1
|
Sennett C, Pula G. Trapped in the NETs: Multiple Roles of Platelets in the Vascular Complications Associated with Neutrophil Extracellular Traps. Cells 2025; 14:335. [PMID: 40072064 PMCID: PMC11898727 DOI: 10.3390/cells14050335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
Neutrophil extracellular traps (NETs) have received significant attention in recent years for their role in both the immune response and the vascular damage associated with inflammation. Platelets have been described as critical components of NETs since the initial description of this physio-pathological response of neutrophils. Platelets have been shown to play a dual role as responders and also as stimulators of NETs. The direct interaction with DNA leads to the entrapment of platelets into NETs, a phenomenon that significantly contributes to the thrombotic complications of inflammation and neutrophil activation, while the direct and paracrine stimulation of neutrophils by platelets has been shown to initiate the process of NET formation. In this review, we provide a comprehensive description of our current understanding of the molecular mechanisms underlying the entrapping of platelets into NETs and, in parallel, the platelet-driven cellular responses promoting NET formation. We then illustrate established examples of the contribution of NETs to vascular pathologies, describe the important questions that remain to be answered regarding the contribution of platelets to NET formation and NET-dependent cardiovascular complication, and highlight the fundamental steps taken towards the application of our understanding of platelets' contribution to NETs for the development of novel cardiovascular therapies.
Collapse
Affiliation(s)
| | - Giordano Pula
- Biomedical Institute for Multimorbidity (BIM), Hull York Medical School (HYMS), University of Hull, Hull HU6 7RX, UK
| |
Collapse
|
2
|
Martínez-Botía P, Tassi Yunga S, Szklanna P, Babur O, Emili A, Wilmarth PA, Heemskerk JWM, Maguire PB, Iding AFJ, Ramström S, García Á, Aslan JE, Gutiérrez L. Toward standardization and a concerted vision for platelet proteomics research: communication from the SSC of the ISTH. J Thromb Haemost 2025:S1538-7836(25)00064-9. [PMID: 39952361 DOI: 10.1016/j.jtha.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/23/2024] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Over the past 3 decades, omics technologies have revolutionized our understanding of platelet molecular content and organization, enabling the systematic analyses of platelet physiology. Among these approaches, proteomics has been especially significant in discovering as well as validating molecular mechanisms of platelet function in health and disease. However, several conceptual and practical challenges continue to limit the full utility of platelet proteomics tools and data. Methodological and analytical inconsistencies remain a key concern, with biological and technical variables exerting substantial influence on study outcomes and interpretation. These issues are compounded by the rapid pace of proteomics tool development and dataset collection, outstripping efforts to standardize best practices and ensure consensus, as platelet proteomics consolidates itself as a tool for research even outside the thrombosis and hemostasis field. In this communication from the International Society on Thrombosis and Haemostasis Scientific and Standardization Committee, we highlight recent advances in platelet proteomics studies, and we identify where collective efforts can strengthen experimental design, execution, and analysis. As a practical recommendation, we encourage platelet biologists to recognize current discrepancies and advance efforts to standardize and customize methods and reporting practices, including blood collection, platelet isolation, data acquisition, and data interpretation. By aligning protocols and ensuring detailed reporting, the field can more effectively integrate proteomics findings and accelerate our understanding of platelet biology.
Collapse
Affiliation(s)
- Patricia Martínez-Botía
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Samuel Tassi Yunga
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA; Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Paulina Szklanna
- UCD Conway SPHERE research group, UCD Institute for Discovery, University College Dublin (Ireland)
| | - Ozgun Babur
- Department of Computer Science, University of Massachusetts, Boston, Boston, Massachusetts, USA
| | - Andrew Emili
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA; Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Phillip A Wilmarth
- Proteomics Shared Resource, Oregon Health & Science University, Portland, Oregon, USA
| | | | - Patricia B Maguire
- UCD Conway SPHERE research group, UCD Institute for Discovery, University College Dublin (Ireland)
| | - Aaron F J Iding
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Sofia Ramström
- Cardiovascular Research Centre, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ángel García
- Platelet Proteomics Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, and Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Joseph E Aslan
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA; Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA; Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Laura Gutiérrez
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Department of Medicine, University of Oviedo, Oviedo, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.
| |
Collapse
|
3
|
Bravaccini S, Boldrin E, Gurioli G, Tedaldi G, Piano MA, Canale M, Curtarello M, Ulivi P, Pilati P. The use of platelets as a clinical tool in oncology: opportunities and challenges. Cancer Lett 2024; 607:217044. [PMID: 38876385 DOI: 10.1016/j.canlet.2024.217044] [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: 02/16/2024] [Revised: 05/17/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024]
Abstract
Platelets are small circulating anucleated cells mainly involved in thrombosis and hemostasis processes. Moreover, platelets play an active role in tumorigenesis and cancer progression, stimulating angiogenesis and vascular remodelling, and protecting circulating cancer cells from shear forces and immune surveillance. Several reports indicate that platelet number in the blood circulation of cancer patients is associated with prognosis and response to treatment. However, the mechanisms of platelets "education" by cancer cells and the crosstalk between platelets and tumor are still unclear, and the role of "tumor educated platelets" (TEPs) is achieving growing interest in cancer research. TEPs are a biological source of cancer-derived biomarkers, especially RNAs that are protected by platelets membrane from circulating RNases, and could serve as a non-invasive tool for tumor detection, molecular profiling and evolution during therapy in clinical practice. Moreover, short platelet lifespan offers the possibility to get a snapshot assessment of cancer molecular profile, providing a real-time tool. We review and discuss the potential and the clinical utility, in terms of cancer diagnosis and monitoring, of platelet count together with other morphological parameters and of the more recent and innovative TEP profiling.
Collapse
Affiliation(s)
- Sara Bravaccini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via P. Maroncelli 40, 47014, Meldola, Italy.
| | - Elisa Boldrin
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128, Padua, Italy.
| | - Giorgia Gurioli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via P. Maroncelli 40, 47014, Meldola, Italy.
| | - Gianluca Tedaldi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via P. Maroncelli 40, 47014, Meldola, Italy.
| | - Maria Assunta Piano
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128, Padua, Italy.
| | - Matteo Canale
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via P. Maroncelli 40, 47014, Meldola, Italy.
| | - Matteo Curtarello
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128, Padua, Italy.
| | - Paola Ulivi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via P. Maroncelli 40, 47014, Meldola, Italy.
| | - Pierluigi Pilati
- Surgical Oncology of Digestive Tract Unit, Veneto Institute of Oncology IOV-IRCCS, 35128, Padova, Italy.
| |
Collapse
|
4
|
Su J, Yang L, Sun Z, Zhan X. Personalized Drug Therapy: Innovative Concept Guided With Proteoformics. Mol Cell Proteomics 2024; 23:100737. [PMID: 38354979 PMCID: PMC10950891 DOI: 10.1016/j.mcpro.2024.100737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/29/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024] Open
Abstract
Personalized medicine can reduce adverse effects, enhance drug efficacy, and optimize treatment outcomes, which represents the essence of personalized medicine in the pharmacy field. Protein drugs are crucial in the field of personalized drug therapy and are currently the mainstay, which possess higher target specificity and biological activity than small-molecule chemical drugs, making them efficient in regulating disease-related biological processes, and have significant potential in the development of personalized drugs. Currently, protein drugs are designed and developed for specific protein targets based on patient-specific protein data. However, due to the rapid development of two-dimensional gel electrophoresis and mass spectrometry, it is now widely recognized that a canonical protein actually includes multiple proteoforms, and the differences between these proteoforms will result in varying responses to drugs. The variation in the effects of different proteoforms can be significant and the impact can even alter the intended benefit of a drug, potentially making it harmful instead of lifesaving. As a result, we propose that protein drugs should shift from being targeted through the lens of protein (proteomics) to being targeted through the lens of proteoform (proteoformics). This will enable the development of personalized protein drugs that are better equipped to meet patients' specific needs and disease characteristics. With further development in the field of proteoformics, individualized drug therapy, especially personalized protein drugs aimed at proteoforms as a drug target, will improve the understanding of disease mechanisms, discovery of new drug targets and signaling pathways, provide a theoretical basis for the development of new drugs, aid doctors in conducting health risk assessments and making more cost-effective targeted prevention strategies conducted by artificial intelligence/machine learning, promote technological innovation, and provide more convenient treatment tailored to individualized patient profile, which will benefit the affected individuals and society at large.
Collapse
Affiliation(s)
- Junwen Su
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lamei Yang
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ziran Sun
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xianquan Zhan
- Medical Science and Technology Innovation Center, Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
5
|
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
|
6
|
Lipshitz M, Visser J, Anderson R, Nel DG, Smit T, Steel HC, Rapoport B. Emerging markers of cancer cachexia and their relationship to sarcopenia. J Cancer Res Clin Oncol 2023; 149:17511-17527. [PMID: 37906352 PMCID: PMC10657295 DOI: 10.1007/s00432-023-05465-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/06/2023] [Indexed: 11/02/2023]
Abstract
PURPOSE Emerging biomarkers of cancer cachexia and their roles in sarcopenia and prognosis are poorly understood. Baseline assessments of anthropometrics, sarcopenia, cachexia status and biomarkers of cachexia were measured in patients with advanced cancer and healthy controls. Thereafter, relationships of the biomarkers with cachexia and sarcopenia were explored. METHODS A prospective case-control design was used, including 40 patients with advanced cancer and 40 gender, age-matched controls. Bioelectrical impedance [skeletal muscle index (SMI)] and hand dynamometry [hand grip strength (HGS)] assessed sarcopenia and a validated tool classified cancer cachexia. Albumin, lymphocyte and platelet counts, haemoglobin, C-reactive protein (CRP), pro-inflammatory cytokines/chemokines and citrullinated histone H3 (H3Cit) were measured. RESULTS Patients had significantly lower SMI (6.67 kg/m2 versus 7.67 kg/m2, p = < 0.01) and HGS (24.42 kg versus 29.62 kg) compared to controls, with 43% being sarcopenic. Significant differences were found for albumin, lymphocyte and platelet counts, haemoglobin, CRP, and tumour necrosis factor α (TNFα), (p < 0.01). Interleukin (IL)-6 (p < 0.04), IL-8 (p = 0.02), neutrophil/lymphocyte ratio (NLR), p = 0.02, platelet/lymphocyte (PLR) ratio, p < 0.01 and systemic immune inflammatory index (SII), p < 0.01 differed significantly. No difference was observed for CXC motif chemokine ligand 5 [CXCL5 or epithelial neutrophil-activating peptide 78 (ENA78)] or H3Cit. Albumin and haemoglobin correlated negatively with total protein, skeletal muscle mass and SMI (all p < 0.01). The presence of sarcopenia associated significantly with albumin, haemoglobin and CRP. CONCLUSION Significant relationships and differences of haemoglobin, CRP and albumin supports future use of these biomarkers in cancer cachexia. CXCL5 and H3Cit as valuable biomarkers in cancer cachexia remains to be defined.
Collapse
Affiliation(s)
- Melanie Lipshitz
- Division of Human Nutrition, Stellenbosch University, Stellenbosch, South Africa.
- Melanie Levy Dietician, 1 Mid Way Road, Glenhazel, Johannesburg, South Africa.
| | - J Visser
- Division of Human Nutrition, Stellenbosch University, Stellenbosch, South Africa
| | - R Anderson
- Department of Immunology, University of Pretoria, Pretoria, South Africa
| | - D G Nel
- Centre for Statistical Consultation, Stellenbosch University, Stellenbosch, South Africa
| | - T Smit
- The Medical Oncology Centre of Rosebank, Johannesburg, South Africa
| | - H C Steel
- Department of Immunology, University of Pretoria, Pretoria, South Africa
| | - B Rapoport
- Department of Immunology, University of Pretoria, Pretoria, South Africa
- The Medical Oncology Centre of Rosebank, Johannesburg, South Africa
| |
Collapse
|
7
|
García Á, Aslan JE. Special review series: provocative questions in platelet omics studies. Platelets 2023; 34:2259169. [PMID: 37726881 DOI: 10.1080/09537104.2023.2259169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Affiliation(s)
- Ángel García
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, and Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Joseph E Aslan
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
8
|
Deshpande AS, Muraoka W, Wait J, Çolak A, Andreescu S. Direct real-time measurements of superoxide release from skeletal muscles in rat limbs and human blood platelets using an implantable Cytochrome C microbiosensor. Biosens Bioelectron 2023; 240:115664. [PMID: 37689016 DOI: 10.1016/j.bios.2023.115664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/19/2023] [Accepted: 09/01/2023] [Indexed: 09/11/2023]
Abstract
Oxidative stress and excessive accumulation of the superoxide (O2.-) anion are at the genesis of many pathological conditions and the onset of several diseases. The real time monitoring of (O2.-) release is important to assess the extent of oxidative stress in these conditions. Herein, we present the design, fabrication and characterization of a robust (O2.-) biosensor using a simple and straightforward procedure involving deposition of a uniform layer of L-Cysteine on a gold wire electrode to which Cytochrome C (Cyt c) was conjugated. The immobilized layers, studied using conductive Atomic Force Microscopy (c-AFM) revealed a stable and uniformly distributed redox protein on the gold surface, visualized as conductivity and surface topographical plots. The biosensor enabled detection of (O2.-) at an applied potential of 0.15 V with a sensitivity of 42.4 nA/μM and a detection limit of 2.4 nM. Utility of the biosensor was demonstrated in measurements of real time (O2.-) release in activated human blood platelets and skeletal rat limb muscles following ischemia reperfusion injury (IRI), confirming the biosensor's stability and robustness for measurements in complex biological systems. The results demonstrate the ability of these biosensors to monitor real time release of (O2.-) and estimate the extent of oxidative injury in models that could easily be translated to human pathologies.
Collapse
Affiliation(s)
- Aaditya S Deshpande
- Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699-5810, USA
| | - Wayne Muraoka
- U.S. Army Institute of Surgical Research, Blood and Shock Resuscitation, Fort Sam Houston, TX, 78234, USA
| | - James Wait
- Department of Physics, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699, USA
| | - Arzu Çolak
- Department of Physics, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699, USA
| | - Silvana Andreescu
- Department of Chemistry and Biomolecular Science, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699-5810, USA.
| |
Collapse
|
9
|
Li X, Wang Q. Platelet-Derived Microparticles and Autoimmune Diseases. Int J Mol Sci 2023; 24:10275. [PMID: 37373420 DOI: 10.3390/ijms241210275] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/11/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Extracellular microparticles provide a means of cell-to-cell communication and can promote information exchanges between adjacent or distant cells. Platelets are cell fragments that are derived from megakaryocytes. Their main functions are to stop bleeding, regulate inflammation, and maintain the integrity of blood vessels. When platelets are activated, they can perform related tasks by secreting platelet-derived microparticles that contain lipids, proteins, nucleic acids, and even organelles. There are differences in the circulating platelet levels in many autoimmune diseases, including rheumatoid arthritis, systemic lupus erythematosus, antiphospholipid antibody syndrome, and Sjogren's syndrome. In this paper, the latest findings in the research field of platelet-derived microparticles are reviewed, including the potential pathogenesis of platelet-derived microparticles in various types of immune diseases, their potential as related markers, and for monitoring the progress and prognosis of disease treatment are expounded.
Collapse
Affiliation(s)
- Xiaoshuai Li
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang 110801, China
| | - Qiushi Wang
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang 110801, China
| |
Collapse
|
10
|
Kreft IC, Huisman EJ, Cnossen MH, van Alphen FPJ, van der Zwaan C, van Leeuwen K, van Spaendonk R, Porcelijn L, Veen CSB, van den Biggelaar M, de Haas M, Meijer AB, Hoogendijk AJ. Proteomic landscapes of inherited platelet disorders with different etiologies. JOURNAL OF THROMBOSIS AND HAEMOSTASIS : JTH 2023; 21:359-372.e3. [PMID: 36700500 DOI: 10.1016/j.jtha.2022.11.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/04/2022] [Accepted: 11/16/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Inherited platelet disorders (IPDs) are a heterogeneous group of rare diseases that are caused by the defects in early megakaryopoiesis, proplatelet formation, and/or mature platelet function. Although genomic sequencing is increasingly used to identify genetic variants underlying IPD, this technique does not disclose resulting molecular changes that impact platelet function. Proteins are the functional units that shape platelet function; however, insights into how variants that cause IPDs impact platelet proteomes are limited. OBJECTIVES The objective of this study was to profile the platelet proteomics signatures of IPDs. METHODS We performed unbiased label-free quantitative mass spectrometry (MS)-based proteome profiling on platelets of 34 patients with IPDs with variants in 13 ISTH TIER1 genes that affect different stages of platelet development. RESULTS In line with the phenotypical heterogeneity between IPDs, proteomes were diverse between IPDs. We observed extensive proteomic alterations in patients with a GFI1B variant and for genetic variants in genes encoding proteins that impact cytoskeletal processes (MYH9, TUBB1, and WAS). Using the diversity between IPDs, we clustered protein dynamics, revealing disrupted protein-protein complexes. This analysis furthermore grouped proteins with similar cellular function and location, classifying mitochondrial protein constituents and identifying both known and putative novel alpha granule associated proteins. CONCLUSIONS With this study, we demonstrate a MS-based proteomics perspective to IPDs. By integrating the effects of IPDs that impact different aspects of platelet function, we dissected the biological contexts of protein alterations to gain further insights into the biology of platelet (dys)function.
Collapse
Affiliation(s)
- Iris C Kreft
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Elise J Huisman
- Department of Pediatric Hematology, Erasmus MC Sophia Children's Hospital, University Medical Center Rotterdam, The Netherlands; Unit of Transfusion Medicine, Sanquin Blood Supply, Amsterdam, The Netherlands
| | - Marjon H Cnossen
- Department of Pediatric Hematology, Erasmus MC Sophia Children's Hospital, University Medical Center Rotterdam, The Netherlands
| | | | - Carmen van der Zwaan
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Karin van Leeuwen
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands
| | - Rosalina van Spaendonk
- Department of Immunohematology Diagnostic, Sanquin Diagnostic Services, Amsterdam, The Netherlands; Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Leendert Porcelijn
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Caroline S B Veen
- Department of Hematology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Maartje van den Biggelaar
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands; Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Masja de Haas
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands; Center for Clinical Transfusion Research, Sanquin Research, Amsterdam and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Alexander B Meijer
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands; Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Arie J Hoogendijk
- Department of Molecular Hematology, Sanquin Research, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Platelet Membrane Proteins as Pain Biomarkers in Patients with Severe Dementia. Biomedicines 2023; 11:biomedicines11020380. [PMID: 36830917 PMCID: PMC9953643 DOI: 10.3390/biomedicines11020380] [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: 12/24/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
Pain is one of the most frequent health problems, and its evaluation and therapeutic approach largely depend on patient self-report. When it is not possible to obtain a self-report, the therapeutic decision becomes more difficult and limited. This study aims to evaluate whether some membrane platelet proteins could be of value in pain characterization. To achieve this goal, we used 53 blood samples obtained from palliative patients, 44 with non-oncological pain and nine without pain. We observed in patients with pain a decrease in the percentage of platelets expressing CD36, CD49f, and CD61 and in the expression levels of CD49f and CD61 when compared with patients without pain. Besides that, an increase in the percentage of platelets expressing CD62p was observed in patients with pain. These results suggest that the levels of these platelet cluster differentiations (CDs) could have some value as pain biomarkers objectively since they are not dependent on the patient's participation. Likewise, CD40 seems to have some importance as a biomarker of moderate and/or severe pain. The identification of pain biomarkers such as CD40, CD49f, CD62p and CD61 can lead to an adjustment of the therapeutic strategy, contributing to a faster and more adequate control of pain and reduction in patient-associated suffering.
Collapse
|
12
|
Filippelli A, Del Gaudio C, Simonis V, Ciccone V, Spini A, Donnini S. Scoping Review on Platelets and Tumor Angiogenesis: Do We Need More Evidence or Better Analysis? Int J Mol Sci 2022; 23:13401. [PMID: 36362186 PMCID: PMC9656254 DOI: 10.3390/ijms232113401] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 08/24/2023] Open
Abstract
Platelets are an active component of the tumor microenvironment (TME), involved in the regulation of multiple tumor processes, including angiogenesis. They are generated rich in angiogenic factors in their granules to actively participate in the hemostatic process by megakaryocytes and further enriched in angiogenic factors by all components of the tumor microenvironment to control the angiogenic process because of their preferential relationship with the endothelial component of vessels. In recent decades, the literature has reported a great deal of evidence on the role of platelets in tumor angiogenesis; however, it is unclear whether the number or mean volume of platelets and/or their content and localization in TME may have clinical relevance in the choice and management of therapy for the cancer patient. In this scoping review, we collected and critically reviewed the scientific evidence supporting a close relationship between platelets, cancer, and angiogenesis. The aim of this work was to define the landscape of platelet-activated angiogenesis in cancer progression and analyze what and how much evidence is present in the last 20 years in the literature at both the preclinical and clinical levels, to answer whether platelets could be a useful determinant for analyzing tumor angiogenesis. In conclusion, this scoping review indicates that there is much evidence, both preclinical and clinical, but in the preclinical context, studies demonstrate the direct involvement of platelets in tumor angiogenesis; in the clinical context the evidence is indirect, though strong, and the indication of how and to what extent platelet content contributes to tumor angiogenesis is lacking. So, do we need more evidence or better analysis? More molecular and quali-quantitative data is needed to translate the results obtained in preclinical studies into the clinical setting. This information about platelets, if correlated with tumor type and its biology, including tumor vasculature, type of angiogenesis, and patient characteristics (age, sex, comorbidities, drug treatments for chronic diseases) could be an important pa- rameter for correlating platelet biology to angiogenesis, for personalizing cancer therapy, and for clinical prognosis.
Collapse
Affiliation(s)
- Arianna Filippelli
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Cinzia Del Gaudio
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Vittoria Simonis
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Valerio Ciccone
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Andrea Spini
- Department of Medical Science, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
- Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
13
|
Ormazabal P, Rodriguez L, Paredes A, Morales G, Fuentes E, Palomo I. Antiplatelet activity of Lampaya medicinalis Phil. in human platelets. NFS JOURNAL 2022. [DOI: 10.1016/j.nfs.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
14
|
Platelets’ Nanomechanics and Morphology in Neurodegenerative Pathologies. Biomedicines 2022; 10:biomedicines10092239. [PMID: 36140340 PMCID: PMC9496241 DOI: 10.3390/biomedicines10092239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
The imaging and force–distance curve modes of atomic force microscopy (AFM) are explored to compare the morphological and mechanical signatures of platelets from patients diagnosed with classical neurodegenerative diseases (NDDs) and healthy individuals. Our data demonstrate the potential of AFM to distinguish between the three NDDs—Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) and Alzheimer’s disease (AD), and normal healthy platelets. The common features of platelets in the three pathologies are reduced membrane surface roughness, area and height, and enhanced nanomechanics in comparison with healthy cells. These changes might be related to general phenomena associated with reorganization in the platelet membrane morphology and cytoskeleton, a key factor for all platelets’ functions. Importantly, the platelets’ signatures are modified to a different extent in the three pathologies, most significant in ALS, less pronounced in PD and the least in AD platelets, which shows the specificity associated with each pathology. Moreover, different degree of activation, distinct pseudopodia and nanocluster formation characterize ALS, PD and AD platelets. The strongest alterations in the biophysical properties correlate with the highest activation of ALS platelets, which reflect the most significant changes in their nanoarchitecture. The specific platelet signatures that mark each of the studied pathologies can be added as novel biomarkers to the currently used diagnostic tools.
Collapse
|
15
|
Chen S, Zhang L, Feng B, Wang W, Liu D, Zhao X, Yu C, Wang X, Gao Y. MiR-550a-3p restores damaged vascular smooth muscle cells by inhibiting thrombomodulin in an <em>in vitro</em> atherosclerosis model. Eur J Histochem 2022; 66. [PMID: 35855629 PMCID: PMC9335314 DOI: 10.4081/ejh.2022.3429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 07/04/2022] [Indexed: 11/22/2022] Open
Abstract
Thrombomodulin (TM) is involved in the pathological process of atherosclerosis; however, the underlying mechanism remains unclear. Oxidised low-density lipoprotein (Ox-LDL; 100 μg/mL) was used to induce human vascular smooth muscle cells (HVSMCs) into a stable atherosclerotic cell model. The expression levels of miR-550a-3p and TM were detected by real-time reverse transcription-polymerase chain reaction. Cell proliferation was estimated using CCK8 and EDU assays. Wound scratch and transwell assays were used to measure the ability of cells to invade and migrate. Propidium iodide fluorescence-activated cell sorting was used to detect apoptosis and cell cycle changes. A dual-luciferase reporter assay was performed to determine the binding of miR-550a-3p to TM. Our results suggested the successful development of a cellular atherosclerosis model. Our data revealed that TM overexpression significantly promoted the proliferation, invasion, migration, and apoptosis of HVSMCs as well as cell cycle changes. Upregulation of miR-550a-3p inhibited the growth and metastasis of HVSMCs. Furthermore, miR-550a-3p was confirmed to be a direct target of TM. Restoration of miR-550a-3p expression rescued the effects of TM overexpression. Thus, miR-550a-3p might play a role in atherosclerosis and, for the first time, normalised the function of injured vascular endothelial cells by simultaneous transfection of TM and miR-550a-3p. These results suggest that the miR-550a-3p/TM axis is a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Shiyuan Chen
- The First Clinical College, Jinan University, Guangzhou, Guangdong; Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui.
| | - Longfei Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui.
| | - Benchi Feng
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui.
| | - Wei Wang
- Department of Oncological Surgery, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui.
| | - Delang Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui.
| | - Xinyu Zhao
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui.
| | - Chaowen Yu
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui.
| | - Xiaogao Wang
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui.
| | - Yong Gao
- The First Clinical College, Jinan University, Guangzhou, Guangdong; Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui.
| |
Collapse
|
16
|
Stampouloglou PK, Siasos G, Bletsa E, Oikonomou E, Vogiatzi G, Kalogeras K, Katsianos E, Vavuranakis MA, Souvaliotis N, Vavuranakis M. The Role of Cell Derived Microparticles in Cardiovascular Diseases: Current Concepts. Curr Pharm Des 2022; 28:1745-1757. [DOI: 10.2174/1381612828666220429081555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/15/2022] [Indexed: 12/07/2022]
Abstract
Abstract:
Cardiovascular disease remains the main cause of human morbidity and mortality in the developed countries. Microparticles (MPs) are small vesicles originating from the cell membrane as a result of various stimuli and particularly of biological processes that constitute the pathophysiology of atherosclerosis, such as endothelial damage. They form vesicles that can transfer various molecules and signals to remote target cells without direct cell to cell interaction. Circulating microparticles have been associated with cardiovascular diseases. Therefore, many studies have been designed to further investigate the role of microparticles as biomarkers for diagnosis, prognosis, and disease monitoring. To this concept the pro-thrombotic and atherogenic potential of platelets and endothelial derived MPs has gain research interest especially concerning accelerate atherosclerosis and acute coronary syndrome triggering and prognosis. MPs especially of endothelial origin have been investigated in different clinical scenarios of heart failure and in association of left ventricular loading conditions. Finally, most cardiovascular risk factors present unique patterns of circulating MPs population, highlighting their pathophysiologic link to cardiovascular disease progression. In this review article we present a synopsis of the biogenesis and characteristics of microparticles, as well as the most recent data concerning their implication in the cardiovascular settings.
Collapse
Affiliation(s)
- Panagiota K. Stampouloglou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Evanthia Bletsa
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Georgia Vogiatzi
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Efstratios Katsianos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Michael-Andrew Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Nektarios Souvaliotis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens. Greece
| |
Collapse
|
17
|
Platelet Membrane: An Outstanding Factor in Cancer Metastasis. MEMBRANES 2022; 12:membranes12020182. [PMID: 35207103 PMCID: PMC8875259 DOI: 10.3390/membranes12020182] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/02/2022]
Abstract
In addition to being biological barriers where the internalization or release of biomolecules is decided, cell membranes are contact structures between the interior and exterior of the cell. Here, the processes of cell signaling mediated by receptors, ions, hormones, cytokines, enzymes, growth factors, extracellular matrix (ECM), and vesicles begin. They triggering several responses from the cell membrane that include rearranging its components according to the immediate needs of the cell, for example, in the membrane of platelets, the formation of filopodia and lamellipodia as a tissue repair response. In cancer, the cancer cells must adapt to the new tumor microenvironment (TME) and acquire capacities in the cell membrane to transform their shape, such as in the case of epithelial−mesenchymal transition (EMT) in the metastatic process. The cancer cells must also attract allies in this challenging process, such as platelets, fibroblasts associated with cancer (CAF), stromal cells, adipocytes, and the extracellular matrix itself, which limits tumor growth. The platelets are enucleated cells with fairly interesting growth factors, proangiogenic factors, cytokines, mRNA, and proteins, which support the development of a tumor microenvironment and support the metastatic process. This review will discuss the different actions that platelet membranes and cancer cell membranes carry out during their relationship in the tumor microenvironment and metastasis.
Collapse
|
18
|
Kim CJ, Kim J, Sabaté Del Río J, Ki DY, Kim J, Cho YK. Fully automated light transmission aggregometry on a disc for platelet function tests. LAB ON A CHIP 2021; 21:4707-4715. [PMID: 34752594 DOI: 10.1039/d1lc00708d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Platelet function tests, a group of assays that measure the ability of platelets to aggregate and promote clotting in a sample of blood, are performed in various medical fields to assess inherited platelet function disorders and monitor antiplatelet therapies. Light transmission aggregometry (LTA) is considered the gold standard for platelet function assessment. However, the lack of a standardized protocol is a major drawback when applied at the point of care. Moreover, it is a time-consuming and labor-intensive assay that requires a large volume of blood. Here, we describe the design, fabrication, and operation of a centrifugal microfluidic disc that can perform a fully automated LTA assay from a small volume of a whole blood sample (<1 mL), achieving highly reproducible results (3.2% coefficient of variation) within a short period (<25 min). The assays performed with this device yield more precise and accurate results than traditional LTA because of the automation of the reaction steps, minimal human operation, robust detection strategy via the distinctive structure of the microfluidic chamber, and quick analysis that minimizes the adverse effects of platelet instability.
Collapse
Affiliation(s)
- Chi-Ju Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Jungmin Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Jonathan Sabaté Del Río
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Dong Yeob Ki
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Junyoung Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| | - Yoon-Kyoung Cho
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea.
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
| |
Collapse
|
19
|
Locatelli L, Colciago A, Castiglioni S, Maier JA. Platelets in Wound Healing: What Happens in Space? Front Bioeng Biotechnol 2021; 9:716184. [PMID: 34760877 PMCID: PMC8572965 DOI: 10.3389/fbioe.2021.716184] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/11/2021] [Indexed: 12/21/2022] Open
Abstract
Beyond their fundamental role in hemostasis, platelets importantly contribute to other processes aimed at maintaining homeostasis. Indeed, platelets are a natural source of growth factors and also release many other substances-such as fibronectin, vitronectin, sphingosine 1-phosphate-that are important in maintaining healthy tissues, and ensuring regeneration and repair. Despite rare thrombotic events have been documented in astronauts, some in vivo and in vitro studies demonstrate that microgravity affects platelet's number and function, thus increasing the risk of hemorrhages and contributing to retard wound healing. Here we provide an overview about events linking platelets to the impairment of wound healing in space, also considering, besides weightlessness, exposure to radiation and psychological stress. In the end we discuss the possibility of utilizing platelet rich plasma as a tool to treat skin injuries eventually occurring during space missions.
Collapse
Affiliation(s)
- Laura Locatelli
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milan, Italy
| | - Alessandra Colciago
- Department of Pharmacological and Biomolecular Sciences, Università di Milano, Milan, Italy
| | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milan, Italy
| | - Jeanette A Maier
- Department of Biomedical and Clinical Sciences L. Sacco, Università di Milano, Milan, Italy.,Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa), Università di Milano, Milan, Italy
| |
Collapse
|
20
|
Antsiferov OV, Cherevatenko RF, Korokin MV, Gureev VV, Gureeva AV, Zatolokina MA, Avdeyeva EV, Zhilinkova LA, Kolesnik IM. A new EPOR/CD131 heteroreceptor agonist EP-11-1: a neuroprotective effect in experimental traumatic brain injury. RESEARCH RESULTS IN PHARMACOLOGY 2021. [DOI: 10.3897/rrpharmacology.7.75301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: EP-11-1 (UEHLERALNSS) is a short-chain erythropoietin derivative without have erythropoietic activity. It was created by modifying a peptide mimicking the spatial structure of the erythropoietin a-helix B pHBSP. One of the promising directions of its administration is the correction of morphofunctional disorders that occur in traumatic brain injury (TBI).
Materials and methods: The study was performed in 160 male Wistar rats, weighing 180–200 g.TBI was simulated using the drop-weight method. To assess the emerging morphofunctional disorders and a degree of their correction, we used the severity of neurological deficit, indicators of locomotor activity and exploration, a marker of brain injury S100B and morphological examination.
Results and discussion: The combined administration of a new EPOR/CD131 heteroreceptor agonist EP-11-1 with citicoline and trimetazidine led to a more pronounced correction of the neurological deficit when compared not only to the group of the ”untreated” animals, but also to the groups of animals to which these drugs had been administered as monotherapy (p < 0.05). The same tendency was also observed in the study of locomotor activity and exploration. A biochemical study showed that the administration of all three combinations led to a statistically significant (p < 0.05) decrease in the S-100B concentration compared not only to the group of “untreated” animals, but also to the groups of animals to which these drugs had been administered as a monotherapy.
Conclusion: The results of the conducted experiments prove the most pronounced positive dynamics in the combined administration of the new EPOR/CD131 heteroreceptor agonist EP-11-1with citicoline and trimetazidine.
Collapse
|
21
|
Kopeikina E, Ponomarev ED. The Role of Platelets in the Stimulation of Neuronal Synaptic Plasticity, Electric Activity, and Oxidative Phosphorylation: Possibilities for New Therapy of Neurodegenerative Diseases. Front Cell Neurosci 2021; 15:680126. [PMID: 34335186 PMCID: PMC8318360 DOI: 10.3389/fncel.2021.680126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/23/2021] [Indexed: 12/04/2022] Open
Abstract
The central nervous system (CNS) is highly vascularized where neuronal cells are located in proximity to endothelial cells, astroglial limitans, and neuronal processes constituting integrated neurovascular units. In contrast to many other organs, the CNS has a blood-brain barrier (BBB), which becomes compromised due to infection, neuroinflammation, neurodegeneration, traumatic brain injury, and other reasons. BBB disruption is presumably involved in neuronal injury during epilepsy and psychiatric disorders. Therefore, many types of neuropsychological disorders are accompanied by an increase in BBB permeability leading to direct contact of circulating blood cells in the capillaries with neuronal cells in the CNS. The second most abundant type of blood cells are platelets, which come after erythrocytes and outnumber ~100-fold circulating leukocytes. When BBB becomes compromised, platelets swiftly respond to the vascular injury and become engaged in thrombosis and hemostasis. However, more recent studies demonstrated that platelets could also enter CNS parenchyma and directly interact with neuronal cells. Within CNS, platelets become activated by recognizing major brain gangliosides on the surface of astrocytes and neurons and releasing a milieu of pro-inflammatory mediators, neurotrophic factors, and neurotransmitters. Platelet-derived factors directly stimulate neuronal electric and synaptic activity and promote the formation of new synapses and axonal regrowth near the site of damage. Despite such active involvement in response to CNS damage, the role of platelets in neurological disorders was not extensively studied, which will be the focus of this review.
Collapse
Affiliation(s)
| | - Eugene D. Ponomarev
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
22
|
Williams K, Carrasquilla GD, Ingerslev LR, Hochreuter MY, Hansson S, Pillon NJ, Donkin I, Versteyhe S, Zierath JR, Kilpeläinen TO, Barrès R. Epigenetic rewiring of skeletal muscle enhancers after exercise training supports a role in whole-body function and human health. Mol Metab 2021; 53:101290. [PMID: 34252634 PMCID: PMC8355925 DOI: 10.1016/j.molmet.2021.101290] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives Regular physical exercise improves health by reducing the risk of a plethora of chronic disorders. We hypothesized that endurance exercise training remodels the activity of gene enhancers in skeletal muscle and that this remodeling contributes to the beneficial effects of exercise on human health. Methods and results By studying changes in histone modifications, we mapped the genome-wide positions and activities of enhancers in skeletal muscle biopsies collected from young sedentary men before and after 6 weeks of endurance exercise. We identified extensive remodeling of enhancer activities after exercise training, with a large subset of the remodeled enhancers located in the proximity of genes transcriptionally regulated after exercise. By overlapping the position of enhancers with genetic variants, we identified an enrichment of disease-associated genetic variants within the exercise-remodeled enhancers. Conclusion Our data provide evidence of a functional link between epigenetic rewiring of enhancers to control their activity after exercise training and the modulation of disease risk in humans. Exercise training changes in skeletal muscle gene expression is enriched for secreted factors. The activity of skeletal muscle enhancers undergoes substantial remodeling after exercise training. Skeletal muscle enhancer activity and gene transcription are strongly associated. Exercise training-remodeled enhancer regions are enriched for GWAS SNPs associated with human traits and diseases.
Collapse
Affiliation(s)
- Kristine Williams
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Germán D Carrasquilla
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Lars Roed Ingerslev
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Mette Yde Hochreuter
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Svenja Hansson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Nicolas J Pillon
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Ida Donkin
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Soetkin Versteyhe
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark; Department of Physiology and Pharmacology, Karolinska Institutet, 171 77, Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark.
| |
Collapse
|
23
|
Kleinbongard P, Andreadou I, Vilahur G. The platelet paradox of injury versus protection in myocardial infarction-has it been overlooked? Basic Res Cardiol 2021; 116:37. [PMID: 34037862 PMCID: PMC8150149 DOI: 10.1007/s00395-021-00876-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Gemma Vilahur
- CIBERCV, Instituto Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair Autonomous University of Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
24
|
Shevchuk O, Begonja AJ, Gambaryan S, Totzeck M, Rassaf T, Huber TB, Greinacher A, Renne T, Sickmann A. Proteomics: A Tool to Study Platelet Function. Int J Mol Sci 2021; 22:ijms22094776. [PMID: 33946341 PMCID: PMC8125008 DOI: 10.3390/ijms22094776] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 12/22/2022] Open
Abstract
Platelets are components of the blood that are highly reactive, and they quickly respond to multiple physiological and pathophysiological processes. In the last decade, it became clear that platelets are the key components of circulation, linking hemostasis, innate, and acquired immunity. Protein composition, localization, and activity are crucial for platelet function and regulation. The current state of mass spectrometry-based proteomics has tremendous potential to identify and quantify thousands of proteins from a minimal amount of material, unravel multiple post-translational modifications, and monitor platelet activity during drug treatments. This review focuses on the role of proteomics in understanding the molecular basics of the classical and newly emerging functions of platelets. including the recently described role of platelets in immunology and the development of COVID-19.The state-of-the-art proteomic technologies and their application in studying platelet biogenesis, signaling, and storage are described, and the potential of newly appeared trapped ion mobility spectrometry (TIMS) is highlighted. Additionally, implementing proteomic methods in platelet transfusion medicine, and as a diagnostic and prognostic tool, is discussed.
Collapse
Affiliation(s)
- Olga Shevchuk
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V, Bunsen-Kirchhoff-Straße 11, 44139 Dortmund, Germany
- Department of Immunodynamics, Institute of Experimental Immunology and Imaging, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
- Correspondence: (O.S.); (A.S.)
| | - Antonija Jurak Begonja
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia;
| | - Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Torez pr. 44, 194223 St. Petersburg, Russia;
| | - Matthias Totzeck
- West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany; (M.T.); (T.R.)
| | - Tienush Rassaf
- West German Heart and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany; (M.T.); (T.R.)
| | - Tobias B. Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Andreas Greinacher
- Institut für Immunologie und Transfusionsmedizin, Universitätsmedizin Greifswald, Sauerbruchstraße, 17475 Greifswald, Germany;
| | - Thomas Renne
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany;
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V, Bunsen-Kirchhoff-Straße 11, 44139 Dortmund, Germany
- Medizinisches Proteom-Center (MPC), Medizinische Fakultät, Ruhr-Universität Bochum, 44801 Bochum, Germany
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen AB24 3FX, UK
- Correspondence: (O.S.); (A.S.)
| |
Collapse
|
25
|
Casagrande N, Borghese C, Agostini F, Durante C, Mazzucato M, Colombatti A, Aldinucci D. In Ovarian Cancer Multicellular Spheroids, Platelet Releasate Promotes Growth, Expansion of ALDH+ and CD133+ Cancer Stem Cells, and Protection against the Cytotoxic Effects of Cisplatin, Carboplatin and Paclitaxel. Int J Mol Sci 2021; 22:ijms22063019. [PMID: 33809553 PMCID: PMC7999151 DOI: 10.3390/ijms22063019] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023] Open
Abstract
A high platelet count is associated with a poor prognosis in ovarian cancer (OvCa). Despite good clinical responses with platinating agents in combination with taxanes, numerous OvCa patients relapse due to chemotherapy resistance. Here, we report that treatment of OvCa cells A2780, OVCAR5 and MDAH with releasate from activated platelets (PR) promoted multicellular tumor spheroid (MCTS) formation. These OvCa-MCTSs had increased percentages of CD133+ and aldehyde dehydrogenase (ALDH)+ cells, bona fide markers of OvCa cancer stem cells (CSCs). PR increased OVCAR5- and MDAH-MCTS viability and decreased the cytotoxic and pro-apoptotic effects of paclitaxel, cisplatin and carboplatin. PR increased the volume of spontaneously formed OVCAR8-MCTSs and counteracted their size reduction due to cisplatin, carboplatin and paclitaxel treatment. PR promoted the survival of ALDH+ and CD133+ OvCa cells during cisplatin, carboplatin and paclitaxel treatment. In conclusion, molecules and growth factors released by activated platelets (EGF, PDGF, TGF-β, IGF and CCL5) may protect tumor cells from chemotherapy by promoting the expansion of ALDH+ and CD133+ OvCa-CSCs, favoring drug resistance and tumor relapse.
Collapse
Affiliation(s)
- Naike Casagrande
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (N.C.); (C.B.); (A.C.)
| | - Cinzia Borghese
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (N.C.); (C.B.); (A.C.)
| | - Francesco Agostini
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (F.A.); (C.D.); (M.M.)
| | - Cristina Durante
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (F.A.); (C.D.); (M.M.)
| | - Mario Mazzucato
- Stem Cell Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (F.A.); (C.D.); (M.M.)
| | - Alfonso Colombatti
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (N.C.); (C.B.); (A.C.)
| | - Donatella Aldinucci
- Molecular Oncology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (N.C.); (C.B.); (A.C.)
- Correspondence:
| |
Collapse
|
26
|
Jurado-Escobar R, Doña I, Bogas-Herrera G, Pérez-Sánchez N, Salas M, Laguna JJ, Muñoz-Cano R, Mayorga C, Torres MJ, Cornejo-García JA. Platelet-Adherent Leukocytes Associated With Cutaneous Cross-Reactive Hypersensitivity to Nonsteroidal Anti-Inflammatory Drugs. Front Pharmacol 2021; 11:594427. [PMID: 33658935 PMCID: PMC7919189 DOI: 10.3389/fphar.2020.594427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/19/2020] [Indexed: 11/13/2022] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are among the most highly consumed drugs worldwide and the main triggers of drug hypersensitivity reactions. The most frequent reaction, named cross-reactive NSAID-hypersensitivity, is due to the pharmacological activity of these drugs by blocking the cyclooxygenase-1 enzyme. Such inhibition leads to cysteinyl-leukotriene synthesis, mainly LTE4, which are responsible for the reaction. Although the complete molecular picture of the underlying mechanisms remains elusive, the participation of platelet-adherent leukocytes (CD61+) and integrins have been described for NSAID-exacerbated respiratory disease (NERD). However, there is a lack of information concerning NSAID-induced urticaria/angioedema (NIUA), by far the most frequent clinical phenotype. Here we have evaluated the potential role of CD61+ leukocytes and integrins (CD18, CD11a, CD11b, and CD11c) in patients with NIUA, and included the other two phenotypes with cutaneous involvement, NSAID-exacerbated cutaneous disease (NECD) and blended reactions (simultaneous skin and airways involvement). A group NSAID-tolerant individuals was also included. During the acute phase of the reaction, the three clinical phenotypes showed increased frequencies of CD61+ neutrophils, eosinophils, and monocytes compared to controls, which correlated with urinary LTE4 levels. However, no correlation was found between these variables at basal state. Furthermore, increased expressions of CD18 and CD11a were found in the three CD61+ leukocytes subsets in NIUA, NECD and blended reactions during the acute phase when compared with CD61-leukocyte subpopulations. During the acute phase, CD61+ neutrophils, eosinophils and monocytes showed increased CD18 and CD11a expression when compared with CD61+ leukocytes at basal state. No differences were found when comparing controls and CD61+ leukocytes at basal state. Our results support the participation of platelet-adherent leukocytes and integrins in cutaneous cross-hypersensitivity to NSAIDs and provide a link between these cells and arachidonic acid metabolism. Our findings also suggest that these reactions do not involve a systemic imbalance in the frequency of CD61+ cells/integrin expression or levels of LTE4, which represents a substantial difference to NERD. Although further studies are needed, our results shed light on the molecular basis of cutaneous cross-reactive NSAID-hypersensitivity, providing potential targets for therapy through the inhibition of platelet-leukocyte interactions.
Collapse
Affiliation(s)
- Raquel Jurado-Escobar
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga-IBIMA, Malaga, Spain.,Departamento de Medicina, Universidad de Málaga, Malaga, Spain
| | - Inmaculada Doña
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga-IBIMA, Malaga, Spain.,Allergy Unit, Hospital Regional Universitario de Málaga, Malaga, Spain.,ARADyAL Network, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - María Salas
- Allergy Unit, Hospital Regional Universitario de Málaga, Malaga, Spain
| | - José J Laguna
- ARADyAL Network, Instituto de Salud Carlos III, Madrid, Spain.,Allergy Unit, Allergo-Anaesthesia Unit, Hospital Central de la Cruz Roja, Faculty of Medicine, Alfonso X El Sabio University, Madrid, Spain
| | - Rosa Muñoz-Cano
- ARADyAL Network, Instituto de Salud Carlos III, Madrid, Spain.,Allergy Section, Pneumology Department, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Cristobalina Mayorga
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga-IBIMA, Malaga, Spain.,Allergy Unit, Hospital Regional Universitario de Málaga, Malaga, Spain.,ARADyAL Network, Instituto de Salud Carlos III, Madrid, Spain.,Nanostructures for Diagnosing and Treatment of Allergic Diseases Laboratory, Andalusian Center for Nanomedicine and Biotechnology-BIONAND, Malaga, Spain
| | - María J Torres
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga-IBIMA, Malaga, Spain.,Departamento de Medicina, Universidad de Málaga, Malaga, Spain.,Allergy Unit, Hospital Regional Universitario de Málaga, Malaga, Spain.,ARADyAL Network, Instituto de Salud Carlos III, Madrid, Spain.,Nanostructures for Diagnosing and Treatment of Allergic Diseases Laboratory, Andalusian Center for Nanomedicine and Biotechnology-BIONAND, Malaga, Spain
| | - José A Cornejo-García
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga-IBIMA, Malaga, Spain.,ARADyAL Network, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
27
|
Aslan JE. Platelet Proteomes, Pathways, and Phenotypes as Informants of Vascular Wellness and Disease. Arterioscler Thromb Vasc Biol 2021; 41:999-1011. [PMID: 33441027 PMCID: PMC7980774 DOI: 10.1161/atvbaha.120.314647] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Platelets rapidly undergo responsive transitions in form and function to repair vascular endothelium and mediate hemostasis. In contrast, heterogeneous platelet subpopulations with a range of primed or refractory phenotypes gradually arise in chronic inflammatory and other conditions in a manner that may indicate or support disease. Qualitatively distinguishable platelet phenotypes are increasingly associated with a variety of physiological and pathological circumstances; however, the origins and significance of platelet phenotypic variation remain unclear and conceptually vague. As changes in platelet function in disease exhibit many similarities to platelets following the activation of platelet agonist receptors, the intracellular responses of platelets common to hemostasis and inflammation may provide insights to the molecular basis of platelet phenotype. Here, we review concepts around how protein-level relations-from platelet receptors through intracellular signaling events-may help to define platelet phenotypes in inflammation, immune responses, aging, and other conditions. We further discuss how representing systems-wide platelet proteomics data profiles as circuit-like networks of causally related intracellular events, or, pathway maps, may inform molecular definitions of platelet phenotype. In addition to offering insights into platelets as druggable targets, maps of causally arranged intracellular relations underlying platelet function can also advance precision and interceptive medicine efforts by leveraging platelets as accessible, dynamic, endogenous, circulating biomarkers of vascular wellness and disease. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Joseph E. Aslan
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Chemical Physiology and Biochemistry and School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
28
|
Droplet Microfluidics with Reagent Micromixing for Investigating Intrinsic Platelet Functionality. Cell Mol Bioeng 2021; 14:223-230. [PMID: 34109001 DOI: 10.1007/s12195-020-00665-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/23/2020] [Indexed: 10/22/2022] Open
Abstract
Introduction Precision mapping of the functional structure of platelet populations holds great promise for the identification of hyper-reactive subtypes that are likely to be disease drivers, having value in prognostics and as therapeutic targets. However, the ability to measure the intrinsic functional capacity of individual platelets is confounded by potent paracrine cross-talk, resulting in phenotypic remodeling of the entire platelet population, and in doing so obscuring the identity of hyper-reactive platelets. Methods To address this we have developed a droplet microfluidics strategy for single platelet confinement to exclude paracrine signaling. Consideration of the Poisson distribution was used for high throughput single platelet encapsulation and the preparation of minimal platelet collectives serving as digital models for understanding the role of hyper-reactive platelets coordinating system-level behavior by paracrine signaling. Platelets are retrieved from the droplets for phenotyping using standard flow cytometry. In addition, we have incorporated a staggered herringbone micromixing element for accurate agonist and antibody dispensing in droplets. Results The methodology was used for characterizing sensitivity distributions from healthy blood donors in response to convulxin (agonist of the GPVI receptor, the major platelet receptor for collagen). P-selectin exposure and α IIb β 3 integrin activation were used as analytical end-points to demonstrate the existence of hyper-reactive platelets that direct 20-fold gains in system level sensitivity. Conclusions The analytical workflow represents an enabling tool for the accurate classification of platelet subtypes and description of their underlying biology. Supplementary information The online version of this article (10.1007/s12195-020-00665-6) contains supplementary material, which is available to authorized users.
Collapse
|
29
|
Moin ASM, Al-Qaissi A, Sathyapalan T, Atkin SL, Butler AE. Platelet Protein-Related Abnormalities in Response to Acute Hypoglycemia in Type 2 Diabetes. Front Endocrinol (Lausanne) 2021; 12:651009. [PMID: 33859620 PMCID: PMC8043308 DOI: 10.3389/fendo.2021.651009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Patients with severe COVID-19 infections have coagulation abnormalities indicative of a hypercoagulable state, with thromboembolic complications and increased mortality. Platelets are recognized as mediators of inflammation, releasing proinflammatory and prothrombotic factors, and are hyperactivated in COVID-19 infected patients. Activated platelets have also been reported in type 2 diabetes (T2D) patients, putting these patients at higher risk for thromboembolic complications of COVID-19 infection. METHODS A case-control study of T2D (n=33) and control subjects (n=30) who underwent a hyperinsulinemic clamp to induce normoglycemia in T2D subjects: T2D: baseline glucose 7.5 ± 0.3mmol/l (135.1 ± 5.4mg/dl), reduced to 4.5 ± 0.07mmol/l (81 ± 1.2mg/dl) with 1-hour clamp; Controls: maintained at 5.1 ± 0.1mmol/l (91.9 ± 1.8mg/dl). Slow Off-rate Modified Aptamer (SOMA)-scan plasma protein measurement was used to determine a panel of platelet proteins. RESULTS Prothrombotic platelet proteins were elevated in T2D versus controls: platelet factor 4 (PF4, p<0.05); platelet glycoprotein VI (PGVI p<0.05); P-selectin (p<0.01) and plasminogen activator inhibitor I (PAI-1, p<0.01). In addition, the antithrombotic platelet-related proteins, plasmin (p<0.05) and heparin cofactor II (HCFII, p<0.05), were increased in T2D. Normalization of glucose in the T2D cohort had no effect on platelet protein levels. CONCLUSION T2D patients have platelet hyperactivation, placing them at higher risk for thromboembolic events. When infected with COVID-19, this risk may be compounded, and their propensity for a more severe COVID-19 disease course increased. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ct2/show/NCT03102801, identifier NCT03102801.
Collapse
Affiliation(s)
- Abu Saleh Md Moin
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Ahmed Al-Qaissi
- Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, Hull, United Kingdom
- Academic Endocrinology, Leeds Medical School, Leeds, United Kingdom
| | - Thozhukat Sathyapalan
- Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, Hull, United Kingdom
| | - Stephen L. Atkin
- School of Postgraduate Studies and Research, Royal College of Surgeons in Ireland Bahrain, Adliya, Bahrain
| | - Alexandra E. Butler
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- *Correspondence: Alexandra E. Butler, ;
| |
Collapse
|
30
|
van Geffen JP, Swieringa F, van Kuijk K, Tullemans BME, Solari FA, Peng B, Clemetson KJ, Farndale RW, Dubois LJ, Sickmann A, Zahedi RP, Ahrends R, Biessen EAL, Sluimer JC, Heemskerk JWM, Kuijpers MJE. Mild hyperlipidemia in mice aggravates platelet responsiveness in thrombus formation and exploration of platelet proteome and lipidome. Sci Rep 2020; 10:21407. [PMID: 33293576 PMCID: PMC7722935 DOI: 10.1038/s41598-020-78522-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/23/2020] [Indexed: 01/21/2023] Open
Abstract
Hyperlipidemia is a well-established risk factor for cardiovascular diseases. Millions of people worldwide display mildly elevated levels of plasma lipids and cholesterol linked to diet and life-style. While the prothrombotic risk of severe hyperlipidemia has been established, the effects of moderate hyperlipidemia are less clear. Here, we studied platelet activation and arterial thrombus formation in Apoe-/- and Ldlr-/- mice fed a normal chow diet, resulting in mildly increased plasma cholesterol. In blood from both knockout mice, collagen-dependent thrombus and fibrin formation under flow were enhanced. These effects did not increase in severe hyperlipidemic blood from aged mice and upon feeding a high-fat diet (Apoe-/- mice). Bone marrow from wild-type or Ldlr-/- mice was transplanted into irradiated Ldlr-/- recipients. Markedly, thrombus formation was enhanced in blood from chimeric mice, suggesting that the hyperlipidemic environment altered the wild-type platelets, rather than the genetic modification. The platelet proteome revealed high similarity between the three genotypes, without clear indication for a common protein-based gain-of-function. The platelet lipidome revealed an altered lipid profile in mildly hyperlipidemic mice. In conclusion, in Apoe-/- and Ldlr-/- mice, modest elevation in plasma and platelet cholesterol increased platelet responsiveness in thrombus formation and ensuing fibrin formation, resulting in a prothrombotic phenotype.
Collapse
Affiliation(s)
- Johanna P van Geffen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Frauke Swieringa
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.,Leibniz Institut für Analytische Wissenschaften - ISAS- e.V, Dortmund, Germany
| | - Kim van Kuijk
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands
| | - Bibian M E Tullemans
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Fiorella A Solari
- Leibniz Institut für Analytische Wissenschaften - ISAS- e.V, Dortmund, Germany
| | - Bing Peng
- Leibniz Institut für Analytische Wissenschaften - ISAS- e.V, Dortmund, Germany
| | - Kenneth J Clemetson
- Department of Haematology, Inselspital, University of Bern, Bern, Switzerland
| | | | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| | - Albert Sickmann
- Leibniz Institut für Analytische Wissenschaften - ISAS- e.V, Dortmund, Germany
| | - René P Zahedi
- Leibniz Institut für Analytische Wissenschaften - ISAS- e.V, Dortmund, Germany.,Segal Cancer Proteomics Centre, Jewish General Hospital, McGill University, Montreal, Canada
| | - Robert Ahrends
- Leibniz Institut für Analytische Wissenschaften - ISAS- e.V, Dortmund, Germany.,Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Wien, Austria
| | - Erik A L Biessen
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands.,Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Judith C Sluimer
- Department of Pathology, CARIM, Maastricht University, Maastricht, The Netherlands.,BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Marijke J E Kuijpers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| |
Collapse
|