1
|
Zhang JY, Xiang XN, Yu X, Liu Y, Jiang HY, Peng JL, He CQ, He HC. Mechanisms and applications of the regenerative capacity of platelets-based therapy in knee osteoarthritis. Biomed Pharmacother 2024; 178:117226. [PMID: 39079262 DOI: 10.1016/j.biopha.2024.117226] [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: 06/03/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/25/2024] Open
Abstract
Osteoarthritis (OA) is the most prevalent joint disease in the elderly population and its substantial morbidity and disability impose a heavy economic burden on patients and society. Knee osteoarthritis (KOA) is the most common subtype of OA, which is characterized by damage to progressive articular cartilage, synovitis, and subchondral bone sclerosis. Most current treatments for OA are palliative, primarily aim at symptom management, and do not prevent the progression of the disease or restore degraded cartilage. The activation of α-granules in platelets releases various growth factors that are involved in multiple stages of tissue repair, suggesting potential for disease modification. In recent years, platelet-based therapies, such as platelet-rich plasma, platelet-rich fibrin, and platelet lysates, have emerged as promising regenerative treatments for KOA, but their related effects and mechanisms are still unclear. Therefore, this review aims to summarize the biological characteristics and functions of platelets, classify the products of platelet-based therapy and related preparation methods. Moreover, we summarize the basic research of platelet-based regeneration strategies for KOA and discuss the cellular effects and molecular mechanisms. Further, we describe the general clinical application of platelet-based therapy in the treatment of KOA and the results of the meta-analysis of randomized controlled trials.
Collapse
Affiliation(s)
- Jiang-Yin Zhang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xiao-Na Xiang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xi Yu
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yan Liu
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Hong-Ying Jiang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Jia-Lei Peng
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Cheng-Qi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Hong-Chen He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
2
|
Pérez HJ, Crombet T. Notable correlation between serum epidermal growth factor values and inflammatory status in patients with COVID-19. Immun Inflamm Dis 2024; 12:e1355. [PMID: 39110087 PMCID: PMC11304898 DOI: 10.1002/iid3.1355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 07/04/2024] [Accepted: 07/12/2024] [Indexed: 08/10/2024] Open
Abstract
INTRODUCTION Despite its crucial role in Epidermal Growth Factor Receptor (EGFR) activation, and the resulting impact on the health-disease process, epidermal growth factor (EGF) is an underexplored molecule in relation to how its serum concentrations relate to other analytes and clinical variables in pathological contexts. OBJECTIVE To clarify the possible correlation between EGF and clinical and analytical variables in the context of COVID-19. METHODS Cross-sectional observational and analytical study, in patients with virological and clinical diagnosis of COVID-19, selected by simple random sampling, admitted between August and September 2021. UMELISA-EGF commercial kits were used. RESULTS Differences in overall EGF values were observed between groups (566.04 vs. 910.53 pg/ml, p = .0430). In COVID-19 patients, no notable correlations were observed for neutrophil, platelet, triglyceride or liver enzyme values (p > .05). Significant correlations were observed with the neutrophil-lymphocyte indicator (r = 0.4711, p = .0128) as well as with the platelet-lymphocyte index (r = 0.4553, p = .0155). Statistical results of multivariate regression analysis suggest NLR (β = .2232, p = .0353) and PLR (β = .2117, p = .0411) are predictors of inflammation in patients with COVID-19. CONCLUSIONS Serum EGF concentrations in COVID-19 correlate positively with prognostic inflammatory markers of severity and could presumably act as an independent risk factor for the development of inflammation in response to new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
Collapse
Affiliation(s)
- Héctor José Pérez
- Critical Care DivisionSaturnino Lora Provincial HospitalSantiago de CubaCuba
| | - Tania Crombet
- Clinical Trials DivisionCentre for Molecular ImmunologyHavanaCuba
| |
Collapse
|
3
|
Wang XB, Cui NH, Fang ZQ, Gao MJ, Cai D. Platelet bioenergetic profiling uncovers a metabolic pattern of high dependency on mitochondrial fatty acid oxidation in type 2 diabetic patients who developed in-stent restenosis. Redox Biol 2024; 72:103146. [PMID: 38579589 PMCID: PMC11000186 DOI: 10.1016/j.redox.2024.103146] [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: 03/21/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024] Open
Abstract
Although platelet bioenergetic dysfunction is evident early in the pathogenesis of diabetic macrovascular complications, the bioenergetic characteristics in type 2 diabetic patients who developed coronary in-stent restenosis (ISR) and their effects on platelet function remain unclear. Here, we performed platelet bioenergetic profiling to characterize the bioenergetic alterations in 28 type 2 diabetic patients with ISR compared with 28 type 2 diabetic patients without ISR (non-ISR) and 28 healthy individuals. Generally, platelets from type 2 diabetic patients with ISR exhibited a specific bioenergetic alteration characterized by high dependency on fatty acid (FA) oxidation, which subsequently induced complex III deficiency, causing decreased mitochondrial respiration, increased mitochondrial oxidant production, and low efficiency of mitochondrial ATP generation. This pattern of bioenergetic dysfunction showed close relationships with both α-granule and dense granule secretion as measured by surface P-selectin expression, ATP release, and profiles of granule cargo proteins in platelet releasates. Importantly, ex vivo reproduction of high dependency on FA oxidation by exposing non-ISR platelets to its agonist mimicked the bioenergetic dysfunction observed in ISR platelets and enhanced platelet secretion, whereas pharmaceutical inhibition of FA oxidation normalized the respiratory and redox states of ISR platelets and diminished platelet secretion. Further, causal mediation analyses identified a strong association between high dependency on FA oxidation and increased angiographical severity of ISR, which was significantly mediated by the status of platelet secretion. Our findings, for the first time, uncover a pattern of bioenergetic dysfunction in ISR and enhance current understanding of the mechanistic link of high dependency on FA oxidation to platelet abnormalities in the context of diabetes.
Collapse
Affiliation(s)
- Xue-Bin Wang
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Ning-Hua Cui
- Zhengzhou Key Laboratory of Children's Infection and Immunity, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zi-Qi Fang
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Mi-Jie Gao
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dan Cai
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| |
Collapse
|
4
|
Houlahan CB, Kong Y, Johnston B, Cielesh M, Chau TH, Fenwick J, Coleman PR, Hao H, Haltiwanger RS, Thaysen-Andersen M, Passam FH, Larance M. Analysis of the Healthy Platelet Proteome Identifies a New Form of Domain-Specific O-Fucosylation. Mol Cell Proteomics 2024; 23:100717. [PMID: 38237698 PMCID: PMC10879016 DOI: 10.1016/j.mcpro.2024.100717] [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: 07/14/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 02/17/2024] Open
Abstract
Platelet activation induces the secretion of proteins that promote platelet aggregation and inflammation. However, detailed analysis of the released platelet proteome is hampered by platelets' tendency to preactivate during their isolation and a lack of sensitive protocols for low abundance releasate analysis. Here, we detail the most sensitive analysis to date of the platelet releasate proteome with the detection of >1300 proteins. Unbiased scanning for posttranslational modifications within releasate proteins highlighted O-glycosylation as being a major component. For the first time, we detected O-fucosylation on previously uncharacterized sites including multimerin-1 (MMRN1), a major alpha granule protein that supports platelet adhesion to collagen and is a carrier for platelet factor V. The N-terminal elastin microfibril interface (EMI) domain of MMRN1, a key site for protein-protein interaction, was O-fucosylated at a conserved threonine within a new domain context. Our data suggest that either protein O-fucosyltransferase 1, or a novel protein O-fucosyltransferase, may be responsible for this modification. Mutating this O-fucose site on the EMI domain led to a >50% reduction of MMRN1 secretion, supporting a key role of EMI O-fucosylation in MMRN1 secretion. By comparing releasates from resting and thrombin-treated platelets, 202 proteins were found to be significantly released after high-dose thrombin stimulation. Complementary quantification of the platelet lysates identified >3800 proteins, which confirmed the platelet origin of releasate proteins by anticorrelation analysis. Low-dose thrombin treatment yielded a smaller subset of significantly regulated proteins with fewer secretory pathway enzymes. The extensive platelet proteome resource provided here (larancelab.com/platelet-proteome) allows identification of novel regulatory mechanisms for drug targeting to address platelet dysfunction and thrombosis.
Collapse
Affiliation(s)
- Callum B Houlahan
- The Heart Research Institute, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Yvonne Kong
- Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Bede Johnston
- The Heart Research Institute, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Michelle Cielesh
- Charles Perkins Centre, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - The Huong Chau
- School of Natural Sciences, Macquarie University, Macquarie Park, New South Wales, Australia
| | - Jemma Fenwick
- The Heart Research Institute, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia; Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul R Coleman
- The Heart Research Institute, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Huilin Hao
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Robert S Haltiwanger
- Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Morten Thaysen-Andersen
- School of Natural Sciences, Macquarie University, Macquarie Park, New South Wales, Australia; Institute for Glyco-Core Research, Nagoya University, Nagoya, Aichi, Japan
| | - Freda H Passam
- The Heart Research Institute, Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia; Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia.
| | - Mark Larance
- Charles Perkins Centre, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
5
|
Akbari P, Vuckovic D, Stefanucci L, Jiang T, Kundu K, Kreuzhuber R, Bao EL, Collins JH, Downes K, Grassi L, Guerrero JA, Kaptoge S, Knight JC, Meacham S, Sambrook J, Seyres D, Stegle O, Verboon JM, Walter K, Watkins NA, Danesh J, Roberts DJ, Di Angelantonio E, Sankaran VG, Frontini M, Burgess S, Kuijpers T, Peters JE, Butterworth AS, Ouwehand WH, Soranzo N, Astle WJ. A genome-wide association study of blood cell morphology identifies cellular proteins implicated in disease aetiology. Nat Commun 2023; 14:5023. [PMID: 37596262 PMCID: PMC10439125 DOI: 10.1038/s41467-023-40679-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/07/2023] [Indexed: 08/20/2023] Open
Abstract
Blood cells contain functionally important intracellular structures, such as granules, critical to immunity and thrombosis. Quantitative variation in these structures has not been subjected previously to large-scale genetic analysis. We perform genome-wide association studies of 63 flow-cytometry derived cellular phenotypes-including cell-type specific measures of granularity, nucleic acid content and reactivity-in 41,515 participants in the INTERVAL study. We identify 2172 distinct variant-trait associations, including associations near genes coding for proteins in organelles implicated in inflammatory and thrombotic diseases. By integrating with epigenetic data we show that many intracellular structures are likely to be determined in immature precursor cells. By integrating with proteomic data we identify the transcription factor FOG2 as an early regulator of platelet formation and α-granularity. Finally, we show that colocalisation of our associations with disease risk signals can suggest aetiological cell-types-variants in IL2RA and ITGA4 respectively mirror the known effects of daclizumab in multiple sclerosis and vedolizumab in inflammatory bowel disease.
Collapse
Affiliation(s)
- Parsa Akbari
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
- Department of Human Genetics, The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1HH, UK
- Medical Research Council Biostatistics Unit, University of Cambridge, East Forvie Building, Cambridge Biomedical Campus, Forvie Site, Robinson Way, Cambridge, CB2 0SR, UK
- The National Institute for Health and Care Research Blood and Transplant Unit in Donor Health and Genomics, Strangeways Research Laboratory, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
| | - Dragana Vuckovic
- Department of Human Genetics, The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1HH, UK
- The National Institute for Health and Care Research Blood and Transplant Unit in Donor Health and Genomics, Strangeways Research Laboratory, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Luca Stefanucci
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Centre, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Tao Jiang
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
- The National Institute for Health and Care Research Blood and Transplant Unit in Donor Health and Genomics, Strangeways Research Laboratory, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, CB2 0BB, UK
| | - Kousik Kundu
- Department of Human Genetics, The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1HH, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
| | - Roman Kreuzhuber
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
| | - Erik L Bao
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, 1 Blackfan Circle, Boston, MA, 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA, 02142, USA
- Harvard-MIT Health Sciences and Technology, Harvard Medical School, 77 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Janine H Collins
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Centre, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- Department of Haematology, Barts Health National Health Service Trust, London, E1 1BB, UK
| | - Kate Downes
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Centre, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
| | - Luigi Grassi
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Centre, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- National Institute for Health and Care Research Cambridge BioResource, Box 229, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Jose A Guerrero
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Centre, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
| | - Stephen Kaptoge
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
- The National Institute for Health and Care Research Blood and Transplant Unit in Donor Health and Genomics, Strangeways Research Laboratory, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, CB2 0BB, UK
| | - Julian C Knight
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Stuart Meacham
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Jennifer Sambrook
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- National Institute for Health and Care Research Cambridge BioResource, Box 229, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Denis Seyres
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Centre, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- National Institute for Health and Care Research Cambridge BioResource, Box 229, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
- European Molecular Biology Laboratory, Genome Biology Unit, 69117, Heidelberg, Germany
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Jeffrey M Verboon
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, 1 Blackfan Circle, Boston, MA, 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA, 02142, USA
| | - Klaudia Walter
- Department of Human Genetics, The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1HH, UK
| | - Nicholas A Watkins
- National Health Service Blood and Transplant, Cambridge Centre, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
- Department of Human Genetics, The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1HH, UK
- The National Institute for Health and Care Research Blood and Transplant Unit in Donor Health and Genomics, Strangeways Research Laboratory, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, CB2 0BB, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - David J Roberts
- The National Institute for Health and Care Research Blood and Transplant Unit in Donor Health and Genomics, Strangeways Research Laboratory, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Headley Way, Headington, Oxford, OX3 9DU, UK
- National Institute for Health Research Oxford Biomedical Research Centre-Haematology Theme, John Radcliffe Hospital, Headley Way, Headington, Oxford, OX3 9DU, UK
- National Health Service Blood and Transplant, Oxford Centre, John Radcliffe Hospital, Headley Way, Headington, Oxford, OX3 9DU, UK
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
- The National Institute for Health and Care Research Blood and Transplant Unit in Donor Health and Genomics, Strangeways Research Laboratory, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, CB2 0BB, UK
- Health Data Science Research Centre, Fondazione Human Technopole, Viale Rita Levi Montalcini 1, Milan, 20157, Italy
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, 1 Blackfan Circle, Boston, MA, 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave, Boston, MA, 02115, USA
- Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA, 02142, USA
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Centre, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, RILD Building, Barrack Road, Exeter, EX2 5DW, UK
| | - Stephen Burgess
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK
- Medical Research Council Biostatistics Unit, University of Cambridge, East Forvie Building, Cambridge Biomedical Campus, Forvie Site, Robinson Way, Cambridge, CB2 0SR, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, CB2 0BB, UK
| | - Taco Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, CB2 0PT, UK
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Sanquin, University of Amsterdam, Amsterdam, Netherlands
| | - James E Peters
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Department of Immunology and Inflammation, Imperial College London, Commonwealth Building, The Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK.
- The National Institute for Health and Care Research Blood and Transplant Unit in Donor Health and Genomics, Strangeways Research Laboratory, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK.
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, CB2 0BB, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK.
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK.
- National Health Service Blood and Transplant, Cambridge Centre, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK.
- Department of Haematology, University College London Hospitals, WC1E 6AS, London, UK.
| | - Nicole Soranzo
- Department of Human Genetics, The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1HH, UK.
- The National Institute for Health and Care Research Blood and Transplant Unit in Donor Health and Genomics, Strangeways Research Laboratory, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK.
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK.
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.
- Genomics Research Centre, Fondazione Human Technopole, Viale Rita Levi Montalcini 1, Milan, 20157, Italy.
| | - William J Astle
- Medical Research Council Biostatistics Unit, University of Cambridge, East Forvie Building, Cambridge Biomedical Campus, Forvie Site, Robinson Way, Cambridge, CB2 0SR, UK.
- The National Institute for Health and Care Research Blood and Transplant Unit in Donor Health and Genomics, Strangeways Research Laboratory, Strangeways Research Laboratory, University of Cambridge, Wort's Causeway, Cambridge, CB1 8RN, UK.
- National Health Service Blood and Transplant, Cambridge Centre, Cambridge Biomedical Campus, Long Road, Cambridge, CB2 0PT, UK.
| |
Collapse
|
6
|
Xulu KR, Augustine TN. Targeting Platelet Activation Pathways to Limit Tumour Progression: Current State of Affairs. Pharmaceuticals (Basel) 2022; 15:1532. [PMID: 36558983 PMCID: PMC9784118 DOI: 10.3390/ph15121532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
The association between cancer and a hypercoagulatory environment is well described. Thrombotic complications serve not only as a major mortality risk but the underlying molecular structure and function play significant roles in enhancing tumour progression, which is defined as the tumour's capacity to survive, invade and metastasise, amongst other hallmarks of the disease. The use of anticoagulant or antiplatelet drugs in cardiovascular disease lessens thrombotic effects, but the consequences on tumour progression require interrogation. Therefore, this review considered developments in the management of platelet activation pathways (thromboxane, ADP and thrombin), focusing on the use of Aspirin, Clopidogrel and Atopaxar, and their potential impacts on tumour progression. Published data suggested a cautionary tale in ensuring we adequately investigate not only drug-drug interactions but also those unforeseen reciprocal interactions between drugs and their targets within the tumour microenvironment that may act as selective pressures, enhancing tumour survival and progression.
Collapse
Affiliation(s)
- Kutlwano R. Xulu
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| | - Tanya N. Augustine
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa
| |
Collapse
|
7
|
Platelet Reactivity and Inflammatory Phenotype Induced by Full-Length Spike SARS-CoV-2 Protein and Its RBD Domain. Int J Mol Sci 2022; 23:ijms232315191. [PMID: 36499540 PMCID: PMC9738415 DOI: 10.3390/ijms232315191] [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: 09/30/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 12/08/2022] Open
Abstract
A state of immunothrombosis has been reported in COVID-19. Platelets actively participate in this process. However, little is known about the ability of SARS-CoV-2 virus proteins to induce platelet activity. Platelet-rich plasma (PRP) was incubated with spike full-length protein and the RBD domain in independent assays. We evaluated platelet activation through the expression of P-selectin and activation of glicoprotein IIbIIIa (GP IIbIIIa), determined by flow cytometry and the ability of the proteins to induce platelet aggregation. We determined concentrations of immunothrombotic biomarkers in PRP supernatant treated with the proteins. We determined that the spike full-length proteins and the RBD domain induced an increase in P-selectin expression and GP IIbIIIa activation (p < 0.0001). We observed that the proteins did not induce platelet aggregation, but favored a pro-aggregating state that, in response to minimal doses of collagen, could re-establish the process (p < 0.0001). On the other hand, the viral proteins stimulated the release of interleukin 6, interleukin 8, P-selectin and the soluble fraction of CD40 ligand (sCD40L), molecules that favor an inflammatory state p < 0.05. These results indicate that the spike full-length protein and its RBD domain can induce platelet activation favoring an inflammatory phenotype that might contribute to the development of an immunothrombotic state.
Collapse
|
8
|
Tiedemann K, Tsao S, Komarova SV. Platelets and osteoblasts: secretome connections. Am J Physiol Cell Physiol 2022; 323:C347-C353. [PMID: 35675640 DOI: 10.1152/ajpcell.00187.2022] [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: 11/22/2022]
Abstract
Megakaryocyte hyperplasia associated with myeloproliferative neoplasms commonly leads to abnormal bone tissue deposition in the bone marrow, known as osteosclerosis. In this study, we aimed to synthesize the known proteomics literature describing factors released by megakaryocytes and platelets and to examine if any of the secreted factors have a known ability to stimulate the bone-forming cells, osteoblasts. Using a systematic search of Medline, we identified 77 articles reporting on factors secreted by platelets and megakaryocytes. After a full-text screening and analysis of the studies, we selected seven papers that reported proteomics data for factors secreted by platelets from healthy individuals. From 60 proteins reported in at least two studies, we focused on 23 that contained a putative signal peptide, which we searched for a potential osteoblast-stimulatory function. From nine proteins with a positive effect on osteoblast formation and function, two extracellular matrix (ECM) proteins, secreted protein acidic and rich in cysteine (SPARC) and tissue inhibitor of metalloproteinase-1 (TIMP1), and three cellular proteins with known extracellular function, the 70-kDa heat shock protein (HSP70), thymosin-β4 (TB4), and super dismutase (SOD), were identified as hypothetical candidate molecules to be examined as potential mediators in mouse models of osteomyelofibrosis. Thus, careful analysis of prior literature can be beneficial in assisting the planning of future experimental studies.
Collapse
Affiliation(s)
- Kerstin Tiedemann
- Faculty of Dental Medicine and Oral Health Sciences, Shriners Hospital for Children-Canada, Montreal, Quebec, Canada
| | - Serena Tsao
- Faculty of Dental Medicine and Oral Health Sciences, Shriners Hospital for Children-Canada, Montreal, Quebec, Canada
| | - Svetlana V Komarova
- Faculty of Dental Medicine and Oral Health Sciences, Shriners Hospital for Children-Canada, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Suades R, Padró T, Vilahur G, Badimon L. Platelet-released extracellular vesicles: the effects of thrombin activation. Cell Mol Life Sci 2022; 79:190. [PMID: 35288766 PMCID: PMC8920058 DOI: 10.1007/s00018-022-04222-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/17/2022] [Accepted: 02/23/2022] [Indexed: 12/13/2022]
Abstract
Platelets exert fundamental roles in thrombosis, inflammation, and angiogenesis, contributing to different pathologies from cardiovascular diseases to cancer. We previously reported that platelets release extracellular vesicles (pEVs) which contribute to thrombus formation. However, pEV composition remains poorly defined. Indeed, pEV quality and type, rather than quantity, may be relevant in intravascular cross-talk with either circulating or vascular cells. We aimed to define the phenotypic characteristics of pEVs released spontaneously and those induced by thrombin activation to better understand their role in disease dissemination. pEVs obtained from washed platelets from healthy donor blood were characterized by flow cytometry. pEVs from thrombin-activated platelets (T-pEVs) showed higher levels of P-selectin and active form of glycoprotein IIb/IIIa than baseline non-activated platelets (B-pEVs). Following mass spectrometry-based differential proteomic analysis, significant changes in the abundance of proteins secreted in T-pEVs compared to B-pEVs were found. These differential proteins were involved in coagulation, adhesion, cytoskeleton, signal transduction, metabolism, and vesicle-mediated transport. Interestingly, release of proteins relevant for cell adhesion, intrinsic pathway coagulation, and platelet activation signalling was significantly modified by thrombin stimulation. A novel pEV-associated protein (protocadherin-α4) was found to be significantly reduced in T-pEVs showing a shift towards increased expression in the membranes of activated platelets. In summary, platelet activation induced by thrombin triggers the shedding of pEVs with a complex proteomic pattern rich in procoagulant and proadhesive proteins. Crosstalk with other vascular and blood cells in a paracrine regulatory mode could extend the prothrombotic signalling as well as promote proteostasic changes in other cellular types.
Collapse
Affiliation(s)
- Rosa Suades
- Cardiovascular Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB Sant Pau, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
| | - Teresa Padró
- Cardiovascular Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB Sant Pau, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB Sant Pau, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Program ICCC, Research Institute Hospital Santa Creu i Sant Pau, IIB Sant Pau, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain.
- CIBERCV Instituto de Salud Carlos III, Madrid, Spain.
- Cardiovascular Research Chair, UAB, Barcelona, Spain.
| |
Collapse
|
10
|
Heinzmann ACA, Coenen DM, Vajen T, Cosemans JMEM, Koenen RR. Combined Antiplatelet Therapy Reduces the Proinflammatory Properties of Activated Platelets. TH OPEN 2021; 5:e533-e542. [PMID: 34901735 PMCID: PMC8651446 DOI: 10.1055/a-1682-3415] [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: 06/18/2021] [Accepted: 10/22/2020] [Indexed: 11/17/2022] Open
Abstract
The cause of atherothrombosis is rupture or erosion of atherosclerotic lesions, leading to an increased risk of myocardial infarction or stroke. Here, platelet activation plays a major role, leading to the release of bioactive molecules, for example, chemokines and coagulation factors, and to platelet clot formation. Several antiplatelet therapies have been developed for secondary prevention of cardiovascular events, in which anticoagulant drugs are often combined. Besides playing a role in hemostasis, platelets are also involved in inflammation. However, it is unclear whether current antiplatelet therapies also affect platelet immune functions. In this study, the possible anti-inflammatory effects of antiplatelet medications on chemokine release were investigated using enzyme-linked immunosorbent assay and on the chemotaxis of THP-1 cells toward platelet releasates. We found that antiplatelet medication acetylsalicylic acid (ASA) led to reduced chemokine (CC motif) ligand 5 (CCL5) and chemokine (CXC motif) ligand 4 (CXCL4) release from platelets, while leukocyte chemotaxis was not affected. Depending on the agonist, α
IIb
β
3
and P2Y
12
inhibitors also affected CCL5 or CXCL4 release. The combination of ASA with a P2Y
12
inhibitor or a phosphodiesterase (PDE) inhibitor did not lead to an additive reduction in CCL5 or CXCL4 release. Interestingly, these combinations did reduce leukocyte chemotaxis. This study provides evidence that combined therapy of ASA and a P2Y
12
or PDE3 inhibitor can decrease the inflammatory leukocyte recruiting potential of the releasate of activated platelets.
Collapse
Affiliation(s)
- Alexandra C A Heinzmann
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Daniëlle M Coenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands.,Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Tanja Vajen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands.,Cardiovascular Research Laboratory, Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Judith M E M Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
11
|
Smith CW. Release of α-granule contents during platelet activation. Platelets 2021; 33:491-502. [PMID: 34569425 DOI: 10.1080/09537104.2021.1913576] [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: 10/20/2022]
Abstract
Upon activation, platelets release a plethora of factors which help to mediate their dynamic functions in hemostasis, inflammation, wound healing, tumor metastasis and angiogenesis. The majority of these bioactive molecules are released from α-granules, which are unique to platelets, and contain an incredibly diverse repertoire of cargo including; integral membrane proteins, pro-coagulant molecules, chemokines, mitogenic, growth and angiogenic factors, adhesion proteins, and microbicidal proteins. Clinically, activation of circulating platelets has increasingly been associated with various disease states. Biomarkers indicating the level of platelet activation in patients can therefore be useful tools to evaluate risk factors to predict future complications and determine treatment strategies or evaluate antiplatelet therapy. The irreversible nature of α-granule secretion makes it ideally suited as a marker of platelet activation. This review outlines the release and contents of platelet α-granules, as well as the membrane bound, and soluble α-granule cargo proteins that can be used as biomarkers of platelet activation.
Collapse
Affiliation(s)
- Christopher W Smith
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| |
Collapse
|
12
|
Coenen DM, Heinzmann ACA, Oggero S, Albers HJ, Nagy M, Hagué P, Kuijpers MJE, Vanderwinden JM, van der Meer AD, Perretti M, Koenen RR, Cosemans JMEM. Inhibition of Phosphodiesterase 3A by Cilostazol Dampens Proinflammatory Platelet Functions. Cells 2021; 10:1998. [PMID: 34440764 PMCID: PMC8392606 DOI: 10.3390/cells10081998] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE platelets possess not only haemostatic but also inflammatory properties, which combined are thought to play a detrimental role in thromboinflammatory diseases such as acute coronary syndromes and stroke. Phosphodiesterase (PDE) 3 and -5 inhibitors have demonstrated efficacy in secondary prevention of arterial thrombosis, partially mediated by their antiplatelet action. Yet it is unclear whether such inhibitors also affect platelets' inflammatory functions. Here, we aimed to examine the effect of the PDE3A inhibitor cilostazol and the PDE5 inhibitor tadalafil on platelet function in various aspects of thromboinflammation. Approach and results: cilostazol, but not tadalafil, delayed ex vivo platelet-dependent fibrin formation under whole blood flow over type I collagen at 1000 s-1. Similar results were obtained with blood from Pde3a deficient mice, indicating that cilostazol effects are mediated via PDE3A. Interestingly, cilostazol specifically reduced the release of phosphatidylserine-positive extracellular vesicles (EVs) from human platelets while not affecting total EV release. Both cilostazol and tadalafil reduced the interaction of human platelets with inflamed endothelium under arterial flow and the release of the chemokines CCL5 and CXCL4 from platelets. Moreover, cilostazol, but not tadalafil, reduced monocyte recruitment and platelet-monocyte interaction in vitro. CONCLUSIONS this study demonstrated yet unrecognised roles for platelet PDE3A and platelet PDE5 in platelet procoagulant and proinflammatory responses.
Collapse
Affiliation(s)
- Daniëlle M. Coenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (D.M.C.); (A.C.A.H.); (M.N.); (M.J.E.K.); (R.R.K.)
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Alexandra C. A. Heinzmann
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (D.M.C.); (A.C.A.H.); (M.N.); (M.J.E.K.); (R.R.K.)
| | - Silvia Oggero
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK; (S.O.); (M.P.)
| | - Hugo J. Albers
- BIOS Lab-on-a-Chip Group, Technical Medical Centre, MESA+ Institute for Nanotechnology, University of Twente, 7522 NB Enschede, The Netherlands;
- Applied Stem Cell Technologies Group, Technical Medical Centre, University of Twente, 7522 NB Enschede, The Netherlands;
| | - Magdolna Nagy
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (D.M.C.); (A.C.A.H.); (M.N.); (M.J.E.K.); (R.R.K.)
| | - Perrine Hagué
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, B-1070 Brussels, Belgium; (P.H.); (J.-M.V.)
| | - Marijke J. E. Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (D.M.C.); (A.C.A.H.); (M.N.); (M.J.E.K.); (R.R.K.)
| | - Jean-Marie Vanderwinden
- Laboratory of Neurophysiology, Faculty of Medicine, Université Libre de Bruxelles, B-1070 Brussels, Belgium; (P.H.); (J.-M.V.)
| | - Andries D. van der Meer
- Applied Stem Cell Technologies Group, Technical Medical Centre, University of Twente, 7522 NB Enschede, The Netherlands;
| | - Mauro Perretti
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK; (S.O.); (M.P.)
| | - Rory R. Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (D.M.C.); (A.C.A.H.); (M.N.); (M.J.E.K.); (R.R.K.)
| | - Judith M. E. M. Cosemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 ER Maastricht, The Netherlands; (D.M.C.); (A.C.A.H.); (M.N.); (M.J.E.K.); (R.R.K.)
| |
Collapse
|
13
|
Braun A, Anders HJ, Gudermann T, Mammadova-Bach E. Platelet-Cancer Interplay: Molecular Mechanisms and New Therapeutic Avenues. Front Oncol 2021; 11:665534. [PMID: 34322381 PMCID: PMC8311658 DOI: 10.3389/fonc.2021.665534] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/17/2021] [Indexed: 12/18/2022] Open
Abstract
Although platelets are critically involved in thrombosis and hemostasis, experimental and clinical evidence indicate that platelets promote tumor progression and metastasis through a wide range of physical and functional interactions between platelets and cancer cells. Thrombotic and thromboembolic events are frequent complications in patients with solid tumors. Hence, cancer modulates platelet function by directly inducing platelet-tumor aggregates and triggering platelet granule release and altering platelet turnover. Also, platelets enhance tumor cell dissemination by activating endothelial cell function and recruiting immune cells to primary and metastatic tumor sites. In this review, we summarize current knowledge on the complex interactions between platelets and tumor cells and the host microenvironment. We also critically discuss the potential of anti-platelet agents for cancer prevention and treatment.
Collapse
Affiliation(s)
- Attila Braun
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Munich, Germany
| | - Thomas Gudermann
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Elmina Mammadova-Bach
- Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilian-University, Member of the German Center for Lung Research (DZL), Munich, Germany.,Division of Nephrology, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Munich, Germany
| |
Collapse
|
14
|
Huang J, Swieringa F, Solari FA, Provenzale I, Grassi L, De Simone I, Baaten CCFMJ, Cavill R, Sickmann A, Frontini M, Heemskerk JWM. Assessment of a complete and classified platelet proteome from genome-wide transcripts of human platelets and megakaryocytes covering platelet functions. Sci Rep 2021; 11:12358. [PMID: 34117303 PMCID: PMC8196183 DOI: 10.1038/s41598-021-91661-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
Novel platelet and megakaryocyte transcriptome analysis allows prediction of the full or theoretical proteome of a representative human platelet. Here, we integrated the established platelet proteomes from six cohorts of healthy subjects, encompassing 5.2 k proteins, with two novel genome-wide transcriptomes (57.8 k mRNAs). For 14.8 k protein-coding transcripts, we assigned the proteins to 21 UniProt-based classes, based on their preferential intracellular localization and presumed function. This classified transcriptome-proteome profile of platelets revealed: (i) Absence of 37.2 k genome-wide transcripts. (ii) High quantitative similarity of platelet and megakaryocyte transcriptomes (R = 0.75) for 14.8 k protein-coding genes, but not for 3.8 k RNA genes or 1.9 k pseudogenes (R = 0.43-0.54), suggesting redistribution of mRNAs upon platelet shedding from megakaryocytes. (iii) Copy numbers of 3.5 k proteins that were restricted in size by the corresponding transcript levels (iv) Near complete coverage of identified proteins in the relevant transcriptome (log2fpkm > 0.20) except for plasma-derived secretory proteins, pointing to adhesion and uptake of such proteins. (v) Underrepresentation in the identified proteome of nuclear-related, membrane and signaling proteins, as well proteins with low-level transcripts. We then constructed a prediction model, based on protein function, transcript level and (peri)nuclear localization, and calculated the achievable proteome at ~ 10 k proteins. Model validation identified 1.0 k additional proteins in the predicted classes. Network and database analysis revealed the presence of 2.4 k proteins with a possible role in thrombosis and hemostasis, and 138 proteins linked to platelet-related disorders. This genome-wide platelet transcriptome and (non)identified proteome database thus provides a scaffold for discovering the roles of unknown platelet proteins in health and disease.
Collapse
Affiliation(s)
- Jingnan Huang
- Department of Biochemistry, CARIM, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-E.V, Dortmund, Germany.
| | - Frauke Swieringa
- Department of Biochemistry, CARIM, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-E.V, Dortmund, Germany
| | - Fiorella A Solari
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-E.V, Dortmund, Germany
| | - Isabella Provenzale
- Department of Biochemistry, CARIM, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Luigi Grassi
- Department of Haematology, University of Cambridge, National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge, UK
| | - Ilaria De Simone
- Department of Biochemistry, CARIM, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Constance C F M J Baaten
- Department of Biochemistry, CARIM, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH, Aachen, Germany
| | - Rachel Cavill
- Department of Data Science and Knowledge Engineering, FSE, Maastricht University, Maastricht, The Netherlands
| | - Albert Sickmann
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-E.V, Dortmund, Germany
- Medizinische Fakultät, Medizinische Proteom-Center, Ruhr-Universität Bochum, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, UK
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, National Health Service Blood and Transplant (NHSBT), Cambridge Biomedical Campus, Cambridge, UK
- Institute of Biomedical & Clinical Science, College of Medicine and Health, University of Exeter Medical School, Exeter, UK
| | - Johan W M Heemskerk
- Department of Biochemistry, CARIM, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| |
Collapse
|
15
|
Hellgren G, Lundgren P, Pivodic A, Löfqvist C, Nilsson AK, Ley D, Sävman K, Smith LE, Hellström A. Decreased Platelet Counts and Serum Levels of VEGF-A, PDGF-BB, and BDNF in Extremely Preterm Infants Developing Severe ROP. Neonatology 2021; 118:18-27. [PMID: 33611321 PMCID: PMC10539685 DOI: 10.1159/000512282] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 09/02/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Thrombocytopenia has been identified as an independent risk factor for retinopathy of prematurity (ROP), although underlying mechanisms are unknown. In this study, the association of platelet count and serum platelet-derived factors with ROP was investigated. METHODS Data for 78 infants born at gestational age (GA) <28 weeks were included. Infants were classified as having no/mild ROP or severe ROP. Serum levels of vascular endothelial growth factor A, platelet-derived growth factor BB, and brain-derived neurotrophic factor were measured in serum samples collected from birth until postmenstrual age (PMA) 40 weeks. Platelet counts were obtained from samples taken for clinical indication. RESULTS Postnatal platelet counts and serum concentrations of the 3 growth factors followed the same postnatal pattern, with lower levels in infants developing severe ROP at PMA 32 and 36 weeks (p < 0.05-0.001). With adjustment for GA, low platelet counts and low serum concentrations of all 3 factors at PMA 32 weeks were significantly associated with severe ROP. Serum concentrations of all 3 factors also strongly correlated with platelet count (p < 0.001). CONCLUSION In this article, we show that ROP, platelet counts, and specific pro-angiogenic factors correlate. These data suggest that platelet-released factors might be involved in the regulation of retinal and systemic angiogenesis after extremely preterm birth. Further investigations are needed.
Collapse
Affiliation(s)
- Gunnel Hellgren
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,
| | - Pia Lundgren
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Aldina Pivodic
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Chatarina Löfqvist
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Health and Care Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders K Nilsson
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - David Ley
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Karin Sävman
- Department of Pediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Neonatology, The Queen Silvia Children's Hospital, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lois E Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ann Hellström
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
16
|
Balkenhol J, Kaltdorf KV, Mammadova-Bach E, Braun A, Nieswandt B, Dittrich M, Dandekar T. Comparison of the central human and mouse platelet signaling cascade by systems biological analysis. BMC Genomics 2020; 21:897. [PMID: 33353544 PMCID: PMC7756956 DOI: 10.1186/s12864-020-07215-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022] Open
Abstract
Background Understanding the molecular mechanisms of platelet activation and aggregation is of high interest for basic and clinical hemostasis and thrombosis research. The central platelet protein interaction network is involved in major responses to exogenous factors. This is defined by systemsbiological pathway analysis as the central regulating signaling cascade of platelets (CC). Results The CC is systematically compared here between mouse and human and major differences were found. Genetic differences were analysed comparing orthologous human and mouse genes. We next analyzed different expression levels of mRNAs. Considering 4 mouse and 7 human high-quality proteome data sets, we identified then those major mRNA expression differences (81%) which were supported by proteome data. CC is conserved regarding genetic completeness, but we observed major differences in mRNA and protein levels between both species. Looking at central interactors, human PLCB2, MMP9, BDNF, ITPR3 and SLC25A6 (always Entrez notation) show absence in all murine datasets. CC interactors GNG12, PRKCE and ADCY9 occur only in mice. Looking at the common proteins, TLN1, CALM3, PRKCB, APP, SOD2 and TIMP1 are higher abundant in human, whereas RASGRP2, ITGB2, MYL9, EIF4EBP1, ADAM17, ARRB2, CD9 and ZYX are higher abundant in mouse. Pivotal kinase SRC shows different regulation on mRNA and protein level as well as ADP receptor P2RY12. Conclusions Our results highlight species-specific differences in platelet signaling and points of specific fine-tuning in human platelets as well as murine-specific signaling differences. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-020-07215-4.
Collapse
Affiliation(s)
- Johannes Balkenhol
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Am Hubland, University of Würzburg, D-97074, Würzburg, Germany
| | - Kristin V Kaltdorf
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Am Hubland, University of Würzburg, D-97074, Würzburg, Germany
| | - Elmina Mammadova-Bach
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Centre, University of Würzburg, Würzburg, Germany.,Present address: Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig, Maximilian University of Munich, D-80336, Munich, Germany
| | - Attila Braun
- Member of the German Center for Lung Research (DZL), Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and Rudolf Virchow Centre, University of Würzburg, Würzburg, Germany
| | - Marcus Dittrich
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Am Hubland, University of Würzburg, D-97074, Würzburg, Germany.,Dept of Genetics, Biocenter, Am Hubland, University of Würzburg, Am Hubland, D 97074, Würzburg, Germany
| | - Thomas Dandekar
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Am Hubland, University of Würzburg, D-97074, Würzburg, Germany.
| |
Collapse
|
17
|
Platelets in Healthy and Disease States: From Biomarkers Discovery to Drug Targets Identification by Proteomics. Int J Mol Sci 2020; 21:ijms21124541. [PMID: 32630608 PMCID: PMC7352998 DOI: 10.3390/ijms21124541] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/15/2020] [Accepted: 06/24/2020] [Indexed: 12/16/2022] Open
Abstract
Platelets are a heterogeneous small anucleate blood cell population with a central role both in physiological haemostasis and in pathological states, spanning from thrombosis to inflammation, and cancer. Recent advances in proteomic studies provided additional important information concerning the platelet biology and the response of platelets to several pathophysiological pathways. Platelets circulate systemically and can be easily isolated from human samples, making proteomic application very interesting for characterizing the complexity of platelet functions in health and disease as well as for identifying and quantifying potential platelet proteins as biomarkers and novel antiplatelet therapeutic targets. To date, the highly dynamic protein content of platelets has been studied in resting and activated platelets, and several subproteomes have been characterized including platelet-derived microparticles, platelet granules, platelet releasates, platelet membrane proteins, and specific platelet post-translational modifications. In this review, a critical overview is provided on principal platelet proteomic studies focused on platelet biology from signaling to granules content, platelet proteome changes in several diseases, and the impact of drugs on platelet functions. Moreover, recent advances in quantitative platelet proteomics are discussed, emphasizing the importance of targeted quantification methods for more precise, robust and accurate quantification of selected proteins, which might be used as biomarkers for disease diagnosis, prognosis and therapy, and their strong clinical impact in the near future.
Collapse
|
18
|
Suzuki‐Inoue K, Tsukiji N. Platelet CLEC-2 and lung development. Res Pract Thromb Haemost 2020; 4:481-490. [PMID: 32548549 PMCID: PMC7292670 DOI: 10.1002/rth2.12338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/05/2020] [Accepted: 02/08/2020] [Indexed: 01/23/2023] Open
Abstract
In this article, the State of the Art lecture "Platelet CLEC-2 and Lung Development" presented at the ISTH congress 2019 is reviewed. During embryonic development, blood cells are often considered as porters of nutrition and oxygen but not as active influencers of cell differentiation. However, recent studies revealed that platelets actively facilitate cell differentiation by releasing biological substances during development. C-type lectin-like receptor 2 (CLEC-2) has been identified as a receptor for the platelet-activating snake venom rhodocytin. An endogenous ligand of CLEC-2 is the membrane protein podoplanin (PDPN), which is expressed on the surface of certain types of tumor cells and lymphatic endothelial cells (LECs). Deletion of CLEC-2 from platelets in mice results in death just after birth due to lung malformation and blood/lymphatic vessel separation. During development, lymphatic vessels are derived from cardinal veins. At this stage, platelets are activated by binding of CLEC-2 to LEC PDPN and release trandforming growth factor-β (TGF-β). This cytokine inhibits LEC migration and proliferation, facilitating blood/lymphatic vessel separation. TGF-β released upon platelet-expressed CLEC-2/LEC PDPN also facilitates differentiation of lung mesothelial cells into alveolar duct myofibroblasts (adMYFs) in the developing lung. AdMYFs generate elastic fibers inside the lung, so that the lung can be properly inflated. Thus, platelets act as an ultimate natural drug delivery system that enables biological substances to be specifically delivered to the target at high concentrations by receptor/ligand interactions during development.
Collapse
Affiliation(s)
- Katsue Suzuki‐Inoue
- Department of Clinical and Laboratory MedicineFaculty of MedicineUniversity of YamanashiChuoJapan
| | - Nagaharu Tsukiji
- Department of Clinical and Laboratory MedicineFaculty of MedicineUniversity of YamanashiChuoJapan
| |
Collapse
|
19
|
Montague SJ, Lim YJ, Lee WM, Gardiner EE. Imaging Platelet Processes and Function-Current and Emerging Approaches for Imaging in vitro and in vivo. Front Immunol 2020; 11:78. [PMID: 32082328 PMCID: PMC7005007 DOI: 10.3389/fimmu.2020.00078] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 01/13/2020] [Indexed: 12/22/2022] Open
Abstract
Platelets are small anucleate cells that are essential for many biological processes including hemostasis, thrombosis, inflammation, innate immunity, tumor metastasis, and wound healing. Platelets circulate in the blood and in order to perform all of their biological roles, platelets must be able to arrest their movement at an appropriate site and time. Our knowledge of how platelets achieve this has expanded as our ability to visualize and quantify discreet platelet events has improved. Platelets are exquisitely sensitive to changes in blood flow parameters and so the visualization of rapid intricate platelet processes under conditions found in flowing blood provides a substantial challenge to the platelet imaging field. The platelet's size (~2 μm), rapid activation (milliseconds), and unsuitability for genetic manipulation, means that appropriate imaging tools are limited. However, with the application of modern imaging systems to study platelet function, our understanding of molecular events mediating platelet adhesion from a single-cell perspective, to platelet recruitment and activation, leading to thrombus (clot) formation has expanded dramatically. This review will discuss current platelet imaging techniques in vitro and in vivo, describing how the advancements in imaging have helped answer/expand on platelet biology with a particular focus on hemostasis. We will focus on platelet aggregation and thrombus formation, and how platelet imaging has enhanced our understanding of key events, highlighting the knowledge gained through the application of imaging modalities to experimental models in vitro and in vivo. Furthermore, we will review the limitations of current imaging techniques, and questions in thrombosis research that remain to be addressed. Finally, we will speculate how the same imaging advancements might be applied to the imaging of other vascular cell biological functions and visualization of dynamic cell-cell interactions.
Collapse
Affiliation(s)
- Samantha J. Montague
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Yean J. Lim
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- Research School of Electrical, Energy and Materials Engineering, The Australian National University, Canberra, ACT, Australia
| | - Woei M. Lee
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
- Research School of Electrical, Energy and Materials Engineering, The Australian National University, Canberra, ACT, Australia
| | - Elizabeth E. Gardiner
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
20
|
Proteomic profiling of the thrombin-activated canine platelet secretome (CAPS). PLoS One 2019; 14:e0224891. [PMID: 31721811 PMCID: PMC6853320 DOI: 10.1371/journal.pone.0224891] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/23/2019] [Indexed: 11/27/2022] Open
Abstract
Domestic dogs share the same environment as humans, and they represent a valuable animal model to study naturally-occurring human disease. Platelet proteomics holds promise for the discovery of biomarkers that capture the contribution of platelets to the pathophysiology of many disease states, however, canine platelet proteomic studies are lacking. Our study objectives were to establish a protocol for proteomic identification and quantification of the thrombin-activated canine platelet secretome (CAPS), and to compare the CAPS proteins to human and murine platelet proteomic data. Washed platelets were isolated from healthy dogs, and stimulated with saline (control) or gamma-thrombin (releasate). Proteins were separated by SDS-page, trypsin-digested and analyzed by liquid chromatography and tandem mass spectrometry (MS). CAPS proteins were defined as those with a MS1-abundance ratio of two or more for releasate vs. unstimulated saline control. A total of 1,918 proteins were identified, with 908 proteins common to all dogs and 693 characterized as CAPS proteins. CAPS proteins were similar to human and murine platelet secretomes and were highly represented in hemostatic pathways. Differences unique to CAPS included replacement of platelet factor 4 with other cleavage products of platelet basic protein (e.g. interleukin-8), novel proteins (e.g. C-C motif chemokine 14), and proteins in relatively high (e.g. protease nexin-1) or low (e.g. von Willebrand factor) abundance. This study establishes the first in-depth platelet releasate proteome from healthy dogs with a reference database of 693 CAPS proteins. Similarities between CAPS and the human secretome confirm the utility of dogs as translational models of human disease, but we also identify differences unique to canine platelets. Our findings provide a resource for further investigations into disease-related CAPS profiles, and for comparative pathway analyses of platelet activation among species.
Collapse
|
21
|
|
22
|
|
23
|
Szklanna PB, Parsons ME, Wynne K, O'Connor H, Egan K, Allen S, Ní Áinle F, Maguire PB. The Platelet Releasate is Altered in Human Pregnancy. Proteomics Clin Appl 2018; 13:e1800162. [PMID: 30318839 DOI: 10.1002/prca.201800162] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 09/28/2018] [Indexed: 01/09/2023]
Abstract
PURPOSE Healthy pregnancy is characterized by an increase in platelet activation and a decrease in the number of circulating platelets with gestation. Despite this recognized importance, proteomic studies investigating platelets in healthy pregnancy have not been performed. As platelet cargo can be altered in different conditions, it is hypothesized that platelets may store a relevant and bespoke collection of molecules during pregnancy. EXPERIMENTAL DESIGN Comparative label-free quantitative proteomic profiling of platelet releasates (PRs) is performed from 18 healthy pregnant and 13 non-pregnant women using an MS/MS approach. RESULTS Of the 723 proteins identified, 69 PR proteins are found to be differentially released from platelets in pregnancy, including proteins only expressed during pregnancy such as pregnancy-specific glycoproteins and human placental lactogen. Moreover, the population of exosomal vesicles present in the PR is also modified in pregnancy. Receiver operating characteristic analysis shows the predictive ability of 11 PR proteins to distinctly classify pregnant and nonpregnant women with an area under the curve of 0.876, a sensitivity of 88.9%, and a specificity of 84.6%. CONCLUSIONS AND CLINICAL RELEVANCE Taken together this demonstrates that platelets and their released cargo are 'educated' in physiologic stressful conditions such as pregnancy and may represent a promising platform to study pregnancy complications.
Collapse
Affiliation(s)
- Paulina B Szklanna
- UCD Conway SPHERE research group, University College Dublin, Dublin, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Martin E Parsons
- UCD Conway SPHERE research group, University College Dublin, Dublin, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Kieran Wynne
- Proteomics Core, Conway Institute, University College Dublin, Dublin, Ireland
| | - Hugh O'Connor
- Department of Haematology, Rotunda Hospital, Dublin, Ireland
| | - Karl Egan
- UCD Conway SPHERE research group, University College Dublin, Dublin, Ireland.,School of Medicine, University College Dublin, Ireland
| | - Seamus Allen
- UCD Conway SPHERE research group, University College Dublin, Dublin, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.,School of Medicine, University College Dublin, Ireland
| | - Fionnuala Ní Áinle
- UCD Conway SPHERE research group, University College Dublin, Dublin, Ireland.,Department of Haematology, Rotunda Hospital, Dublin, Ireland.,School of Medicine, University College Dublin, Ireland.,Departament of Haematology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Patricia B Maguire
- UCD Conway SPHERE research group, University College Dublin, Dublin, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.,UCD Institute for Discovery, O'Brien Centre for Science, University College Dublin, Ireland
| |
Collapse
|
24
|
Chen R, Jin G, Li W, McIntyre TM. Epidermal Growth Factor (EGF) Autocrine Activation of Human Platelets Promotes EGF Receptor-Dependent Oral Squamous Cell Carcinoma Invasion, Migration, and Epithelial Mesenchymal Transition. THE JOURNAL OF IMMUNOLOGY 2018; 201:2154-2164. [PMID: 30150285 DOI: 10.4049/jimmunol.1800124] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 07/25/2018] [Indexed: 12/11/2022]
Abstract
Activated platelets release functional, high m.w. epidermal growth factor (HMW-EGF). In this study, we show platelets also express epidermal growth factor (EGF) receptor (EGFR) protein, but not ErbB2 or ErbB4 coreceptors, and so might respond to HMW-EGF. We found HMW-EGF stimulated platelet EGFR autophosphorylation, PI3 kinase-dependent AKT phosphorylation, and a Ca2+ transient that were blocked by EGFR tyrosine kinase inhibition. Strong (thrombin) and weak (ADP, platelet-activating factor) G protein-coupled receptor agonists and non-G protein-coupled receptor collagen recruited EGFR tyrosine kinase activity that contributed to platelet activation because EGFR kinase inhibition reduced signal transduction and aggregation induced by each agonist. EGF stimulated ex vivo adhesion of platelets to collagen-coated microfluidic channels, whereas systemic EGF injection increased initial platelet deposition in FeCl3-damaged murine carotid arteries. EGFR signaling contributes to oral squamous cell carcinoma (OSCC) tumorigenesis, but the source of its ligand is not established. We find individual platelets were intercalated within OSCC tumors. A portion of these platelets expressed stimulation-dependent Bcl-3 and IL-1β and so had been activated. Stimulated platelets bound OSCC cells, and material released from stimulated platelets induced OSCC epithelial-mesenchymal transition and stimulated their migration and invasion through Matrigel barriers. Anti-EGF Ab or EGFR inhibitors abolished platelet-induced tumor cell phenotype transition, migration, and invasion; so the only factor released from activated platelets necessary for OSCC metastatic activity was HMW-EGF. These results establish HMW-EGF in platelet function and elucidate a previously unsuspected connection between activated platelets and tumorigenesis through rapid, and prolonged, autocrine-stimulated release of HMW-EGF by tumor-associated platelets.
Collapse
Affiliation(s)
- Rui Chen
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Ge Jin
- Case Western Reserve University School of Dental Medicine, Cleveland, OH 44106
| | - Wei Li
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195.,Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106; and.,Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| | - Thomas M McIntyre
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; .,Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106; and.,Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195
| |
Collapse
|
25
|
Parsons MEM, Szklanna PB, Guerrero JA, Wynne K, Dervin F, O'Connell K, Allen S, Egan K, Bennett C, McGuigan C, Gheveart C, Ní Áinle F, Maguire PB. Platelet Releasate Proteome Profiling Reveals a Core Set of Proteins with Low Variance between Healthy Adults. Proteomics 2018; 18:e1800219. [PMID: 29932309 DOI: 10.1002/pmic.201800219] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/09/2018] [Indexed: 12/18/2022]
Abstract
Upon activation, platelets release a powerful cocktail of soluble and vesicular signals, collectively termed the "platelet releasate" (PR). Although several studies have used qualitative/quantitative proteomic approaches to characterize PR; with debated content and significant inter-individual variability reported, confident, and reliable insights have been hindered. Using label-free quantitative (LFQ)-proteomics analysis, a reproducible, quantifiable investigation of the 1U mL-1 thrombin-induced PR from 32 healthy adults was conducted. MS proteomics data are available via ProteomeXchange, identifier PXD009310. Of the 894 proteins identified, 277 proteins were quantified across all donors and form a "core" PR. Bioinformatics and further LFQ-proteomic analysis revealed that the majority (84%) of "core" PR proteins overlapped with the protein composition of human platelet-derived exosomes. Vesicles in the exosomal-size range were confirmed in healthy-human PR and reduced numbers of similar-sized vesicles were observed in the PR of a mouse model of gray platelet syndrome, known to be deficient in platelet alpha-granules. Lastly, the variability of proteins in the PR was assessed, and reproducible secretion levels were found across all 32 healthy donors. Taken together, the PR contains valuable soluble and vesicular cargo and has low-population variance among healthy adults, rendering it a potentially useful platform for diagnostic fingerprinting of platelet-related disease.
Collapse
Affiliation(s)
- Martin E M Parsons
- SPHERE research group, Conway Institute, University College Dublin, Dublin 4, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
| | - Paulina B Szklanna
- SPHERE research group, Conway Institute, University College Dublin, Dublin 4, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
| | - Jose A Guerrero
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Camebridge, United Kingdom.,National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Kieran Wynne
- Proteomics Core, Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Feidhlim Dervin
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland
| | - Karen O'Connell
- Department of Neurology, St Vincent's University Hospital, Dublin 4, Ireland.,School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Seamus Allen
- SPHERE research group, Conway Institute, University College Dublin, Dublin 4, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland.,School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Karl Egan
- SPHERE research group, Conway Institute, University College Dublin, Dublin 4, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Cavan Bennett
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Camebridge, United Kingdom.,National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Christopher McGuigan
- Department of Neurology, St Vincent's University Hospital, Dublin 4, Ireland.,School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Cedric Gheveart
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Camebridge, United Kingdom.,National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Fionnuala Ní Áinle
- SPHERE research group, Conway Institute, University College Dublin, Dublin 4, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,School of Medicine, University College Dublin, Dublin 4, Ireland.,Department of Haematology, Rotunda Hospital, Dublin 1, Ireland.,Department of Haematology, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - Patricia B Maguire
- SPHERE research group, Conway Institute, University College Dublin, Dublin 4, Ireland.,Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland.,UCD Institute for Discovery, O'Brien Centre for Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
26
|
Haemmerle M, Stone RL, Menter DG, Afshar-Kharghan V, Sood AK. The Platelet Lifeline to Cancer: Challenges and Opportunities. Cancer Cell 2018; 33:965-983. [PMID: 29657130 PMCID: PMC5997503 DOI: 10.1016/j.ccell.2018.03.002] [Citation(s) in RCA: 392] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 01/08/2018] [Accepted: 03/01/2018] [Indexed: 12/21/2022]
Abstract
Besides their function in limiting blood loss and promoting wound healing, experimental evidence has highlighted platelets as active players in all steps of tumorigenesis including tumor growth, tumor cell extravasation, and metastasis. Additionally, thrombocytosis in cancer patients is associated with adverse patient survival. Due to the secretion of large amounts of microparticles and exosomes, platelets are well positioned to coordinate both local and distant tumor-host crosstalk. Here, we present a review of recent discoveries in the field of platelet biology and the role of platelets in cancer progression as well as challenges in targeting platelets for cancer treatment.
Collapse
Affiliation(s)
- Monika Haemmerle
- Department of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Institute of Pathology, Martin Luther University Halle-Wittenberg, 06112 Halle, Germany
| | - Rebecca L Stone
- Department of Obstetrics and Gynecology, Johns Hopkins Hospital, Baltimore, MD 21287-1281, USA
| | - David G Menter
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Benign Hematology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Anil K Sood
- Department of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
27
|
Looße C, Swieringa F, Heemskerk JWM, Sickmann A, Lorenz C. Platelet proteomics: from discovery to diagnosis. Expert Rev Proteomics 2018; 15:467-476. [PMID: 29787335 DOI: 10.1080/14789450.2018.1480111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Platelets are the smallest cells within the circulating blood with key roles in physiological hemostasis and pathological thrombosis regulated by the onset of activating/inhibiting processes via receptor responses and signaling cascades. Areas covered: Proteomics as well as genomic approaches have been fundamental in identifying and quantifying potential targets for future diagnostic strategies in the prevention of bleeding and thrombosis, and uncovering the complexity of platelet functions in health and disease. In this article, we provide a critical overview on current functional tests used in diagnostics and the future perspectives for platelet proteomics in clinical applications. Expert commentary: Proteomics represents a valuable tool for the identification of patients with diverse platelet associated defects. In-depth validation of identified biomarkers, e.g. receptors, signaling proteins, post-translational modifications, in large cohorts is decisive for translation into routine clinical diagnostics.
Collapse
Affiliation(s)
- Christina Looße
- a Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund , Germany
| | - Frauke Swieringa
- a Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund , Germany
| | - Johan W M Heemskerk
- b Department of Biochemistry , CARIM, Maastricht University , Maastricht , The Netherlands
| | - Albert Sickmann
- a Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund , Germany.,c Medizinisches Proteom-Center , Medizinische Fakultät, Ruhr-Universität Bochum , Bochum , Germany.,d Department of Chemistry, College of Physical Sciences , University of Aberdeen , Aberdeen , UK
| | - Christin Lorenz
- a Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund , Germany
| |
Collapse
|
28
|
Abstract
Platelet granules are unique among secretory vesicles in both their content and their life cycle. Platelets contain three major granule types—dense granules, α-granules, and lysosomes—although other granule types have been reported. Dense granules and α-granules are the most well-studied and the most physiologically important. Platelet granules are formed in large, multilobulated cells, termed megakaryocytes, prior to transport into platelets. The biogenesis of dense granules and α-granules involves common but also distinct pathways. Both are formed from the
trans-Golgi network and early endosomes and mature in multivesicular bodies, but the formation of dense granules requires trafficking machinery different from that of α-granules. Following formation in the megakaryocyte body, both granule types are transported through and mature in long proplatelet extensions prior to the release of nascent platelets into the bloodstream. Granules remain stored in circulating platelets until platelet activation triggers the exocytosis of their contents. Soluble
N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins, located on both the granules and target membranes, provide the mechanical energy that enables membrane fusion during both granulogenesis and exocytosis. The function of these core fusion engines is controlled by SNARE regulators, which direct the site, timing, and extent to which these SNAREs interact and consequently the resulting membrane fusion. In this review, we assess new developments in the study of platelet granules, from their generation to their exocytosis.
Collapse
Affiliation(s)
- Anish Sharda
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| |
Collapse
|
29
|
Raque VJX, Carlos SGJ, Eduardo RR, Rafael BH, Ángeles RTMDL, Adriana RC, Honorio TA, José BA, Roberto ARS. Modification of immunological features in human platelets during sepsis. Immunol Invest 2017; 47:196-211. [DOI: 10.1080/08820139.2017.1413113] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Valle-Jiménez Xareni Raque
- Department of Experimental Biomedicine, Medicine and Surgery Faculty, Autonomous University ‘‘Benito Juárez’’ of Oaxaca, Oaxaca City, México
- Department of Immunology, National School of Biological Sciences, National Polytechnic Institute, Mexico City, Mexico
| | | | | | - Baltierrez-Hoyos Rafael
- Department of Experimental Biomedicine, Medicine and Surgery Faculty, Autonomous University ‘‘Benito Juárez’’ of Oaxaca, Oaxaca City, México
| | | | - Ramírez-Cosmes Adriana
- Department of Experimental Biomedicine, Medicine and Surgery Faculty, Autonomous University ‘‘Benito Juárez’’ of Oaxaca, Oaxaca City, México
| | - Torres-Aguilar Honorio
- Department of Experimental Biomedicine, Medicine and Surgery Faculty, Autonomous University ‘‘Benito Juárez’’ of Oaxaca, Oaxaca City, México
- Chemical Sciences Faculty, Autonomous University ‘‘Benito Juárez’’, Oaxaca City, Mexico
| | - Bustos-Arriaga José
- Laboratory of molecular biology and immunology of arboviruses, Biomedicine Unit, School of Higher Studies-Iztacala, National Autonomous University of Mexico, State of Mexico, Mexico
| | - Aguilar-Ruiz Sergio Roberto
- Department of Experimental Biomedicine, Medicine and Surgery Faculty, Autonomous University ‘‘Benito Juárez’’ of Oaxaca, Oaxaca City, México
| |
Collapse
|
30
|
Poole LG, Massey VL, Siow DL, Torres-Gonzáles E, Warner NL, Luyendyk JP, Ritzenthaler JD, Roman J, Arteel GE. Plasminogen Activator Inhibitor-1 Is Critical in Alcohol-Enhanced Acute Lung Injury in Mice. Am J Respir Cell Mol Biol 2017; 57:315-323. [PMID: 28445073 PMCID: PMC5625219 DOI: 10.1165/rcmb.2016-0184oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 04/19/2017] [Indexed: 01/07/2023] Open
Abstract
Chronic alcohol exposure is a clinically important risk factor for the development of acute respiratory distress syndrome, the most severe form of acute lung injury (ALI). However, the mechanisms by which alcohol sensitizes the lung to development of this disease are poorly understood. We determined the role of the antifibrinolytic protein plasminogen activator inhibitor-1 (PAI-1) in alcohol enhancement of experimental endotoxin-induced ALI. Wild-type, PAI-1-/-, and integrin β3-/- mice were fed ethanol-containing Lieber-DeCarli liquid or a control diet for 6 weeks, followed by systemic LPS challenge. LPS administration triggered coagulation cascade activation as evidenced by increased plasma thrombin-antithrombin levels and pulmonary fibrin deposition. Ethanol-exposed animals showed enhanced PAI-1 expression and pulmonary fibrin deposition with coincident exaggeration of pulmonary inflammatory edematous injury. PAI-1 deficiency markedly reduced pulmonary fibrin deposition and greatly reduced inflammation and injury without impacting upstream coagulation. Interestingly, pulmonary platelet accumulation was effectively abolished by PAI-1 deficiency in ethanol/LPS-challenged mice. Moreover, mice lacking integrin αIIBβ3, the primary platelet receptor for fibrinogen, displayed a dramatic reduction in early inflammatory changes after ethanol/LPS challenge. These results indicate that the mechanism whereby alcohol exaggerates LPS-induced lung injury requires PAI-1-mediated pulmonary fibrin accumulation, and suggest a novel mechanism whereby alcohol contributes to inflammatory ALI by enhancing fibrinogen-platelet engagement.
Collapse
Affiliation(s)
- Lauren G. Poole
- Department of Pharmacology and Toxicology
- Alcohol Research Center, University of Louisville, Louisville, Kentucky; and
| | - Veronica L. Massey
- Department of Pharmacology and Toxicology
- Alcohol Research Center, University of Louisville, Louisville, Kentucky; and
| | - Deanna L. Siow
- Department of Pharmacology and Toxicology
- Alcohol Research Center, University of Louisville, Louisville, Kentucky; and
| | - Edilson Torres-Gonzáles
- Division of Pulmonary, Critical Care and Sleep Disorders Medicine, Department of Medicine, and
| | - Nikole L. Warner
- Department of Microbiology and Immunology, University of Louisville Health Sciences Center, Louisville, Kentucky
| | - James P. Luyendyk
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Jeffrey D. Ritzenthaler
- Division of Pulmonary, Critical Care and Sleep Disorders Medicine, Department of Medicine, and
| | - Jesse Roman
- Division of Pulmonary, Critical Care and Sleep Disorders Medicine, Department of Medicine, and
| | - Gavin E. Arteel
- Department of Pharmacology and Toxicology
- Alcohol Research Center, University of Louisville, Louisville, Kentucky; and
| |
Collapse
|
31
|
Platelet releasate promotes breast cancer growth and angiogenesis via VEGF-integrin cooperative signalling. Br J Cancer 2017; 117:695-703. [PMID: 28697175 PMCID: PMC5572171 DOI: 10.1038/bjc.2017.214] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 06/13/2017] [Accepted: 06/14/2017] [Indexed: 12/21/2022] Open
Abstract
Background: Selective platelet release of pro- or anti-angiogenic factors distinctly regulated angiogenesis. We hypothesised that selective release of platelet angiogenic factors could differently regulate tumour growth. Methods: Breast cancer cell proliferation, cancer cell-induced endothelial tube formation in vitro, and tumour growth in vivo were studied in the presence of protease-activated receptor 1-stimulated platelet releasate (PAR1-PR; rich in pro-angiogenic factors) or PAR4-PR (rich in anti-angiogenic factors). Results: The PAR1-PR and PAR4-PR supplementation (10%) similarly enhanced cell proliferation of MCF-7 and MDA-MB-231 breast cancer cells. The cancer cells triggered capillary-like tube formation of endothelial cells that was further enhanced by pro-angiogenic factor-rich PAR1-PR. The VEGF, but not SDF-1α, receptor blockade abolished PAR1-PR/PAR4-PR-enhanced cancer cell proliferation. Integrin blockade by RGDS had identical effects as VEGF inhibition. The Src and ERK inhibition diminished, whereas PI3K and PKC blockade abolished platelet releasate-enhanced cancer cell proliferation. Using a model of subcutaneous implantation of MDA-MB-231 cells in nude mice, PAR1-PR enhanced tumour growth more markedly than PAR4-PR, and seemed to achieve the exaggeration by promoting more profound tumour angiogenesis. Conclusions: Platelet releasate increases breast cancer cell proliferation through VEGF–integrin cooperative signalling. Pro-angiogenic factor-rich platelet releasate enhances cancer cell-induced angiogenesis more markedly, and thus exaggerates tumour growth in vivo.
Collapse
|
32
|
Yadav S, Storrie B. The cellular basis of platelet secretion: Emerging structure/function relationships. Platelets 2017; 28:108-118. [PMID: 28010140 PMCID: PMC5627609 DOI: 10.1080/09537104.2016.1257786] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 10/12/2016] [Accepted: 10/27/2016] [Indexed: 12/27/2022]
Abstract
Platelet activation has long been known to be accompanied by secretion from at least three types of compartments. These include dense granules, the major source of small molecules; α-granules, the major protein storage organelle; and lysosomes, the site of acid hydrolase storage. Despite ~60 years of research, there are still many unanswered questions about the cell biology of platelet secretion: for example, how are these secretory organelles organized to support cargo release and what are the key routes of cargo release, granule to plasma membrane or granule to canalicular system. Moreover, in recent years, increasing evidence points to the platelet being organized for secretion of the contents from other organelles, namely the dense tubular system (endoplasmic reticulum) and the Golgi apparatus. Conceivably, protein secretion is a widespread property of the platelet and its organelles. In this review, we concentrate on the cell biology of the α-granule and its structure/function relationships. We both review the literature and discuss the wide array of 3-dimensional, high-resolution structural approaches that have emerged in the last few years. These have begun to reveal new and unanticipated outcomes and some of these are discussed. We are hopeful that the next several years will bring rapid advances to this field that will resolve past controversies and be clinically relevant.
Collapse
Affiliation(s)
- Shilpi Yadav
- a Department of Physiology and Biophysics , University of Arkansas for Medical Sciences , Little Rock , AR , USA
| | - Brian Storrie
- a Department of Physiology and Biophysics , University of Arkansas for Medical Sciences , Little Rock , AR , USA
| |
Collapse
|
33
|
Rampichová M, Buzgo M, Míčková A, Vocetková K, Sovková V, Lukášová V, Filová E, Rustichelli F, Amler E. Platelet-functionalized three-dimensional poly-ε-caprolactone fibrous scaffold prepared using centrifugal spinning for delivery of growth factors. Int J Nanomedicine 2017; 12:347-361. [PMID: 28123295 PMCID: PMC5229261 DOI: 10.2147/ijn.s120206] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Bone and cartilage are tissues of a three-dimensional (3D) nature. Therefore, scaffolds for their regeneration should support cell infiltration and growth in all 3 dimensions. To fulfill such a requirement, the materials should possess large, open pores. Centrifugal spinning is a simple method for producing 3D fibrous scaffolds with large and interconnected pores. However, the process of bone regeneration is rather complex and requires additional stimulation by active molecules. In the current study, we introduced a simple composite scaffold based on platelet adhesion to poly-ε-caprolactone 3D fibers. Platelets were used as a natural source of growth factors and cytokines active in the tissue repair process. By immobilization in the fibrous scaffolds, their bioavailability was prolonged. The biological evaluation of the proposed system in the MG-63 model showed improved metabolic activity, proliferation and alkaline phosphatase activity in comparison to nonfunctionalized fibrous scaffold. In addition, the response of cells was dose dependent with improved biocompatibility with increasing platelet concentration. The results demonstrated the suitability of the system for bone tissue.
Collapse
Affiliation(s)
- Michala Rampichová
- Indoor Environmental Quality, University Center for Energy Efficient Buildings, Czech Technical University in Prague, Buštěhrad; Laboratory of Tissue Engineering, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Matej Buzgo
- Indoor Environmental Quality, University Center for Energy Efficient Buildings, Czech Technical University in Prague, Buštěhrad
| | - Andrea Míčková
- Indoor Environmental Quality, University Center for Energy Efficient Buildings, Czech Technical University in Prague, Buštěhrad; Laboratory of Tissue Engineering, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Karolína Vocetková
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Věra Sovková
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Věra Lukášová
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Eva Filová
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Franco Rustichelli
- Laboratory of Tissue Engineering, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Evžen Amler
- Indoor Environmental Quality, University Center for Energy Efficient Buildings, Czech Technical University in Prague, Buštěhrad; Laboratory of Tissue Engineering, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
34
|
Pagel O, Walter E, Jurk K, Zahedi RP. Taking the stock of granule cargo: Platelet releasate proteomics. Platelets 2016; 28:119-128. [PMID: 27928935 DOI: 10.1080/09537104.2016.1254762] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human platelets are key players in a multitude of physiological and pathological processes. Upon activation they release cargo from different types of granules as well as microparticles in an apparently well-regulated and orchestrated manner. The resulting specific platelet releasates create microenvironments of biologically active compounds and proteins during platelet aggregation and thrombus formation, allowing efficient delivery of growth factors and immune modulators to their sites of effect and enhancing the coagulative response in a positive feedback loop. Thus, platelet releasates play a central role in the regulation of platelet homeostasis and heterotypic cell interaction. Additionally, it recently emerged that both the qualitative and quantitative composition of the releasate as well as release dynamics may be stimulus dependent and therefore more complex than expected. Mass spectrometry-based proteomics is an important asset for studying platelet releasates in vitro, as it allows not only (i) identifying released proteins, but moreover (ii) determining their quantities and the dynamics of release as well as (iii) differentially comparing releasates across a variety of conditions. Though owing to the high sensitivity and comprehensiveness of modern proteomic techniques, a thorough experimental design and a standardized and robust sample preparation are essential to obtain highly confident and reliable insights into platelet biology and pathology. Here, we review releasate proteome studies and crucial sample preparation strategies to summarize possible achievements of state-of-the-art technologies and furthermore discuss potential pitfalls and limitations. We provide a future perspective of platelet releasate proteomics including targeted analyses, post-translational modifications and multi-omics approaches that should be adopted by platelet releasate researchers due to their tremendous depth and comprehensiveness.
Collapse
Affiliation(s)
- Oliver Pagel
- a Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V ., Dortmund , Germany
| | - Elena Walter
- b Center for Thrombosis and Hemostasis (CTH) , Universitätsklinikum der Johannes Gutenberg-Universität Mainz , Mainz , Germany
| | - Kerstin Jurk
- b Center for Thrombosis and Hemostasis (CTH) , Universitätsklinikum der Johannes Gutenberg-Universität Mainz , Mainz , Germany
| | - René P Zahedi
- a Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V ., Dortmund , Germany
| |
Collapse
|
35
|
Leunissen TC, Wisman PP, van Holten TC, de Groot PG, Korporaal SJ, Koekman AC, Moll FL, Teraa M, Verhaar MC, de Borst GJ, Urbanus RT, Roest M. The effect of P2Y12 inhibition on platelet activation assessed with aggregation- and flow cytometry-based assays. Platelets 2016; 28:567-575. [PMID: 27885904 DOI: 10.1080/09537104.2016.1246713] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Patients on P2Y12 inhibitors may still develop thrombosis or bleeding complications. Tailored antiplatelet therapy, based on platelet reactivity testing, might reduce these complications. Several tests have been used, but failed to show a benefit of tailored antiplatelet therapy. This could be due to the narrowness of current platelet reactivity tests, which are limited to analysis of platelet aggregation after stimulation of the adenosine diphosphate (ADP)-pathway. However, the response to ADP does not necessarily reflect the effect of P2Y12 inhibition on platelet function in vivo. Therefore, we investigated whether measuring platelet reactivity toward other physiologically relevant agonists could provide more insight in the efficacy of P2Y12 inhibitors. The effect of in vitro and in vivo P2Y12 inhibition on αIIbβ3-activation, P-selectin and CD63-expression, aggregate formation, release of alpha, and dense granules content was assessed after stimulation of different platelet activation pathways. Platelet reactivity measured with flow cytometry in 72 patients on P2Y12 inhibitors was compared to VerifyNow results. P2Y12 inhibitors caused strongly attenuated platelet fibrinogen binding after stimulation with peptide agonists for protease activated receptor (PAR)-1 and -4, or glycoprotein VI ligand crosslinked collagen-related peptide (CRP-xl), while aggregation was normal at high agonist concentration. P2Y12 inhibitors decreased PAR-agonist and CRP-induced dense granule secretion, but not alpha granule secretion. A proportion of P2Y12-inhibitor responsive patients according to VerifyNow, displayed normal fibrinogen binding assessed with flow cytometry after stimulation with PAR-agonists or CRP despite full inhibition of the response to ADP, indicating suboptimal platelet inhibition. Concluding, measurement of platelet fibrinogen binding with flow cytometry after stimulation of thrombin- or collagen receptors in addition to ADP response identifies different patients as nonresponders to P2Y12 inhibitors, compared to only ADP-induced aggregation-based assays. Future studies should investigate the value of both assays for monitoring on-treatment platelet reactivity.
Collapse
Affiliation(s)
- Tesse C Leunissen
- a Department of Clinical Chemistry and Hematology , University Medical Center Utrecht , The Netherlands.,b Department of Vascular Surgery , University Medical Center Utrecht , The Netherlands
| | - Peter Paul Wisman
- a Department of Clinical Chemistry and Hematology , University Medical Center Utrecht , The Netherlands.,b Department of Vascular Surgery , University Medical Center Utrecht , The Netherlands
| | - Thijs C van Holten
- a Department of Clinical Chemistry and Hematology , University Medical Center Utrecht , The Netherlands
| | - Philip G de Groot
- a Department of Clinical Chemistry and Hematology , University Medical Center Utrecht , The Netherlands
| | - Suzanne J Korporaal
- a Department of Clinical Chemistry and Hematology , University Medical Center Utrecht , The Netherlands
| | - Arnold C Koekman
- a Department of Clinical Chemistry and Hematology , University Medical Center Utrecht , The Netherlands
| | - Frans L Moll
- b Department of Vascular Surgery , University Medical Center Utrecht , The Netherlands
| | - Martin Teraa
- b Department of Vascular Surgery , University Medical Center Utrecht , The Netherlands.,c Department of Nephrology and Hypertension , University Medical Center Utrecht , The Netherlands
| | - Marianne C Verhaar
- c Department of Nephrology and Hypertension , University Medical Center Utrecht , The Netherlands
| | - Gert Jan de Borst
- b Department of Vascular Surgery , University Medical Center Utrecht , The Netherlands
| | - Rolf T Urbanus
- a Department of Clinical Chemistry and Hematology , University Medical Center Utrecht , The Netherlands
| | - Mark Roest
- a Department of Clinical Chemistry and Hematology , University Medical Center Utrecht , The Netherlands
| |
Collapse
|
36
|
Abstract
Secretion is essential to many of the roles that platelets play in the vasculature, e.g., thrombosis, angiogenesis, and inflammation, enabling platelets to modulate the microenvironment at sites of vascular lesions with a myriad of bioactive molecules stored in their granules. Past studies demonstrate that granule cargo release is mediated by Soluble NSF Attachment Protein Receptor (SNARE) proteins, which are required for granule-plasma membrane fusion. Several SNARE regulators, which control when, where, and how the SNAREs interact, have been identified in platelets. Additionally, platelet SNAREs are controlled by post-translational modifications, e.g., phosphorylation and acylation. Although there have been many recent insights into the mechanisms of platelet secretion, many questions remain: have we identified all the important regulators, does calcium directly control the process, and is platelet secretion polarized. In this review, we focus on the mechanics of platelet secretion and discuss how the secretory machinery functions in the pathway leading to membrane fusion and cargo release.
Collapse
Affiliation(s)
- Smita Joshi
- a Department of Molecular and Cellular Biochemistry , University of Kentucky , Lexington , KY , USA
| | - Sidney W Whiteheart
- a Department of Molecular and Cellular Biochemistry , University of Kentucky , Lexington , KY , USA
| |
Collapse
|
37
|
Izquierdo I, García Á. Platelet proteomics applied to the search for novel antiplatelet therapeutic targets. Expert Rev Proteomics 2016; 13:993-1006. [DOI: 10.1080/14789450.2016.1246188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
38
|
Respective contributions of single and compound granule fusion to secretion by activated platelets. Blood 2016; 128:2538-2549. [PMID: 27625359 DOI: 10.1182/blood-2016-03-705681] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 09/09/2016] [Indexed: 12/25/2022] Open
Abstract
Although granule secretion is pivotal in many platelet responses, the fusion routes of α and δ granule release remain uncertain. We used a 3D reconstruction approach based on electron microscopy to visualize the spatial organization of granules in unstimulated and activated platelets. Two modes of exocytosis were identified: a single mode that leads to release of the contents of individual granules and a compound mode that leads to the formation of granule-to-granule fusion, resulting in the formation of large multigranular compartments. Both modes occur during the course of platelet secretion. Single fusion events are more visible at lower levels of stimulation and early time points, whereas large multigranular compartments are present at higher levels of agonist and at later time points. Although α granules released their contents through both modes of exocytosis, δ granules underwent only single exocytosis. To define the underlying molecular mechanisms, we examined platelets from vesicle-associated membrane protein 8 (VAMP8) null mice. After weak stimulation, compound exocytosis was abolished and single exocytosis decreased in VAMP8 null platelets. Higher concentrations of thrombin bypassed the VAMP8 requirement, indicating that this isoform is a key but not a required factor for single and/or compound exocytosis. Concerning the biological relevance of our findings, compound exocytosis was observed in thrombi formed after severe laser injury of the vessel wall with thrombin generation. After superficial injury without thrombin generation, no multigranular compartments were detected. Our studies suggest that platelets use both modes of membrane fusion to control the extent of agonist-induced exocytosis.
Collapse
|
39
|
Miao X, Zhang W, Huang Z, Li N. Unaltered Angiogenesis-Regulating Activities of Platelets in Mild Type 2 Diabetes Mellitus despite a Marked Platelet Hyperreactivity. PLoS One 2016; 11:e0162405. [PMID: 27612088 PMCID: PMC5017716 DOI: 10.1371/journal.pone.0162405] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 08/22/2016] [Indexed: 12/23/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with platelet dysfunction and impaired angiogenesis. Aim of the study is to investigate if platelet dysfunction might hamper platelet angiogenic activities in T2DM patients. Sixteen T2DM patients and gender/age-matched non-diabetic controls were studied. Flow cytometry and endothelial colony forming cell (ECFC) tube formation on matrigel were used to assess platelet reactivity and angiogenic activity, respectively. Thrombin receptor PAR1-activating peptide (PAR1-AP) induced higher platelet P-selectin expression, and evoked more rapid and intense platelet annexin V binding in T2DM patients, seen as a more rapid increase of annexin V+ platelets (24.3±6.4% vs 12.6±3.8% in control at 2 min) and a higher elevation (30.9±5.1% vs 24.3±3.0% at 8 min). However, PAR1-AP and PAR4-AP induced similar releases of angiogenic regulators from platelets, and both stimuli evoked platelet release of platelet angiogenic regulators to similar extents in T2DM and control subjects. Thus, PAR1-stimulated platelet releasate (PAR1-PR) and PAR4-PR similarly enhanced capillary-like network/tube formation of ECFCs, and the enhancements did not differ between T2DM and control subjects. Direct supplementation of platelets to ECFCs at the ratio of 1:200 enhanced ECFC tube formation even more markedly, leading to approximately 100% increases of the total branch points of ECFC tube formation, for which the enhancements were also similar between patients and controls. In conclusion, platelets from T2DM subjects are hyperreactive. Platelet activation induced by high doses of PAR1-AP, however, results in similar releases of angiogenic regulators in mild T2DM and control subjects. Platelets from T2DM and control subjects also demonstrate similar enhancements on ECFC angiogenic activities.
Collapse
Affiliation(s)
- Xinyan Miao
- Karolinska Institutet, Department of Medicine-Solna, Clinical Pharmacology, Karolinska University Hospital-Solna, 171 76, Stockholm, Sweden
| | - Wei Zhang
- Karolinska Institutet, Department of Medicine-Solna, Clinical Pharmacology, Karolinska University Hospital-Solna, 171 76, Stockholm, Sweden
| | - Zhangsen Huang
- Karolinska Institutet, Department of Medicine-Solna, Clinical Pharmacology, Karolinska University Hospital-Solna, 171 76, Stockholm, Sweden
| | - Nailin Li
- Karolinska Institutet, Department of Medicine-Solna, Clinical Pharmacology, Karolinska University Hospital-Solna, 171 76, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
40
|
CCL5 derived from platelets increases megakaryocyte proplatelet formation. Blood 2015; 127:921-6. [PMID: 26647394 DOI: 10.1182/blood-2015-05-644583] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 11/25/2015] [Indexed: 02/07/2023] Open
Abstract
In times of physiological stress, platelet count can transiently rise. What initiates this reactive thrombocytosis is poorly understood. Intriguingly, we found that treating megakaryocytes (MKs) with the releasate from activated platelets increased proplatelet production by 47%. Platelets store inflammatory cytokines, including the chemokine ligand 5 (CCL5, RANTES); after TRAP activation, platelets release over 25 ng/mL CCL5. We hypothesized that CCL5 could regulate platelet production by binding to its receptor, CCR5, on MKs. Maraviroc (CCR5 antagonist) or CCL5 immunodepletion diminished 95% and 70% of the effect of platelet releasate, respectively, suggesting CCL5 derived from platelets is sufficient to drive increased platelet production through MK CCR5. MKs cultured with recombinant CCL5 increased proplatelet production by 50% and had significantly higher ploidy. Pretreating the MK cultures with maraviroc prior to exposure to CCL5 reversed the augmented proplatelet formation and ploidy, suggesting that CCL5 increases MK ploidy and proplatelet formation in a CCR5-dependent manner. Interrogation of the Akt signaling pathway suggested that CCL5/CCR5 may influence proplatelet production by suppressing apoptosis. In an in vivo murine acute colitis model, platelet count significantly correlated with inflammation whereas maraviroc treatment abolished this correlation. We propose that CCL5 signaling through CCR5 may increase platelet counts during physiological stress.
Collapse
|
41
|
Heijnen H, van der Sluijs P. Platelet secretory behaviour: as diverse as the granules … or not? J Thromb Haemost 2015; 13:2141-51. [PMID: 26391322 DOI: 10.1111/jth.13147] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 09/05/2015] [Indexed: 12/19/2022]
Abstract
Platelets play a central role in the arrest of bleeding after damage to a blood vessel and in the development of thrombosis. Platelets rapidly respond after interaction with sub-endothelial components and release cargo from their storage granules. The three principal granule types of platelets are α-granules, dense granules and lysosomes. Timed release of granule contents and regulated expression of critical receptors are essential for maintenance of the platelet thrombus, yet also have important functions beyond hemostasis (i.e. inflammatory reactions and immune responses). α-granules store adhesive molecules such as von Willebrand factor and fibrinogen, growth factors and inflammatory and angiogenic mediators, which play crucial roles in inflammatory responses and tumor genesis. The α-granules comprise a group of subcellular compartments with a unique composition and ultrastructure. Recent studies have suggested that differential secretory kinetics of α-granule subtypes is responsible for a thematic release of adhesive and inflammatory mediators. In addition, new results indicate that activation-dependent synthesis and release of cytokines also contribute to the inflammatory role of platelets. We will discuss the various methods that platelets use to regulate secretory processes and how these relate to potential differential secretion patterns, thereby promoting adhesiveness and/or inflammatory functions. We will focus on the heterogenic granule population, open canalicular system (OCS) plasticity, the role of contractile and mechanobiological forces, and the fusogenic machinery.
Collapse
Affiliation(s)
- H Heijnen
- Department of Clinical Chemistry and Hematology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - P van der Sluijs
- Department of Cell Biology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
42
|
|
43
|
Li N. Platelets in cancer metastasis: To help the "villain" to do evil. Int J Cancer 2015; 138:2078-87. [PMID: 26356352 DOI: 10.1002/ijc.29847] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 07/27/2015] [Accepted: 08/24/2015] [Indexed: 12/16/2022]
Abstract
Cancer progress is accompanied by platelet activation and thrombotic complications. Platelets are a dangerous alliance of cancer cells, and are a close engager in multiple processes of cancer metastasis. Platelet adhesion to cancer cells forms a protective cloak that helps cancer cells to escape immune surveillance and natural killer cell-mediated cytolysis. Platelets facilitate tethering and arrest of disseminated cancer cells in the vasculature, enhance invasive potentials and thus extravasation of cancer cells. Moreover, platelets recruit monocytes and granulocytes to the sites of cancer cell arrest, and collaborate with them to establish a pro-metastatic microenvironment and metastatic niches. Platelets also secret a number of growth factors to stimulate cancer cell proliferation, release various angiogenic regulators to regulate tumor angiogenesis and subsequently promote cancer growth and progress. Albeit platelets are helping the "villain" cancer to do evil, the close engagements of platelets in cancer metastasis and progress can be used as the intervention targets for new anti-cancer therapeutic developments. Platelet-targeted anti-cancer strategy may bring in novel anti-cancer treatments that can synergize the therapeutic effects of chemotherapies and surgical treatments of cancer.
Collapse
Affiliation(s)
- Nailin Li
- Karolinska Institutet Department of Medicine-Solna, Clinical Pharmacology Group, Karolinska University Hospital-Solna, 171 76, Stockholm, Sweden
| |
Collapse
|
44
|
Nguyen KA, Hamzeh-Cognasse H, Laradi S, Pozzetto B, Garraud O, Cognasse F. Specific activation, signalling and secretion profiles of human platelets following PAR-1 and PAR-4 stimulation. Platelets 2015; 26:795-8. [PMID: 26083907 DOI: 10.3109/09537104.2015.1050585] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Blood platelets play a central haemostatic function; however, they also play a role in inflammation and are capable of secreting various cytokines, chemokines and related products. The purpose of this study was to identify subtle variations in platelet physiology using proteomics. We compared the levels of membrane proteins (n = 3), α and δ granule proteins (n = 18), and signalling proteins (n = 30) from unstimulated platelets with those of protease-activated receptor (PAR)-1- and PAR-4-stimulated platelets (n = 10). The vast majority of these proteins responded similarly to PAR-1 or PAR-4 engagement. However, differences were observed within membrane CD40L expressed, and α granule GRO-α and MDC secreted proteins.
Collapse
Affiliation(s)
| | | | - Sandrine Laradi
- a Université de Lyon , Saint Etienne , France .,b Etablissement Français du Sang - Auvergne-Loire , Saint-Etienne , France , and
| | | | - Olivier Garraud
- a Université de Lyon , Saint Etienne , France .,c INTS - Institut National de la Transfusion Sanguine , Paris , France
| | - Fabrice Cognasse
- a Université de Lyon , Saint Etienne , France .,b Etablissement Français du Sang - Auvergne-Loire , Saint-Etienne , France , and
| |
Collapse
|
45
|
Etulain J, Mena HA, Negrotto S, Schattner M. Stimulation of PAR-1 or PAR-4 promotes similar pattern of VEGF and endostatin release and pro-angiogenic responses mediated by human platelets. Platelets 2015; 26:799-804. [PMID: 26082997 DOI: 10.3109/09537104.2015.1051953] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Platelets mediate angiogenesis through the secretion of several factors, including the pro-angiogenic vascular endothelial growth factor (VEGF) and the anti-angiogenic endostatin. Although previous findings indicated that these molecules are packed into different alpha-granules and selectively released by specific stimulation of protease-activated receptor (PAR)-1 or PAR-4, recent evidences are against this hypothesis. OBJECTIVES To elucidate the controversies about the VEGF and endostatin release and the overall angiogenic effect of PARs-stimulated platelets. METHODS VEGF and endostatin were quantified by enzyme linked-immunosorbent assay (ELISA). Endothelial proliferation (pNPP assay), wound healing (scratch assay) and tubule formation (matrigel) of human microvascular endothelial cells (HMEC-1) and endothelial progenitor cells (EPC) were determined using supernatants from PAR-1- or PAR-4-stimulated platelets. RESULTS Activation of washed platelets (WPs) by PAR-1- or PAR-4-activating peptide (AP) promoted the VEGF and endostatin secretion in a concentration-dependent manner, being PAR-1-AP more potent than PAR-4-AP. The release of both molecules was abrogated by pre-incubation of platelets with PAR antagonists. Activation of platelet-rich plasma (PRP) with either PAR-1-AP or PAR-4-AP induced a significant VEGF secretion. Quantification of platelet-endostatin secretion was not possible in PRP due to the high levels of plasmatic endostatin vs. platelet content. Releasates from PAR-1- or PAR-4-activated WPs promoted similar pattern of angiogenic responses of HMEC-1 or EPC. Moreover, proliferation of HMEC-1 mediated by PAR-stimulated PRP releasates was delayed and significantly lower compared with that induced by PAR-stimulated WPs. CONCLUSIONS Our results are in contrast with the previously described differential release of VEGF and endostatin induced by the selective PAR-1 or PAR-4 stimulation, and support the notion that while circulating endostatin accounts for the maintenance of a systemic anti-angiogenic state, locally, the release of platelet alpha-granule content promotes angiogenesis.
Collapse
Affiliation(s)
- J Etulain
- a Laboratory of Experimental Thrombosis , Institute of Experimental Medicine, CONICET-National Academy of Medicine , Buenos Aires , Argentina
| | - H A Mena
- a Laboratory of Experimental Thrombosis , Institute of Experimental Medicine, CONICET-National Academy of Medicine , Buenos Aires , Argentina
| | - S Negrotto
- a Laboratory of Experimental Thrombosis , Institute of Experimental Medicine, CONICET-National Academy of Medicine , Buenos Aires , Argentina
| | - M Schattner
- a Laboratory of Experimental Thrombosis , Institute of Experimental Medicine, CONICET-National Academy of Medicine , Buenos Aires , Argentina
| |
Collapse
|
46
|
Marcone S, Dervin F, Fitzgerald DJ. Proteomic signatures of antiplatelet drugs: new approaches to exploring drug effects. J Thromb Haemost 2015; 13 Suppl 1:S323-31. [PMID: 26149042 DOI: 10.1111/jth.12943] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Antiplatelet agents represent the mainstay of acute coronary syndrome (ACS) therapy to prevent ischemic events and to improve safety in patients undergoing percutaneous coronary intervention. However, despite the availability of several drugs and the use of dual antiplatelet therapy, the pharmacological response is highly variable with a subset of patients continuing to experience recurrent thrombotic events, revealing a wide variability in platelet response to antiplatelet drugs. Several factors may explain this, including genetic variation and environmental factors. Here we look at the application of proteomic analysis, an approach that provides an integrated readout of these diverse influences.
Collapse
Affiliation(s)
- S Marcone
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - F Dervin
- School of Biomedical and Biomolecular Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - D J Fitzgerald
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
47
|
Dzieciatkowska M, D'Alessandro A, Hill RC, Hansen KC. Plasma QconCATs reveal a gender-specific proteomic signature in apheresis platelet plasma supernatants. J Proteomics 2015; 120:1-6. [PMID: 25743772 DOI: 10.1016/j.jprot.2015.02.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 02/20/2015] [Accepted: 02/21/2015] [Indexed: 11/29/2022]
Abstract
UNLABELLED Clinical translation of proteomic technologies is often hampered by technical limitations, including inter-laboratory inconsistencies of label-free derived relative quantification, time-consuming analytical approaches and the subsequent challenge of performing proteomic analyses on large cohorts of subjects. Here we introduce plasma QconCAT-based targeted proteomics, an approach that allows the simultaneous absolute quantitation down to the picogram level of hundreds of proteins in a single liquid chromatography-selected reaction monitoring mass spectrometry run. We demonstrate the robustness of the approach by analyzing apheresis platelet concentrate supernatants at storage day 1 and the end of the shelf life for this blood-derived therapeutic, day 5. The targeted approach was repeatable and robust revealing potential gender-specific signatures across a set of three male and female donors. This technical note represents a proof-of-principle of the application of QconCAT-based MRM strategies to transfusion-medicine relevant issues, such as storage and gender-dependent proteomic signatures in blood-derived therapeutics. BIOLOGICAL SIGNIFICANCE Gender differences in the proteome composition of apheresis platelet supernatants have always been postulated, and might underlie a higher risk of adverse reactions when transfusing apheresis products from female donors. Preliminary proteomic studies provided an overview of gender-dependent relative compositional differences in the proteome of apheresis platelet supernatants during routine storage in the blood bank. Here we apply a proteomics approach for absolute quantitation of approximately 100 proteins in apheresis platelet supernatants from male and female donors at storage days 1 and 5. Absolute quantitative proteomic analyses allowed us to confirm and expand on previous observations about gender and storage-dependency of platelet supernatant protein profiles.
Collapse
Affiliation(s)
- Monika Dzieciatkowska
- University of Colorado Denver School of Medicine, Biochemistry and Molecular Genetics, School of Medicine University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Angelo D'Alessandro
- University of Colorado Denver School of Medicine, Biochemistry and Molecular Genetics, School of Medicine University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Ryan C Hill
- University of Colorado Denver School of Medicine, Biochemistry and Molecular Genetics, School of Medicine University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Kirk C Hansen
- University of Colorado Denver School of Medicine, Biochemistry and Molecular Genetics, School of Medicine University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
48
|
Cognasse F, Nguyen KA, Damien P, McNicol A, Pozzetto B, Hamzeh-Cognasse H, Garraud O. The Inflammatory Role of Platelets via Their TLRs and Siglec Receptors. Front Immunol 2015; 6:83. [PMID: 25784910 PMCID: PMC4345914 DOI: 10.3389/fimmu.2015.00083] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/12/2015] [Indexed: 12/16/2022] Open
Abstract
Platelets are non-nucleated cells that play central roles in the processes of hemostasis, innate immunity, and inflammation; however, several reports show that these distinct functions are more closely linked than initially thought. Platelets express numerous receptors and contain hundreds of secretory products. These receptors and secretory products are instrumental to the platelet functional responses. The capacity of platelets to secrete copious amounts of cytokines, chemokines, and related molecules appears intimately related to the role of the platelet in inflammation. Platelets exhibit non-self-infectious danger detection molecules on their surfaces, including those belonging to the “toll-like receptor” family, as well as pathogen sensors of other natures (Ig- or complement receptors, etc.). These receptors permit platelets to both bind infectious agents and deliver differential signals leading to the secretion of cytokines/chemokines, under the control of specific intracellular regulatory pathways. In contrast, dysfunctional receptors or dysregulation of the intracellular pathway may increase the susceptibility to pathological inflammation. Physiological vs. pathological inflammation is tightly controlled by the sensors of danger expressed in resting, as well as in activated, platelets. These sensors, referred to as pathogen recognition receptors, primarily sense danger signals termed pathogen associated molecular patterns. As platelets are found in inflamed tissues and are involved in auto-immune disorders, it is possible that they can also be stimulated by internal pathogens. In such cases, platelets can also sense danger signals using damage associated molecular patterns (DAMPs). Some of the most significant DAMP family members are the alarmins, to which the Siglec family of molecules belongs. This review examines the role of platelets in anti-infection immunity via their TLRs and Siglec receptors.
Collapse
Affiliation(s)
- Fabrice Cognasse
- Etablissement Français du Sang Auvergne-Loire , Saint-Etienne , France ; GIMAP-EA3064, Université de Lyon , Saint Etienne , France
| | - Kim Anh Nguyen
- GIMAP-EA3064, Université de Lyon , Saint Etienne , France
| | - Pauline Damien
- GIMAP-EA3064, Université de Lyon , Saint Etienne , France
| | - Archibald McNicol
- Faculty of Health Sciences, Colleges of Pharmacy and Medicine, University of Manitoba , Winnipeg, MB , Canada
| | - Bruno Pozzetto
- GIMAP-EA3064, Université de Lyon , Saint Etienne , France
| | | | - Olivier Garraud
- GIMAP-EA3064, Université de Lyon , Saint Etienne , France ; Institut National de Transfusion Sanguine (INTS) , Paris , France
| |
Collapse
|
49
|
Huang Z, Miao X, Luan Y, Zhu L, Kong F, Lu Q, Pernow J, Nilsson G, Li N. PAR1-stimulated platelet releasate promotes angiogenic activities of endothelial progenitor cells more potently than PAR4-stimulated platelet releasate. J Thromb Haemost 2015; 13:465-76. [PMID: 25495701 DOI: 10.1111/jth.12815] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 12/07/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) are important for endothelial regeneration and angiogenesis. Thrombin protease-activated receptor 1 (PAR1) PAR1 and PAR4 stimulation induces selective release of platelet proangiogenic and antiangiogenic regulators. OBJECTIVE To investigate if PAR1-stimulated platelet releasate (PAR1-PR) and PAR4-PR regulate angiogenic properties of EPCs in different manners. METHODS AND RESULTS EPCs were generated from peripheral mononuclear cell culture. Washed platelets (2 × 10(9) mL(-1)) were stimulated by PAR1-activating peptide (PAR1-AP; 10 μmol L(-1)) or PAR4-AP (100 μmol L(-1)) to prepare PAR1-PR and PAR4-PR, respectively. PAR1-PR or PAR4-PR had little influence on EPC proliferation. EPC migration experiments using a modified Boyden chamber showed that both platelet releasates facilitated EPC migration. As for in vitro tube formation on Matrigel, PAR1-PR and PAR4-PR similarly enhanced capillary-like network formation of EPCs in the complete EPC medium containing 10% FBS and a cocktail of growth factors, while PAR1-PR more profoundly increased EPC tube formation in basal culture medium supplemented with only 0.5% FBS than did PAR4-PR. The latter was confirmed in the murine angiogenesis model of subcutaneous Matrigel implantation. Moreover, blockade of vascular endothelial growth factor, stromal cell-derived factor 1α, or matrix metalloproteinases attenuated EPC migration and tube formation, suggesting a cooperation of these factors in the enhancements. CONCLUSIONS PAR1-PR enhances vasculogenesis more potently than PAR4-PR, and the enhancements require a cooperation of multiple platelet-derived angiogenic regulators.
Collapse
Affiliation(s)
- Z Huang
- Clinical Pharmacology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital-Solna, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Platelet hyaluronidase-2: an enzyme that translocates to the surface upon activation to function in extracellular matrix degradation. Blood 2014; 125:1460-9. [PMID: 25411425 DOI: 10.1182/blood-2014-07-590513] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Following injury, platelets rapidly interact with the exposed extracellular matrix (ECM) of the vessel wall and the surrounding tissues. Hyaluronan (HA) is a major glycosaminoglycan component of the ECM and plays a significant role in regulating inflammation. We have recently reported that human platelets degrade HA from the surfaces of activated endothelial cells into fragments capable of inducing immune responses by monocytes. We also showed that human platelets contain the enzyme hyaluronidase-2 (HYAL2), one of two major hyaluronidases that digest HA in somatic tissues. The deposition of HA increases in inflamed tissues in several inflammatory diseases, including inflammatory bowel disease (IBD). We therefore wanted to define the mechanism by which platelets degrade HA in the inflamed tissues. In this study, we show that human platelets degrade the proinflammatory matrix HA through the activity of HYAL2 and that platelet activation causes the immediate translocation of HYAL2 from a distinct population of α-granules to platelet surfaces where it exerts its catalytic activity. Finally, we show that patients with IBD have lower platelet HYAL2 levels and activity than healthy controls.
Collapse
|