascade to breaches of vascular integrity identified by patrolling platelets. Platelet-specific genetic loss of either CypD or TMEM16F as well as combined blockade of platelet GPIIBIIIA and GPVI reduce platelet PA in vivo and aggravate pulmonary inflammatory hemorrhage. Our findings illustrate a novel role of procoagulant platelets in the prevention of inflammatory bleeding and provide evidence that PA of patrolling platelet sentinels effectively targets and confines activation of coagulation to breaches of vascular integrity.
Collapse
|
29
|
Alarabi AB, Mohsen A, Mizuguchi K, Alshbool FZ, Khasawneh FT. Co-expression analysis to identify key modules and hub genes associated with COVID-19 in platelets. BMC Med Genomics 2022; 15:83. [PMID: 35421970 PMCID: PMC9008611 DOI: 10.1186/s12920-022-01222-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/21/2022] [Indexed: 01/23/2023] Open
Abstract
Corona virus disease 2019 (COVID-19) increases the risk of cardiovascular occlusive/thrombotic events and is linked to poor outcomes. The underlying pathophysiological processes are complex, and remain poorly understood. To this end, platelets play important roles in regulating the cardiovascular system, including via contributions to coagulation and inflammation. There is ample evidence that circulating platelets are activated in COVID-19 patients, which is a primary driver of the observed thrombotic outcome. However, the comprehensive molecular basis of platelet activation in COVID-19 disease remains elusive, which warrants more investigation. Hence, we employed gene co-expression network analysis combined with pathways enrichment analysis to further investigate the aforementioned issues. Our study revealed three important gene clusters/modules that were closely related to COVID-19. These cluster of genes successfully identify COVID-19 cases, relative to healthy in a separate validation data set using machine learning, thereby validating our findings. Furthermore, enrichment analysis showed that these three modules were mostly related to platelet metabolism, protein translation, mitochondrial activity, and oxidative phosphorylation, as well as regulation of megakaryocyte differentiation, and apoptosis, suggesting a hyperactivation status of platelets in COVID-19. We identified the three hub genes from each of three key modules according to their intramodular connectivity value ranking, namely: COPE, CDC37, CAPNS1, AURKAIP1, LAMTOR2, GABARAP MT-ND1, MT-ND5, and MTRNR2L12. Collectively, our results offer a new and interesting insight into platelet involvement in COVID-19 disease at the molecular level, which might aid in defining new targets for treatment of COVID-19–induced thrombosis.
Collapse
|
30
|
Bendas G, Schlesinger M. The GPIb-IX complex on platelets: insight into its novel physiological functions affecting immune surveillance, hepatic thrombopoietin generation, platelet clearance and its relevance for cancer development and metastasis. Exp Hematol Oncol 2022; 11:19. [PMID: 35366951 PMCID: PMC8976409 DOI: 10.1186/s40164-022-00273-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/19/2022] [Indexed: 12/13/2022] Open
Abstract
The glycoprotein (GP) Ib-IX complex is a platelet receptor that mediates the initial interaction with subendothelial von Willebrand factor (VWF) causing platelet arrest at sites of vascular injury even under conditions of high shear. GPIb-IX dysfunction or deficiency is the reason for the rare but severe Bernard-Soulier syndrome (BSS), a congenital bleeding disorder. Although knowledge on GPIb-IX structure, its basic functions, ligands, and intracellular signaling cascades have been well established, several advances in GPIb-IX biology have been made in the recent years. Thus, two mechanosensitive domains and a trigger sequence in GPIb were characterized and its role as a thrombin receptor was deciphered. Furthermore, it became clear that GPIb-IX is involved in the regulation of platelet production, clearance and thrombopoietin secretion. GPIb is deemed to contribute to liver cancer development and metastasis. This review recapitulates these novel findings highlighting GPIb-IX in its multiple functions as a key for immune regulation, host defense, and liver cancer development.
Collapse
Affiliation(s)
- Gerd Bendas
- Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Martin Schlesinger
- Department of Pharmacy, Rheinische Friedrich-Wilhelms-University Bonn, An der Immenburg 4, 53121, Bonn, Germany. .,Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany.
| |
Collapse
|
31
|
Lozano PA, Alarabi AB, Garcia SE, Boakye ET, Kingbong HT, Naddour E, Villalobos-García D, Badejo P, El-Halawany MS, Khasawneh FT, Alshbool FZ. The Antidepressant Duloxetine Inhibits Platelet Function and Protects against Thrombosis. Int J Mol Sci 2022; 23:ijms23052587. [PMID: 35269729 PMCID: PMC8910021 DOI: 10.3390/ijms23052587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 02/23/2022] [Accepted: 02/23/2022] [Indexed: 01/27/2023] Open
Abstract
While cardiovascular disease (CVD) is the leading cause of death, major depressive disorder (MDD) is the primary cause of disability, affecting more than 300 million people worldwide. Interestingly, there is evidence that CVD is more prevalent in people with MDD. It is well established that neurotransmitters, namely serotonin and norepinephrine, are involved in the biochemical mechanisms of MDD, and consequently, drugs targeting serotonin-norepinephrine reuptake, such as duloxetine, are commonly prescribed for MDD. In this connection, serotonin and norepinephrine are also known to play critical roles in primary hemostasis. Based on these considerations, we investigated if duloxetine can be repurposed as an antiplatelet medication. Our results-using human and/or mouse platelets show that duloxetine dose-dependently inhibited agonist-induced platelet aggregation, compared to the vehicle control. Furthermore, it also blocked agonist-induced dense and α-granule secretion, integrin αIIbβ3 activation, phosphatidylserine expression, and clot retraction. Moreover duloxetine-treated mice had a significantly prolonged occlusion time. Finally, duloxetine was also found to impair hemostasis. Collectively, our data indicate that the antidepressant duloxetine, which is a serotonin-norepinephrine antagonist, exerts antiplatelet and thromboprotective effects and inhibits hemostasis. Consequently, duloxetine, or a rationally designed derivative, presents potential benefits in the context of CVD, including that associated with MDD.
Collapse
Affiliation(s)
- Patricia A. Lozano
- Department of Pharmacy Practice, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (P.A.L.); (A.B.A.)
| | - Ahmed B. Alarabi
- Department of Pharmacy Practice, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (P.A.L.); (A.B.A.)
| | - Sarah E. Garcia
- School of Pharmacy, The University of Texas at El Paso, El Paso, TX 79902, USA; (S.E.G.); (E.T.B.); (H.T.K.); (E.N.)
| | - Erica T. Boakye
- School of Pharmacy, The University of Texas at El Paso, El Paso, TX 79902, USA; (S.E.G.); (E.T.B.); (H.T.K.); (E.N.)
| | - Hendreta T. Kingbong
- School of Pharmacy, The University of Texas at El Paso, El Paso, TX 79902, USA; (S.E.G.); (E.T.B.); (H.T.K.); (E.N.)
| | - Elie Naddour
- School of Pharmacy, The University of Texas at El Paso, El Paso, TX 79902, USA; (S.E.G.); (E.T.B.); (H.T.K.); (E.N.)
| | - Daniel Villalobos-García
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (D.V.-G.); (P.B.); (M.S.E.-H.)
| | - Precious Badejo
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (D.V.-G.); (P.B.); (M.S.E.-H.)
| | - Medhat S. El-Halawany
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (D.V.-G.); (P.B.); (M.S.E.-H.)
| | - Fadi T. Khasawneh
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (D.V.-G.); (P.B.); (M.S.E.-H.)
- Correspondence: (F.T.K.); (F.Z.A.); Tel.: +1-(361)-221-0755 (F.T.K.); +1-(361)-221-0793 (F.Z.A.)
| | - Fatima Z. Alshbool
- Department of Pharmacy Practice, Irma Lerma Rangel College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (P.A.L.); (A.B.A.)
- Correspondence: (F.T.K.); (F.Z.A.); Tel.: +1-(361)-221-0755 (F.T.K.); +1-(361)-221-0793 (F.Z.A.)
| |
Collapse
|
32
|
Wong J, Gu BJ, Teoh H, Krupa M, Monif M, Slee M, Wiley JS. Flow Cytometry Identifies an Early Stage of Platelet Apoptosis Produced by Agonists of the P2X1 and P2X7 Receptors. Platelets 2022; 33:621-631. [PMID: 35042433 DOI: 10.1080/09537104.2021.1981844] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Platelets express P2X1 receptors and our data also show the expression of P2X7 receptors. We studied the role of both receptors in platelet apoptosis by incubation of PRP with P2X agonists, then centrifuged to remove viable platelets, and analyzed the supernatant by flow cytometry to identify a sparse platelet-derived population that stained with MitoTracker dyes and CD41. BzATP, a potent agonist of P2X receptors, and ABT737, an activator of intrinsic apoptosis, produced altered platelets that stained moderately for annexin V and corresponded to an early stage apoptotic platelet (ESAP). Over a range of BzATP concentrations, we observed a dose-dependent formation of ESAPs between 5 and 500 uM BzATP, together with a variable formation of ESAPs at nanomolar ATP or BzATP (50-200 nM). Production of ESAPs occurred with αβ-meATP, while responses with either BzATP or αβ-meATP showed desensitization at a higher agonist concentration. Formation of ESAPs by either 100 nM or 0.5 mM BzATP was inhibited by preincubation of platelets with latrunculin A, an inhibitor of the actin cytoskeleton that prevents apoptosis. ESAP production was totally inhibited by preincubation of platelets with methyl-beta-cyclodextrin, which removes cholesterol from lipid rafts. Our data show that both P2X1 and P2X7 receptors are localized in platelet lipid rafts where P2X-agonists act to produce early stage apoptotic platelets.
Collapse
Affiliation(s)
- Joelyn Wong
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Ben J Gu
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Harry Teoh
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, Australia
| | - Malgorzata Krupa
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, Australia
| | - Mastura Monif
- Department of Neurology, Royal Melbourne Hospital, Parkville, Australia.,Department of Neuroscience, Monash University, Clayton, Australia
| | - Mark Slee
- College of Medicine and Public Health, Flinders University of South Australia, Bedford Park, Australia
| | - James S Wiley
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia.,Haematology Department, Box Hill Hospital, Australia
| |
Collapse
|
33
|
Allan HE, Hayman MA, Marcone S, Chan MV, Edin ML, Maffucci T, Joshi A, Menke L, Crescente M, Mayr M, Zeldin DC, Armstrong PC, Warner TD. Proteome and functional decline as platelets age in the circulation. J Thromb Haemost 2021; 19:3095-3112. [PMID: 34390534 PMCID: PMC8604765 DOI: 10.1111/jth.15496] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/28/2021] [Accepted: 08/11/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND Platelets circulate in the blood of healthy individuals for approximately 7-10 days regulated by finely balanced processes of production and destruction. As platelets are anucleate we reasoned that their protein composition would change as they age and that this change would be linked to alterations in structure and function. OBJECTIVE To isolate platelets of different ages from healthy individuals to test the hypothesis that changes in protein content cause alterations in platelet structure and function. METHODS Platelets were separated according to thiazole orange fluorescence intensity as a surrogate indicator of mRNA content and so a marker of platelet age and then subjected to proteomics, imaging, and functional assays to produce an in-depth analysis of platelet composition and function. RESULTS Total protein content was 45 ± 5% lower in old platelets compared to young platelets. Predictive proteomic pathway analysis identified associations with 28 biological processes, notably higher hemostasis in young platelets whilst apoptosis and senescence were higher in old platelets. Further studies confirmed platelet ageing was linked to a decrease in cytoskeletal protein and associated capability to spread and adhere, a reduction in mitochondria number, and lower calcium dynamics and granule secretion. CONCLUSIONS Our findings demonstrate changes in protein content are linked to alterations in function as platelets age. This work delineates physical and functional changes in platelets as they age and serves as a base to examine differences associated with altered mean age of platelet populations in conditions such as immune thrombocytopenia and diabetes.
Collapse
Affiliation(s)
- Harriet E. Allan
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom
| | - Melissa A. Hayman
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom
| | - Simone Marcone
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Melissa V. Chan
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom
| | - Matthew L. Edin
- National Institutes of Health, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Tania Maffucci
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom
| | - Abhishek Joshi
- King’s British Heart Foundation Centre, Kings College London, London, United Kingdom
| | - Laura Menke
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom
| | - Marilena Crescente
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom
| | - Manuel Mayr
- King’s British Heart Foundation Centre, Kings College London, London, United Kingdom
| | - Darryl C. Zeldin
- National Institutes of Health, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Paul C. Armstrong
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom
| | - Timothy D. Warner
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University London, London, United Kingdom
| |
Collapse
|
34
|
Upregulation of cAMP prevents antibody-mediated thrombus formation in COVID-19. Blood Adv 2021; 6:248-258. [PMID: 34753174 PMCID: PMC8580563 DOI: 10.1182/bloodadvances.2021005210] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/21/2021] [Indexed: 11/20/2022] Open
Abstract
Thromboembolic events are frequently reported in patients infected with the SARS-CoV-2 virus. The exact mechanisms of COVID-19 associated hypercoagulopathy, however, remain elusive. Recently, we observed that platelets (PLTs) from patients with severe COVID-19 infection express high levels of procoagulant markers, which were found to be associated with increased risk for thrombosis. In the current study, we investigated the time course as well as the mechanisms leading to procoagulant PLTs in COVID-19. Our study demonstrates the presence of PLT-reactive IgG antibodies that induce marked changes in PLTs in terms of increased inner-mitochondrial-transmembrane potential (Δψ) depolarization, phosphatidylserine (PS) externalization and P-selectin expression. The IgG-induced procoagulant PLTs and increased thrombus formation was mediated by ligation of PLT Fc gamma RIIA (FcγRIIA). In addition, PLTs´ contents of calcium and cyclic-adenosine-monophosphate (cAMP) were identified to play central role in antibody-induced procoagulant PLT formation. Most importantly, antibody-induced procoagulant events as well as increased thrombus formation in severe COVID-19 were inhibited by Iloprost a clinically approved therapeutic agent that increases the intracellular cAMP levels in PLTs. Our data indicate that upregulation of cAMP could be a potential therapeutic target to prevent antibody-mediated coagulopathy in COVID-19 disease.
Collapse
|
35
|
A novel mechanism of thrombocytopenia by PS exposure through TMEM16F in sphingomyelin synthase 1 deficiency. Blood Adv 2021; 5:4265-4277. [PMID: 34478523 PMCID: PMC8945624 DOI: 10.1182/bloodadvances.2020002922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 05/27/2021] [Indexed: 01/04/2023] Open
Abstract
Membrane SM reduction by SMS1deficiency enhances PS exposure and thrombocytopenia. Depression of membrane SM potentiates Ca2+ influx and PS externalization through TMEM16F.
Sphingomyelin synthase 1 (SMS1) contributes to the generation of membrane sphingomyelin (SM) and affects SM-mediated physiological functions. Here, we describe the hematologic phenotypes, such as reduced circulating platelets and dysfunctional hemostasis, in SMS1-deficient (SMS1-KO) mice. SMS1-KO mice display pathologic manifestations related to idiopathic thrombocytopenia (ITP), including relatively high amounts of peripheral blood reticulated platelets, enhanced megakaryopoiesis in the bone marrow and spleen, and splenomegaly. Deficiency of SMS1, but not SMS2, prevented SM production and enhanced phosphatidylserine (PS) externalization on the plasma membranes of platelets and megakaryocytes. Consequently, SMS1-KO platelets were excessively cleared by macrophages in the spleen. Multimer formation in the plasma membrane of TMEM16F, a known calcium (Ca2+)-activated nonselective ion channel and Ca2+-dependent PS scramblase, was enhanced; the result was PS externalization to outer leaflets through increased Ca2+ influx in immortalized mouse embryonic fibroblasts established from SMS1-KO mice (SMS1-KO tMEFs), as seen with SMS1-KO platelets. Thus, SMS1 deficiency changed the TMEM16F distribution on the membrane microdomain, regulating Ca2+ influx-dependent PS exposure. SMS1-KO tMEFs in which TMEM16F was knocked out by using the CRISPR/Cas9 system lacked both the Ca2+ influx and excess PS exposure seen in SMS1-KO tMEFs. Therefore, SM depletion on platelet membrane microdomains due to SMS1 deficiency enhanced PS externalization via a Ca2+ influx through TMEM16F activation, leading to elevated platelet clearance and causing hemostasis dysfunction through thrombocytopenia. Our current findings show that the SM-rich microdomain generated by SMS1 is a potent regulator of thrombocytopenia through TMEM16F, suggesting that its dysfunction may be a novel additional mechanism of ITP.
Collapse
|
36
|
Xiao W, Zhou K, Yang M, Sun C, Dai L, Gu J, Yan R, Dai K. Carbamazepine Induces Platelet Apoptosis and Thrombocytopenia Through Protein Kinase A. Front Pharmacol 2021; 12:749930. [PMID: 34658890 PMCID: PMC8513130 DOI: 10.3389/fphar.2021.749930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/13/2021] [Indexed: 11/13/2022] Open
Abstract
Carbamazepine is extensively used worldwide to treat a wide range of disorders such as epilepsy, peripheral neuralgia and bipolar disorder. Thrombocytopenia and hemorrhage have been identified in multiple carbamazepine-treated patients. However, the underlying mechanism remains poorly understood. Here, we show that platelets undergo apoptosis after carbamazepine treatment. The apoptotic platelets induced by carbamazepine are rapidly removed in vivo, which accounts for thrombocytopenia. We found that carbamazepine treatment attenuates the phosphorylation level of bcl-xl/bcl-2-associated death promoter (BAD), vasodilator-associated stimulated phosphoprotein (VASP) and GPIbβ in platelets, indicating an inhibition effect on protein kinase A (PKA). We further demonstrated that carbamazepine reduced PKA activity through PI3K/Akt/PDE3A signaling pathway. Pharmacological activation of PKA or inhibition of PI3K/Akt/PDE3A protects platelets from apoptosis induced by carbamazepine. Importantly, PDE3A inhibitors or PKA activator ameliorates carbamazepine-mediated thrombocytopenia in vivo. These findings shed light on a possible mechanism of carbamazepine-induced thrombocytopenia, designating PDE3A/PKA as a potential therapeutic target in the treatment of carbamazepine-induced thrombocytopenia.
Collapse
Affiliation(s)
- Weiling Xiao
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China.,Department of Immunology, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Kangxi Zhou
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China
| | - Mengnan Yang
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China
| | - Chenglin Sun
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China
| | - Lan Dai
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China
| | - Jian Gu
- Department of Hematology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Rong Yan
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China
| | - Kesheng Dai
- Jiangsu Institute of Hematology, Cyrus Tang Medical Institute, The First Affiliated Hospital and Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, Suzhou, China
| |
Collapse
|
37
|
Alam M, Ali S, Mohammad T, Hasan GM, Yadav DK, Hassan MI. B Cell Lymphoma 2: A Potential Therapeutic Target for Cancer Therapy. Int J Mol Sci 2021; 22:ijms221910442. [PMID: 34638779 PMCID: PMC8509036 DOI: 10.3390/ijms221910442] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/15/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
Defects in the apoptosis mechanism stimulate cancer cell growth and survival. B cell lymphoma 2 (Bcl-2) is an anti-apoptotic molecule that plays a central role in apoptosis. Bcl-2 is the founding constituent of the Bcl-2 protein family of apoptosis controllers, the primary apoptosis regulators linked with cancer. Bcl-2 has been identified as being over-expressed in several cancers. Bcl-2 is induced by protein kinases and several signaling molecules which stimulate cancer development. Identifying the important function played by Bcl-2 in cancer progression and development, and treatment made it a target related to therapy for multiple cancers. Among the various strategies that have been proposed to block Bcl-2, BH3-mimetics have appeared as a novel group of compounds thanks to their favorable effects on many cancers within several clinical settings. Because of the fundamental function of Bcl-2 in the regulation of apoptosis, the Bcl-2 protein is a potent target for the development of novel anti-tumor treatments. Bcl-2 inhibitors have been used against several cancers and provide a pre-clinical platform for testing novel therapeutic drugs. Clinical trials of multiple investigational agents targeting Bcl-2 are ongoing. This review discusses the role of Bcl-2 in cancer development; it could be exploited as a potential target for developing novel therapeutic strategies to combat various types of cancers. We further highlight the therapeutic activity of Bcl-2 inhibitors and their implications for the therapeutic management of cancer.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (M.A.); (S.A.); (T.M.)
| | - Sabeeha Ali
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (M.A.); (S.A.); (T.M.)
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (M.A.); (S.A.); (T.M.)
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Dharmendra Kumar Yadav
- Department of Pharmacy and Gachon Institute of Pharmaceutical Science, College of Pharmacy, Gachon University, Hambakmoeiro 191, Yeonsu-gu, Incheon 21924, Korea
- Correspondence: (D.K.Y.); (M.I.H.)
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; (M.A.); (S.A.); (T.M.)
- Correspondence: (D.K.Y.); (M.I.H.)
| |
Collapse
|
38
|
Abstract
Coronary artery disease is a leading cause of morbidity and mortality worldwide. Despite significant advances in revascularization strategies and antiplatelet therapy with aspirin and/or P2Y12 receptor antagonist, patients with acute coronary syndrome (ACS) continue to be at long-term risk of further cardiovascular events. Besides platelet activation, the role of thrombin generation (TG) in atherothrombotic complications is widely recognized. In this study, we hypothesized that there is an elevation of coagulation activation persists beyond 12 months in patients with ACS and chronic coronary syndrome (CCS) when compared with healthy controls. We measured TG profiles of patients within 72 h after percutaneous coronary intervention, at 6-month, 12-month and 24-month. Our results demonstrated that TG of patients with ACS (n = 114) and CCS (n = 40) were persistently elevated when compared to healthy individuals (n = 50) in peak thrombin (ACS 273.1 nM vs CCS 287.3 nM vs healthy 234.3 nM) and velocity index (ACS 110.2 nM/min vs CCS 111.0 nM/min vs healthy 72.9 nM/min) at 24-month of follow-up. Our results suggest a rationale for addition of anticoagulation to antiplatelet therapy in preventing long-term ischemic events after ACS. Further research could clarify whether the use of TG parameters to enable risk stratification of patients at heightened long-term procoagulant risk who may benefit most from dual pathway inhibition.
Collapse
|
39
|
Curcumin at Low Doses Potentiates and at High Doses Inhibits ABT-737-Induced Platelet Apoptosis. Int J Mol Sci 2021; 22:ijms22105405. [PMID: 34065600 PMCID: PMC8161296 DOI: 10.3390/ijms22105405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 11/01/2022] Open
Abstract
Curcumin is a natural bioactive component derived from the turmeric plant Curcuma longa, which exhibits a range of beneficial activities on human cells. Previously, an inhibitory effect of curcumin on platelets was demonstrated. However, it is unknown whether this inhibitory effect is due to platelet apoptosis or procoagulant platelet formation. In this study, curcumin did not activate caspase 3-dependent apoptosis of human platelets, but rather induced the formation of procoagulant platelets. Interestingly, curcumin at low concentration (5 µM) potentiated, and at high concentration (50 µM) inhibited ABT-737-induced platelet apoptosis, which was accompanied by inhibition of ABT-737-mediated thrombin generation. Platelet viability was not affected by curcumin at low concentration and was reduced by 17% at high concentration. Furthermore, curcumin-induced autophagy in human platelets via increased translocation of LC3I to LC3II, which was associated with activation of adenosine monophosphate (AMP) kinase and inhibition of protein kinase B activity. Because curcumin inhibits P-glycoprotein (P-gp) in cancer cells and contributes to overcoming multidrug resistance, we showed that curcumin similarly inhibited platelet P-gp activity. Our results revealed that the platelet inhibitory effect of curcumin is mediated by complex processes, including procoagulant platelet formation. Thus, curcumin may protect against or enhance caspase-dependent apoptosis in platelets under certain conditions.
Collapse
|
40
|
Fraser M, Matuschewski K, Maier AG. Of membranes and malaria: phospholipid asymmetry in Plasmodium falciparum-infected red blood cells. Cell Mol Life Sci 2021; 78:4545-4561. [PMID: 33713154 PMCID: PMC11071739 DOI: 10.1007/s00018-021-03799-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/04/2021] [Accepted: 02/23/2021] [Indexed: 11/29/2022]
Abstract
Malaria is a vector-borne parasitic disease with a vast impact on human history, and according to the World Health Organisation, Plasmodium parasites still infect over 200 million people per year. Plasmodium falciparum, the deadliest parasite species, has a remarkable ability to undermine the host immune system and cause life-threatening disease during blood infection. The parasite's host cells, red blood cells (RBCs), generally maintain an asymmetric distribution of phospholipids in the two leaflets of the plasma membrane bilayer. Alterations to this asymmetry, particularly the exposure of phosphatidylserine (PS) in the outer leaflet, can be recognised by phagocytes. Because of the importance of innate immune defence numerous studies have investigated PS exposure in RBCs infected with P. falciparum, but have reached different conclusions. Here we review recent advancements in our understanding of the molecular mechanisms which regulate asymmetry in RBCs, and whether infection with the P. falciparum parasite results in changes to PS exposure. On the balance of evidence, it is likely that membrane asymmetry is disrupted in parasitised RBCs, though some methodological issues need addressing. We discuss the potential causes and consequences of altered asymmetry in parasitised RBCs, particularly for in vivo interactions with the immune system, and the role of host-parasite co-evolution. We also examine the potential asymmetric state of parasite membranes and summarise current knowledge on the parasite proteins, which could regulate asymmetry in these membranes. Finally, we highlight unresolved questions at this time and the need for interdisciplinary approaches to uncover the machinery which enables P. falciparum parasites to hide in mature erythrocytes.
Collapse
Affiliation(s)
- Merryn Fraser
- Research School of Biology, The Australian National University, Canberra, Australia
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - Kai Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - Alexander G Maier
- Research School of Biology, The Australian National University, Canberra, Australia.
| |
Collapse
|
41
|
Millington-Burgess SL, Harper MT. A double-edged sword: antibody-mediated procoagulant platelets in COVID-19. Platelets 2021; 32:579-581. [PMID: 33890850 DOI: 10.1080/09537104.2021.1912315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| |
Collapse
|
42
|
Podoplelova NA, Nechipurenko DY, Ignatova AA, Sveshnikova AN, Panteleev MA. Procoagulant Platelets: Mechanisms of Generation and Action. Hamostaseologie 2021; 41:146-153. [PMID: 33860522 DOI: 10.1055/a-1401-2706] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
During the past decades, it has been increasingly recognized that the major function of accelerating membrane-dependent reactions of blood coagulation is predominantly implemented by a subset of activated platelets. These procoagulant platelets (also called collagen- and thrombin-activated or COAT, coated, necrotic, although there could be subtle differences between these definitions) are uniquely characterized by both procoagulant activity and, at the same time, inactivated integrins and profibrinolytic properties. The mechanisms of their generation both in vitro and in situ have been increasingly becoming clear, suggesting unique and multidirectional roles in hemostasis and thrombosis. In this mini-review, we shall highlight the existing concepts and challenges in this field.
Collapse
Affiliation(s)
- N A Podoplelova
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow, Russia
| | - D Y Nechipurenko
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow, Russia.,Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia
| | - A A Ignatova
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow, Russia
| | - A N Sveshnikova
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow, Russia.,Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia
| | - M A Panteleev
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russia.,National Medical Research Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev, Moscow, Russia.,Faculty of Physics, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
43
|
Wang J, Zhou P, Han Y, Zhang H. Platelet transfusion for cancer secondary thrombocytopenia: Platelet and cancer cell interaction. Transl Oncol 2021; 14:101022. [PMID: 33545547 PMCID: PMC7868729 DOI: 10.1016/j.tranon.2021.101022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 01/14/2023] Open
Abstract
Chemoradiotherapy and autoimmune disorder often lead to secondary thrombocytopenia in cancer patients, and thus, platelet transfusion is needed to stop or prevent bleeding. However, the effect of platelet transfusion remains controversial for the lack of agreement on transfusion strategies. Before being transfused, platelets are stored in blood banks, and their activation is usually stimulated. Increasing evidence shows activated platelets may promote metastasis and the proliferation of cancer cells, while cancer cells also induce platelet activation. Such a vicious cycle of interaction between activated platelets and cancer cells is harmful for the prognosis of cancer patients, which results in an increased tumor recurrence rate and decreased five-year survival rate. Therefore, it is important to explore platelet transfusion strategies, summarize mechanisms of interaction between platelets and tumor cells, and carefully evaluate the pros and cons of platelet transfusion for better treatment and prognosis for patients with cancer with secondary thrombocytopenia.
Collapse
Affiliation(s)
- Juan Wang
- Class 2016 Clinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Pan Zhou
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China.
| | - Hongwei Zhang
- Department of Blood Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China.
| |
Collapse
|
44
|
Chu Y, Guo H, Zhang Y, Qiao R. Procoagulant platelets: Generation, characteristics, and therapeutic target. J Clin Lab Anal 2021; 35:e23750. [PMID: 33709517 PMCID: PMC8128296 DOI: 10.1002/jcla.23750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/17/2021] [Accepted: 02/20/2021] [Indexed: 12/19/2022] Open
Abstract
Platelets play a pivotal role in hemostasis. Activated platelets are classified into two groups, according to their agonist response: aggregating and procoagulant platelets. Aggregating platelets consist of activated integrin αIIbβ3 and stretch out pseudopods to further attract platelets to the site of injury by connecting with fibrinogen. They mainly gather in the core of the thrombus and perform a secretory function, such as releasing adenosine diphosphate (ADP). Procoagulant platelets promote the formation of thrombin and fibrin by interacting with coagulation factors and can thus be considered as the connector between primary and secondary hemostasis. In addition to their functions in blood coagulation, procoagulant platelets play a proinflammatory role by releasing platelet microparticles and inorganic polyphosphate. Considering these important functions of procoagulant platelets, this subpopulation warrants detailed study to analyze their potential in preventing human diseases. This review summarizes the generation and important characteristics of procoagulant platelets, as well as their potential for preventing the adverse effects associated with current antiplatelet therapies.
Collapse
Affiliation(s)
- Yaxin Chu
- The Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Han Guo
- The Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Yuncong Zhang
- The Department of Laboratory Medicine, Peking University International Hospital, Beijing, China
| | - Rui Qiao
- The Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
45
|
Ravanat C, Pongérard A, Freund M, Heim V, Rudwill F, Ziessel C, Eckly A, Proamer F, Isola H, Gachet C. Human platelets labeled at two discrete biotin densities are functional in vitro and are detected in vivo in the murine circulation: A promising approach to monitor platelet survival in vivo in clinical research. Transfusion 2021; 61:1642-1653. [PMID: 33580977 DOI: 10.1111/trf.16312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 01/17/2021] [Accepted: 01/17/2021] [Indexed: 01/15/2023]
Abstract
BACKGROUND The production of platelet concentrates (PCs) is evolving, and their survival capacity needs in vivo evaluation. This requires that the transfused platelets (PLTs) be distinguished from those of the recipient. Labeling at various biotin (Bio) densities allows one to concurrently trace multiple PLT populations, as reported for red blood cells. STUDY DESIGN AND METHODS A method is described to label human PLTs at two densities of Bio for future clinical trials. Injectable-grade PLTs were prepared in a sterile environment, using injectable-grade buffers and good manufacturing practices (GMP)-grade Sulfo-NHS-Biotin. Sulfo-NHS-Biotin concentrations were chosen to maintain PLT integrity and avoid potential alloimmunization while enabling the detection of circulating BioPLTs. The impact of biotinylation on human PLT recirculation was evaluated in vivo in a severe immunodeficient mouse model using ex vivo flow cytometry. RESULTS BioPLTs labeled with 1.2 or 10 μg/ml Sulfo-NHS-Biotin displayed normal ultrastructure and retained aggregation and secretion capacity and normal expression of the main surface glycoproteins. The procedure avoided detrimental PLT activation or apoptosis signals. Transfused human BioPLT populations could be distinguished from one another and from unlabeled circulating mouse PLTs, and their survival was comparable to that of unlabeled human PLTs in the mouse model. CONCLUSIONS Provided low Sulfo-NHS-Biotin concentrations (<10 μg/ml) are used, injectable-grade BioPLTs comply with safety regulations, conserve PLT integrity, and permit accurate in vivo detection. This alternative to radioisotopes, which allows one to follow different PLT populations in the same recipient, should be valuable when assessing new PC preparations and monitoring PLT survival in clinical research.
Collapse
Affiliation(s)
- Catherine Ravanat
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Anaïs Pongérard
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Monique Freund
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Véronique Heim
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Floriane Rudwill
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Catherine Ziessel
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Anita Eckly
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Fabienne Proamer
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Hervé Isola
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Christian Gachet
- Université de Strasbourg, INSERM, Etablissement Français du Sang (EFS) Grand-Est, BPPS UMR_S 1255, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| |
Collapse
|
46
|
Furuta Y, Pena-Ramos O, Li Z, Chiao L, Zhou Z. Calcium ions trigger the exposure of phosphatidylserine on the surface of necrotic cells. PLoS Genet 2021; 17:e1009066. [PMID: 33571185 PMCID: PMC7904182 DOI: 10.1371/journal.pgen.1009066] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 02/24/2021] [Accepted: 01/18/2021] [Indexed: 11/18/2022] Open
Abstract
Intracellular Ca2+ level is under strict regulation through calcium channels and storage pools including the endoplasmic reticulum (ER). Mutations in certain ion channel subunits, which cause mis-regulated Ca2+ influx, induce the excitotoxic necrosis of neurons. In the nematode Caenorhabditis elegans, dominant mutations in the DEG/ENaC sodium channel subunit MEC-4 induce six mechanosensory (touch) neurons to undergo excitotoxic necrosis. These necrotic neurons are subsequently engulfed and digested by neighboring hypodermal cells. We previously reported that necrotic touch neurons actively expose phosphatidylserine (PS), an “eat-me” signal, to attract engulfing cells. However, the upstream signal that triggers PS externalization remained elusive. Here we report that a robust and transient increase of cytoplasmic Ca2+ level occurs prior to the exposure of PS on necrotic touch neurons. Inhibiting the release of Ca2+ from the ER, either pharmacologically or genetically, specifically impairs PS exposure on necrotic but not apoptotic cells. On the contrary, inhibiting the reuptake of cytoplasmic Ca2+ into the ER induces ectopic necrosis and PS exposure. Remarkably, PS exposure occurs independently of other necrosis events. Furthermore, unlike in mutants of DEG/ENaC channels, in dominant mutants of deg-3 and trp-4, which encode Ca2+ channels, PS exposure on necrotic neurons does not rely on the ER Ca2+ pool. Our findings indicate that high levels of cytoplasmic Ca2+ are necessary and sufficient for PS exposure. They further reveal two Ca2+-dependent, necrosis-specific pathways that promote PS exposure, a “two-step” pathway initiated by a modest influx of Ca2+ and further boosted by the release of Ca2+ from the ER, and another, ER-independent, pathway. Moreover, we found that ANOH-1, the worm homolog of mammalian phospholipid scramblase TMEM16F, is necessary for efficient PS exposure in thapsgargin-treated worms and trp-4 mutants, like in mec-4 mutants. We propose that both the ER-mediated and ER-independent Ca2+ pathways promote PS externalization through activating ANOH-1. Necrosis is a type of cell death that exhibits distinct morphological features such as cell and organelle swelling. Necrotic cells expose phosphatidylserine (PS)–a type of phospholipid—on their outer surfaces. Receptor molecules on phagocytes detect PS on necrotic cells and subsequently initiate the engulfment process. As necrosis is associated with stroke, cancer, neurodegenerative diseases, and heart diseases, studying necrotic cell clearance has important medical relevance. In the model organism the nematode C. elegans, we previously identified membrane proteins that promote the exposure of PS on necrotic cell surfaces by studying neurons that are induced to undergo necrosis by dominant mutations in ion channels. Here, in C. elegans, we have discovered that the necrotic insults trigger an increase of the cytoplasmic calcium ion (Ca2+), which in turn promotes PS externalization on necrotic cell surfaces. Furthermore, we have identified two different mechanisms that increase cytoplasmic Ca2+ levels, one dependent on the Ca2+ contribution from the endoplasmic reticulum (ER), the other independent of the ER. The Ca2+ signal targets ANOH-1, a worm homolog of mammalian proteins capable of externalizing PS, for promoting PS exposure on necrotic cells. Our findings reveal novel upstream regulatory mechanisms that promote necrotic cell clearance in animals.
Collapse
Affiliation(s)
- Yoshitaka Furuta
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- School of Pharmacy, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, Japan
| | - Omar Pena-Ramos
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Zao Li
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Lucia Chiao
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Zheng Zhou
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
47
|
Repsold L, Pool R, Karodia M, Tintinger G, Becker P, Joubert AM. Apoptotic profiling of chronic myeloid leukaemia patients' platelets ex vivo before and after treatment with Imatinib. Cell Biochem Funct 2021; 39:562-570. [PMID: 33569808 DOI: 10.1002/cbf.3625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/18/2021] [Accepted: 01/26/2021] [Indexed: 11/05/2022]
Abstract
Chronic myeloid leukaemia (CML) is a malignancy of the haematopoietic stem cells. The first line of treatment for CML, especially in developing countries, remains the first-generation tyrosine kinase inhibitor, Imatinib. Patients with CML are frequently diagnosed with platelet abnormalities. However, the specific mechanism of platelet abnormalities in CML remains unclear and poorly understood. The aim of this study was therefore to determine the apoptotic profiles of CML patients ex vivo on platelets before and after treatment with Imatinib. Blood samples of healthy volunteers and CML patients at diagnosis and after 6 months treatment with Imatinib were collected. Platelet counts, viability and activation were determined. Results showed that CML patients' platelet counts were elevated upon diagnosis and these levels statistically significantly decreased after 6 months of treatment. Platelet activation was significantly increased after 6 months of treatment compared to levels at diagnosis (P-value < .05). Similarly, platelet apoptosis was also increased after 6 months of treatment. Abnormalities in platelet functioning found in this study may partly be due to clonal proliferation of haematopoietic cells in CML patients, specifically of megakaryocyte precursors as well as the inhibition of platelet tyrosine kinase's and the inhibition of platelet-derived growth factor.
Collapse
Affiliation(s)
- Lisa Repsold
- Department of Physiology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Roger Pool
- Department of Haematology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Mohammed Karodia
- Department of Haematology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Gregory Tintinger
- Department of Internal Medicine, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Piet Becker
- Research Office, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria, South Africa
| | - Anna Margaretha Joubert
- Department of Physiology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
48
|
The necroptotic cell death pathway operates in megakaryocytes, but not in platelet synthesis. Cell Death Dis 2021; 12:133. [PMID: 33510145 PMCID: PMC7843594 DOI: 10.1038/s41419-021-03418-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 02/06/2023]
Abstract
Necroptosis is a pro-inflammatory cell death program executed by the terminal effector, mixed lineage kinase domain-like (MLKL). Previous studies suggested a role for the necroptotic machinery in platelets, where loss of MLKL or its upstream regulator, RIPK3 kinase, impacted thrombosis and haemostasis. However, it remains unknown whether necroptosis operates within megakaryocytes, the progenitors of platelets, and whether necroptotic cell death might contribute to or diminish platelet production. Here, we demonstrate that megakaryocytes possess a functional necroptosis signalling cascade. Necroptosis activation leads to phosphorylation of MLKL, loss of viability and cell swelling. Analyses at steady state and post antibody-mediated thrombocytopenia revealed that platelet production was normal in the absence of MLKL, however, platelet activation and haemostasis were impaired with prolonged tail re-bleeding times. We conclude that MLKL plays a role in regulating platelet function and haemostasis and that necroptosis signalling in megakaryocytes is dispensable for platelet production.
Collapse
|
49
|
Goelz N, Eekels JJM, Pantic M, Kamber CT, Speer O, Franzoso FD, Schmugge M. Platelets express adaptor proteins of the extrinsic apoptosis pathway and can activate caspase-8. PLoS One 2021; 16:e0244848. [PMID: 33428668 PMCID: PMC7799768 DOI: 10.1371/journal.pone.0244848] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 12/17/2020] [Indexed: 01/22/2023] Open
Abstract
Background Apoptotic pathways in platelets are important for their survival and function. Platelet apoptosis may be involved in the pathogenesis of immune thrombocytopenia (ITP), an autoimmune-mediated disease. In contrast to the intrinsic apoptosis pathway, not much is known about the extrinsic pathway mechanisms in platelets. Objectives To investigate the expression of proteins involved in the extrinsic apoptosis pathway, including the death receptors, adaptor and regulator proteins in human platelets. To determine a possible trigger of the extrinsic apoptosis pathway in platelets. Methods To investigate the expression of key markers of the extrinsic pathway we used targeted immunofluorescence and flow cytometry assays. To study their expression and interaction we performed Western blotting and co-immunoprecipitation. Treated platelets with different apoptosis triggers were subjected to flow cytometry. Results We could identify the protein expression of the pro-apoptotic proteins TRADD (Tumor Necrosis Factor Receptor type 1- Associated DEATH Domain protein), TRAF2/5, (TNF Associated Factor) and DEDAF (Death Effector Domain- Associated Factor), FADD (Fas-Associated protein with death domain) as well as the anti-apoptotic proteins DJ-1 (Deglycase 1) and c-FLIP in human platelets. ABT-737 treatment induced a disruption in the co-localization of DJ-1 with FADD. Platelets treated with ABT-737 showed an activation in caspase-3 and -8. The exposure to TNF (Tumor Necrosis Factor), FasL (Fas ligand), and TWEAK or to plasma derived from ITP patients, did not lead to changes in caspase-3 and -8 activation in platelets. Conclusions Human platelets express some proteins of the extrinsic apoptosis pathway which can be modulated only by ABT-737 treatment. However so far, no other apoptosis trigger or interaction with an external receptor have been yet identified.
Collapse
Affiliation(s)
- Nadine Goelz
- Division of Haematology and Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Julia J. M. Eekels
- Division of Transfusion Medicine, University Hospital Greifswald, Greifswald, Germany
| | - Milica Pantic
- Division of Haematology and Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Christoph T. Kamber
- Division of Haematology and Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Oliver Speer
- Institute for Laboratory Medicine, Hospital Thurgau AG, Münsterlingen, Switzerland
| | - Francesca D. Franzoso
- Division of Haematology and Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Markus Schmugge
- Division of Haematology and Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
50
|
Wu J, Heemskerk JWM, Baaten CCFMJ. Platelet Membrane Receptor Proteolysis: Implications for Platelet Function. Front Cardiovasc Med 2021; 7:608391. [PMID: 33490118 PMCID: PMC7820117 DOI: 10.3389/fcvm.2020.608391] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
The activities of adhesion and signaling receptors in platelets are controlled by several mechanisms. An important way of regulation is provided by proteolytic cleavage of several of these receptors, leading to either a gain or a loss of platelet function. The proteases involved are of different origins and types: (i) present as precursor in plasma, (ii) secreted into the plasma by activated platelets or other blood cells, or (iii) intracellularly activated and cleaving cytosolic receptor domains. We provide a comprehensive overview of the proteases acting on the platelet membrane. We describe how these are activated, which are their target proteins, and how their proteolytic activity modulates platelet functions. The review focuses on coagulation-related proteases, plasmin, matrix metalloproteinases, ADAM(TS) isoforms, cathepsins, caspases, and calpains. We also describe how the proteolytic activities are determined by different platelet populations in a thrombus and conversely how proteolysis contributes to the formation of such populations.
Collapse
Affiliation(s)
- Jiayu Wu
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Johan W. M. Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
| | - Constance C. F. M. J. Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen, Aachen, Germany
| |
Collapse
|