1
|
Ding Z, Qi F, Liu L, Wang Z, Zhang N, Lyu X, Sun W, Du J, Song H, Hou H, Guo Y, Wang X, Liu ML, Wei W. Circulating extracellular vesicles as novel biomarkers for pulmonary arterial hypertension in patients with systemic lupus erythematosus. Front Immunol 2024; 15:1374100. [PMID: 39364410 PMCID: PMC11446868 DOI: 10.3389/fimmu.2024.1374100] [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: 01/21/2024] [Accepted: 08/28/2024] [Indexed: 10/05/2024] Open
Abstract
Introduction Pulmonary arterial hypertension (PAH) is a serious complication of systemic lupus erythematosus (SLE) with increased mortality. A prothrombotic state may contribute to pathogenesis of SLE-PAH. Extracellular vesicles (EVs) are known to be associated with thrombosis. Here, we investigated circulating EVs and their associations with SLE-PAH. Methods Eighteen SLE-PAH patients, 36 SLE-non-PAH patients, and 36 healthy controls (HCs) were enrolled. Flow cytometry was used to analyze circulating EVs from leukocytes (LEVs), red blood cells (REVs), platelets (PEVs), endothelial cells (EEVs), and Annexin V+ EVs with membrane phosphatidylserine (PS) exposure. Results Plasma levels of all EV subgroups were elevated in SLE patients with or without PAH compared to HCs. Furthermore, plasma Annexin V+ EVs, LEVs, PEVs, REVs, EEVs, and Annexin V+ REVs were significantly elevated in SLE-PAH patients compared to SLE-non-PAH patients. Additionally, PAH patients with moderate/high SLE showed a significant increase in LEVs, PEVs, REVs, Annexin V+ EVs, and Annexin V+ REVs compared to SLE-non-PAH patients. However, PAH patients with inactive/mild SLE only exhibited elevations in Annexin V+ EVs, REVs, and Annexin V+ REVs. In the SLE-PAH patients, EEVs were positively correlated with pulmonary arterial systolic pressure, while PEVs and EEVs were positively correlated with right ventricular diameter. Moreover, the receiver operating characteristic curve indicated that Annexin V+ EVs, LEVs, PEVs, REVs, EEVs and Annexin V+ REVs could predict the presence of PAH in SLE patients. Importantly, multivariate logistic regression analysis showed that circulating levels of LEVs or REVs, anti-nRNP antibody, and serositis were independent risk factors for PAH in SLE patients. Discussion Findings reveal that specific subgroups of circulating EVs contribute to the hypercoagulation state and the severity of SLE-PAH. Higher plasma levels of LEVs or REVs may serve as biomarkers for SLE-PAH.
Collapse
Affiliation(s)
- Zhe Ding
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Fumin Qi
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Li Liu
- Department of Neurosurgery, Tianjin Institute of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhouming Wang
- Department of Cardiovascular, Tianjin Medical University General Hospital, Tianjin, China
| | - Na Zhang
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Xing Lyu
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Wenwen Sun
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Jun Du
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Haoming Song
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hou Hou
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Ying Guo
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Xiaomei Wang
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| | - Ming-Lin Liu
- Corporal Michael J. Crescenz Veterans Affairs Medical Center (VAMC), Philadelphia, PA, United States
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wei Wei
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Clinical Research Center for Rheumatic and Immune Diseases, Tianjin Science and Technology Bureau, Tianjin, China
| |
Collapse
|
2
|
Gibson AD, Bayrón-Marrero Z, Nieves-Lopez B, Maldonado-Martínez G, Washington AV. High Levels of Triggering Receptor Expressed in Myeloid Cells-Like Transcript-1 Positive, but Not Glycoprotein 1b+, Microparticles Are Associated With Poor Outcomes in Acute Respiratory Distress Syndrome. Crit Care Explor 2024; 6:e1108. [PMID: 38935146 PMCID: PMC11213581 DOI: 10.1097/cce.0000000000001108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024] Open
Abstract
OBJECTIVES To identify triggering receptor expressed in myeloid cells-like transcript-1 positive (TLT-1+) microparticles (MPs) and evaluate if their presence is associated with clinical outcomes and/or disease severity in acute respiratory distress syndrome (ARDS). DESIGN Retrospective cohort study. SETTING ARDS Network clinical trials. PATIENTS A total of 564 patients were diagnosed with ARDS. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Using flow cytometry, we demonstrated the presence of TLT-1+ platelet-derived microparticles (PMP) that bind fibrinogen in plasma samples from fresh donors. We retrospectively quantified TLT-1, glycoprotein (Gp) 1b, or αIIbβIIIa immunopositive microparticles in plasma samples from patients with ARDS enrolled in the ARMA, KARMA, and LARMA (Studies 01 and 03 lower versus higher tidal volume, ketoconazole treatment, and lisofylline treatment Clincial Trials) ARDS Network clinical trials and evaluated the relationship between these measures and clinical outcomes. No associations were found between Gp1b+ MPs and clinical outcomes for any of the cohorts. When stratified by quartile, associations were found for survival, ventilation-free breathing, and thrombocytopenia with αIIbβIIIa+ and TLT-1+ MPs (χ2p < 0.001). Notably, 63 of 64 patients in this study who failed to achieve unassisted breathing had TLT+ PMP in the 75th percentile. In all three cohorts, patients whose TLT+ MP counts were higher than the median had higher Acute Physiology and Chronic Health Evaluation III scores, were more likely to present with thrombocytopenia and were 3.7 times (p < 0.001) more likely to die than patients with lower TLT+ PMP after adjusting for other risk factors. CONCLUSIONS Although both αIIbβIIIa+ and TLT+ microparticles (αIIbβIIIa, TLT-1) were associated with mortality, TLT-1+ MPs demonstrated stronger correlations with Acute Physiology and Chronic Health Evaluation III scores, unassisted breathing, and multiple system organ failure. These findings warrant further exploration of the mechanistic role of TLT-1+ PMP in ARDS or acute lung injury progression.
Collapse
Affiliation(s)
| | | | - Benjamin Nieves-Lopez
- Department of Medicine, University of Puerto Rico Medical Sciences Campus, San Juan, PR
| | | | | |
Collapse
|
3
|
Guo J, Cui B, Zheng J, Yu C, Zheng X, Yi L, Zhang S, Wang K. Platelet-derived microparticles and their cargos: The past, present and future. Asian J Pharm Sci 2024; 19:100907. [PMID: 38623487 PMCID: PMC11016590 DOI: 10.1016/j.ajps.2024.100907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/05/2024] [Accepted: 01/31/2024] [Indexed: 04/17/2024] Open
Abstract
All eukaryotic cells can secrete extracellular vesicles, which have a double-membrane structure and are important players in the intercellular communication involved in a variety of important biological processes. Platelets form platelet-derived microparticles (PMPs) in response to activation, injury, or apoptosis. This review introduces the origin, pathway, and biological functions of PMPs and their importance in physiological and pathological processes. In addition, we review the potential applications of PMPs in cancer, vascular homeostasis, thrombosis, inflammation, neural regeneration, biomarkers, and drug carriers to achieve targeted drug delivery. In addition, we comprehensively report on the origin, biological functions, and applications of PMPs. The clinical transformation, high heterogeneity, future development direction, and limitations of the current research on PMPs are also discussed in depth. Evidence has revealed that PMPs play an important role in cell-cell communication, providing clues for the development of PMPs as carriers for relevant cell-targeted drugs. The development history and prospects of PMPs and their cargos are explored in this guidebook.
Collapse
Affiliation(s)
- Jingwen Guo
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Bufeng Cui
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Jie Zheng
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Chang Yu
- School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xuran Zheng
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Lixin Yi
- School of Pharmacy, China Medical University, Shenyang 110122, China
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Simeng Zhang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang 110001, China
| | - Keke Wang
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang 110001 China
- School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
4
|
Veilleux V, Pichaud N, Boudreau LH, Robichaud GA. Mitochondria Transfer by Platelet-Derived Microparticles Regulates Breast Cancer Bioenergetic States and Malignant Features. Mol Cancer Res 2024; 22:268-281. [PMID: 38085263 DOI: 10.1158/1541-7786.mcr-23-0329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/25/2023] [Accepted: 12/07/2023] [Indexed: 03/02/2024]
Abstract
An increasing number of studies show that platelets as well as platelet-derived microparticles (PMP) play significant roles in cancer malignancy and disease progression. Particularly, PMPs have the capacity to interact and internalize within target cells resulting in the transfer of their bioactive cargo, which can modulate the signaling and activation processes of recipient cells. We recently identified a new subpopulation of these vesicles (termed mitoMPs), which contain functional mitochondria. Given the predominant role of mitochondria in cancer cell metabolism and disease progression, we set out to investigate the impact of mitoMPs on breast cancer metabolic reprograming and phenotypic processes leading to malignancy. Interestingly, we observed that recipient cell permeability to PMP internalization varied among the breast cancer cell types evaluated in our study. Specifically, cells permissive to mitoMPs acquire mitochondrial-dependent functions, which stimulate increased cellular oxygen consumption rates and intracellular ATP levels. In addition, cancer cells co-incubated with PMPs display enhanced malignant features in terms of migration and invasion. Most importantly, the cancer aggressive processes and notable metabolic plasticity induced by PMPs were highly dependent on the functional status of the mitoMP-packaged mitochondria. These findings characterize a new mechanism by which breast cancer cells acquire foreign mitochondria resulting in the gain of metabolic processes and malignant features. A better understanding of these mechanisms may provide therapeutic opportunities through PMP blockade to deprive cancer cells from resources vital in disease progression. IMPLICATIONS We show that the transfer of foreign mitochondria by microparticles modulates recipient cancer cell metabolic plasticity, leading to greater malignant processes.
Collapse
Affiliation(s)
- Vanessa Veilleux
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- New Brunswick Center for Precision Medicine, Moncton, New Brunswick, Canada
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | - Nicolas Pichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- New Brunswick Center for Precision Medicine, Moncton, New Brunswick, Canada
| | - Luc H Boudreau
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- New Brunswick Center for Precision Medicine, Moncton, New Brunswick, Canada
| | - Gilles A Robichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada
- New Brunswick Center for Precision Medicine, Moncton, New Brunswick, Canada
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| |
Collapse
|
5
|
Gharib E, Veilleux V, Boudreau LH, Pichaud N, Robichaud GA. Platelet-derived microparticles provoke chronic lymphocytic leukemia malignancy through metabolic reprogramming. Front Immunol 2023; 14:1207631. [PMID: 37441073 PMCID: PMC10333545 DOI: 10.3389/fimmu.2023.1207631] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/30/2023] [Indexed: 07/15/2023] Open
Abstract
Background It is well established that inflammation and platelets promote multiple processes of cancer malignancy. Recently, platelets have received attention for their role in carcinogenesis through the production of microvesicles or platelet-derived microparticles (PMPs), which transfer their biological content to cancer cells. We have previously characterized a new subpopulation of these microparticles (termed mito-microparticles), which package functional mitochondria. The potential of mitochondria transfer to cancer cells is particularly impactful as many aspects of mitochondrial biology (i.e., cell growth, apoptosis inhibition, and drug resistance) coincide with cancer hallmarks and disease progression. These metabolic aspects are particularly notable in chronic lymphocytic leukemia (CLL), which is characterized by a relentless accumulation of proliferating, immunologically dysfunctional, mature B-lymphocytes that fail to undergo apoptosis. The present study aimed to investigate the role of PMPs on CLL metabolic plasticity leading to cancer cell phenotypic changes. Methods CLL cell lines were co-incubated with different concentrations of human PMPs, and their impact on cell proliferation, mitochondrial DNA copy number, OCR level, ATP production, and ROS content was evaluated. Essential genes involved in metabolic-reprogramming were identified using the bioinformatics tools, examined between patients with early and advanced CLL stages, and then validated in PMP-recipient CLLs. Finally, the impact of the induced metabolic reprogramming on CLLs' growth, survival, mobility, and invasiveness was tested against anti-cancer drugs Cytarabine, Venetoclax, and Plumbagin. Results The data demonstrated the potency of PMPs in inducing tumoral growth and invasiveness in CLLs through mitochondrial internalization and OXPHOS stimulation which was in line with metabolic shift reported in CLL patients from early to advanced stages. This metabolic rewiring also improved CLL cells' resistance to Cytarabine, Venetoclax, and Plumbagin chemo drugs. Conclusion Altogether, these findings depict a new platelet-mediated pathway of cancer pathogenesis. We also highlight the impact of PMPs in CLL metabolic reprogramming and disease progression.
Collapse
Affiliation(s)
- Ehsan Gharib
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- Atlantic Cancer Research Institute, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| | - Vanessa Veilleux
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- Atlantic Cancer Research Institute, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| | - Luc H Boudreau
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| | - Nicolas Pichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| | - Gilles A Robichaud
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
- Atlantic Cancer Research Institute, Moncton, NB, Canada
- New Brunswick Center for Precision Medicine, Moncton, NB, Canada
| |
Collapse
|
6
|
Dahal S, Chaudhary P, Jung YS, Kim JA. Megakaryocyte-Derived IL-8 Acts as a Paracrine Factor for Prostate Cancer Aggressiveness through CXCR2 Activation and Antagonistic AR Downregulation. Biomol Ther (Seoul) 2023; 31:210-218. [PMID: 36787954 PMCID: PMC9970838 DOI: 10.4062/biomolther.2023.005] [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: 01/09/2023] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 02/16/2023] Open
Abstract
Prostate cancer is the fifth leading cause of cancer-related mortality in men, primarily because of treatment resistance, recurrence, and metastasis. In the present study, we investigated the role of paracrine interleukin-8 (IL-8) in the antagonistic expression of IL-8 and androgen receptor (AR), and the contribution of IL-8 to prostate cancer aggressiveness. In hormone-responsive LNCaP cells that do not express IL-8, recombinant IL-8 treatment significantly increased expressions of IL-8, CXC chemokine receptor 2 (CXCR2), matrix metalloproteinase (MMP)-2/9, Snail, and vimentin. IL-8 treatment significantly decreased AR and E-cadherin expression. IL-8-induced gene expression changes were suppressed by navarixin, a CXCR1/2 inhibitor, and gallein, a Gβγ inhibitor. In PC-3 androgen-refractory prostate cancer cells, IL-8 knockdown reduced expressions of CXCR2, MMP-2/9, Snail, and vimentin, and increased AR and E-cadherin expressions at the mRNA and protein levels. Co-culture with MEG-01 human megakaryocytic cells secreting high levels of IL-8 induced gene expression changes in both LNCaP and PC-3 cells, similar to those induced by IL-8 treatment. The altered gene expressions were accompanied by significant activation of transcription factor Snail in LNCaP and PC-3 cells. Treatment with the CXCR blocker navarixin inhibited the invasion of PC-3 cells but not LNCaP cells. However, invasion induced by MEG-01 was inhibited by navarixin in both LNCaP and PC-3 cells. The collective findings demonstrate that IL-8 enhances CXCR2 expression, which antagonistically regulates AR expression. More importantly, through changes in IL-8/CXCR2-regulated gene expression, IL-8 induces antiandrogen therapy resistance and epithelial-mesenchymal transition in prostate cancer.
Collapse
Affiliation(s)
- Sadan Dahal
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Prakash Chaudhary
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Yi-Sook Jung
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
| | - Jung-Ae Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
7
|
Al-Koussa H, AlZaim I, El-Sabban ME. Pathophysiology of Coagulation and Emerging Roles for Extracellular Vesicles in Coagulation Cascades and Disorders. J Clin Med 2022; 11:jcm11164932. [PMID: 36013171 PMCID: PMC9410115 DOI: 10.3390/jcm11164932] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 11/16/2022] Open
Abstract
The notion of blood coagulation dates back to the ancient Greek civilization. However, the emergence of innovative scientific discoveries that started in the seventeenth century formulated the fundamentals of blood coagulation. Our understanding of key coagulation processes continues to evolve, as novel homeostatic and pathophysiological aspects of hemostasis are revealed. Hemostasis is a dynamic physiological process, which stops bleeding at the site of injury while maintaining normal blood flow within the body. Intrinsic and extrinsic coagulation pathways culminate in the homeostatic cessation of blood loss, through the sequential activation of the coagulation factors. Recently, the cell-based theory, which combines these two pathways, along with newly discovered mechanisms, emerged to holistically describe intricate in vivo coagulation mechanisms. The complexity of these mechanisms becomes evident in coagulation diseases such as hemophilia, Von Willebrand disease, thrombophilia, and vitamin K deficiency, in which excessive bleeding, thrombosis, or unnecessary clotting, drive the development and progression of diseases. Accumulating evidence implicates cell-derived and platelet-derived extracellular vesicles (EVs), which comprise microvesicles (MVs), exosomes, and apoptotic bodies, in the modulation of the coagulation cascade in hemostasis and thrombosis. As these EVs are associated with intercellular communication, molecular recycling, and metastatic niche creation, emerging evidence explores EVs as valuable diagnostic and therapeutic approaches in thrombotic and prothrombotic diseases.
Collapse
Affiliation(s)
- Houssam Al-Koussa
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
| | - Marwan E. El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
- Correspondence: ; Tel.: +961-01-350-000 (ext. 4765)
| |
Collapse
|
8
|
Mi Z, Gong L, Kong Y, Zhao P, Yin Y, Xu H, Tian L, Liu Z. Differential expression of exosomal microRNAs in fresh and senescent apheresis platelet concentrates. Platelets 2022; 33:1260-1269. [PMID: 35968647 DOI: 10.1080/09537104.2022.2108541] [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: 10/15/2022]
Abstract
Patients have a high risk of suffering adverse reactions after receiving platelet products stored for 5 days. Bioactive exosomes in platelet products can be accumulated during storage, which is associated with adverse reactions. MicroRNAs are one of the critical cargoes in exosomes, which participate in cell differentiation, metabolism, and immunomodulation. This study intends to elucidate and analyze the differential expression of exosomal microRNAs in apheresis platelet concentrates during storage and predict the potential functions of target genes. Apheresis platelet concentrates were used to isolate exosomes by ultracentrifugation. Exosomes were phenotyped by western blot, transmission electron microscopy, and nano flow cytometry. The differential expression of the exosomal microRNAs was obtained by a microarray test using four bags of apheresis platelets stored for 5 days compared with 1 day. The differentially expressed microRNAs between the two time points were identified, and their target genes were analyzed by miRWalk and miRDB. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to predict the target genes' functions. Fifteen bags of apheresis platelet concentrates stored for 1 day and 5 days were used to verify the microarray results by quantitative reverse transcription-polymerase chain reactions (qRT-PCR). There were 134 microRNAs in total expressed differently in the two groups (day 1 and day 5), with 57 microRNAs up-regulated and 77 down-regulated (|fold change| > 2.0 and P < .05). Thirteen up-regulated microRNAs (hsa-miR-22-3p, hsa-miR-223-3p, hsa-miR-21-5p, hsa-miR-23a-3p, hsa-miR-320b, hsa-let-7a-5p, hsa-miR-25-3p, hsa-miR-126-3p, hsa-miR-320c, hsa-miR-342-3p, hsa-miR-320d, hsa-miR-328-3p, and hsa-miR-320e) detected in all samples were selected to validate the results. The qRT-PCR results showed that five (hsa-miR-22-3p, hsa-miR-223-3p, hsa-miR-21-5p, hsa-miR-23a-3p, and hsa-miR-320b) of them were increased more than 10-fold (P < .001); four (hsa-let-7a-5p, hsa-miR-25-3p, hsa-miR-126-3p, hsa-miR-320c) more than five-fold (P < .001); two (hsa-miR-342-3p and hsa-miR-320d) more than two-fold (P < .05); and two (hsa-miR-328-3p and hsa-miR-320e) more than two-fold (P > .05). Specifically, hsa-miR-22-3p increased 14.6-fold; hsa-miR-223-3p increased 13.0-fold; and hsa-miR-21-5p increased 12.0-fold. Based on bioinformatics functional analysis, target genes of top nine microRNAs (hsa-miR-22-3p, hsa-miR-223-3p, hsa-miR-21-5p, hsa-miR-23a-3p, hsa-miR-320b, hsa-let-7a-5p, hsa-miR-25-3p, hsa-miR-126-3p, and hsa-miR-320c) were annotated with positive regulation of cell proliferation and nervous system development, and mainly enriched in regulating pluripotency of stem cells signaling pathway, prolactin signaling pathway, and FoxO signaling pathway, etc. The prolactin, FoxO, ErbB, and TNF signaling pathway were relevant to immunomodulation. In particular, hsa-miR-22-3p expression was the most different during storage, with a fold change of 14.6, which might be a key mediator.
Collapse
Affiliation(s)
- Ziyue Mi
- Clinical Transfusion Research Center, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, SC, China.,Key Laboratory of Transfusion Adverse Reactions, Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, SC, China
| | - Li Gong
- Key Laboratory of Transfusion Adverse Reactions, Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, SC, China.,School of Public Health, Anhui Medical University, Hefei, AH, China
| | - Yujie Kong
- Clinical Transfusion Research Center, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, SC, China.,Key Laboratory of Transfusion Adverse Reactions, Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, SC, China
| | - Peizhe Zhao
- Clinical Transfusion Research Center, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, SC, China.,Key Laboratory of Transfusion Adverse Reactions, Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, SC, China
| | - Yonghua Yin
- Clinical Transfusion Research Center, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, SC, China.,Key Laboratory of Transfusion Adverse Reactions, Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, SC, China
| | - Haixia Xu
- Clinical Transfusion Research Center, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, SC, China.,Key Laboratory of Transfusion Adverse Reactions, Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, SC, China
| | - Li Tian
- Clinical Transfusion Research Center, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, SC, China.,Key Laboratory of Transfusion Adverse Reactions, Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, SC, China
| | - Zhong Liu
- Clinical Transfusion Research Center, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, SC, China.,Key Laboratory of Transfusion Adverse Reactions, Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, SC, China
| |
Collapse
|
9
|
Wang X, Zhao S, Wang Z, Gao T. Platelets involved tumor cell EMT during circulation: communications and interventions. Cell Commun Signal 2022; 20:82. [PMID: 35659308 PMCID: PMC9166407 DOI: 10.1186/s12964-022-00887-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 04/24/2022] [Indexed: 12/18/2022] Open
Abstract
AbstractDistant spreading of metastatic tumor cells is still the leading cause of tumor death. Metastatic spreading is a complex process, in which epithelial-mesenchymal transition (EMT) is the primary and key event to promote it. Presently, extensive reviews have given insights on the occurrence of EMT at the primary tumor site that depends on invasive properties of tumor cells and the tumor-associated microenvironment. However, essential roles of circulation environment involved in tumor cell EMT is not well summarized. As a main constituent of the blood, platelet is increasingly found to work as an important activator to induce EMT. Therefore, this review aims to emphasize the novel role of platelet in EMT through signal communications between platelets and circulation tumor cells, and illustrate potent interventions aiming at their communications. It may give a complementary view of EMT in addition to the tissue microenvironment, help for better understand the hematogenous metastasis, and also illustrate theoretical and practical basis for the targeted inhibition.
Collapse
|
10
|
Ostermeier B, Soriano-Sarabia N, Maggirwar SB. Platelet-Released Factors: Their Role in Viral Disease and Applications for Extracellular Vesicle (EV) Therapy. Int J Mol Sci 2022; 23:2321. [PMID: 35216433 PMCID: PMC8876984 DOI: 10.3390/ijms23042321] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
Platelets, which are small anuclear cell fragments, play important roles in thrombosis and hemostasis, but also actively release factors that can both suppress and induce viral infections. Platelet-released factors include sCD40L, microvesicles (MVs), and alpha granules that have the capacity to exert either pro-inflammatory or anti-inflammatory effects depending on the virus. These factors are prime targets for use in extracellular vesicle (EV)-based therapy due to their ability to reduce viral infections and exert anti-inflammatory effects. While there are some studies regarding platelet microvesicle-based (PMV-based) therapy, there is still much to learn about PMVs before such therapy can be used. This review provides the background necessary to understand the roles of platelet-released factors, how these factors might be useful in PMV-based therapy, and a critical discussion of current knowledge of platelets and their role in viral diseases.
Collapse
Affiliation(s)
| | | | - Sanjay B. Maggirwar
- Department of Microbiology Immunology and Tropical Medicine, The George Washington University, 2300 I Street NW, Washington, DC 20037, USA; (B.O.); (N.S.-S.)
| |
Collapse
|
11
|
The bone marrow niche from the inside out: how megakaryocytes are shaped by and shape hematopoiesis. Blood 2022; 139:483-491. [PMID: 34587234 PMCID: PMC8938937 DOI: 10.1182/blood.2021012827] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/10/2021] [Indexed: 01/29/2023] Open
Abstract
Megakaryocytes (MKs), the largest of the hematopoietic cells, are responsible for producing platelets by extending and depositing long proplatelet extensions into the bloodstream. The traditional view of megakaryopoiesis describes the cellular journey from hematopoietic stem cells (HSCs) along the myeloid branch of hematopoiesis. However, recent studies suggest that MKs can be generated from multiple pathways, some of which do not require transit through multipotent or bipotent MK-erythroid progenitor stages in steady-state and emergency conditions. Growing evidence suggests that these emergency conditions are due to stress-induced molecular changes in the bone marrow (BM) microenvironment, also called the BM niche. These changes can result from insults that affect the BM cellular composition, microenvironment, architecture, or a combination of these factors. In this review, we explore MK development, focusing on recent studies showing that MKs can be generated from multiple divergent pathways. We highlight how the BM niche may encourage and alter these processes using different mechanisms of communication, such as direct cell-to-cell contact, secreted molecules (autocrine and paracrine signaling), and the release of cellular components (eg, extracellular vesicles). We also explore how MKs can actively build and shape the surrounding BM niche.
Collapse
|
12
|
Brown PA. Differential and targeted vesiculation: pathologic cellular responses to elevated arterial pressure. Mol Cell Biochem 2022; 477:1023-1040. [PMID: 34989921 DOI: 10.1007/s11010-021-04351-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/23/2021] [Indexed: 11/29/2022]
Abstract
Extracellular vesicles are small membrane-enclosed particles released during cell activation or injury. They have been investigated for several decades and found to be secreted in various diseases. Their pathogenic role is further supported by the presence of several important molecules among their cargo, including proteins, lipids, and nucleic acids. Many studies have reported enhanced and targeted extracellular vesicle biogenesis in diseases that involve chronic or transient elevation of arterial pressure resulting in endothelial dysfunction, within either the general circulatory system or specific local vascular beds. In addition, several associated pathologic processes have been studied and reported. However, the role of elevated pressure as a common pathogenic trigger across vascular domains and disease chronicity has not been previously described. This review will therefore summarize our current knowledge of the differential and targeted biogenesis of extracellular vesicles in major diseases that are characterized by elevated arterial pressure leading to endothelial dysfunction and propose a unified theory of pressure-induced extracellular vesicle-mediated pathogenesis.
Collapse
Affiliation(s)
- Paul A Brown
- Department of Basic Medical Sciences, Faculty of Medical Sciences Teaching and Research Complex, The University of the West Indies, Mona, Kingston 7, Jamaica.
| |
Collapse
|
13
|
Wurtzel JGT, Lazar S, Sikder S, Cai KQ, Astsaturov I, Weyrich AS, Rowley JW, Goldfinger LE. Platelet microRNAs inhibit primary tumor growth via broad modulation of tumor cell mRNA expression in ectopic pancreatic cancer in mice. PLoS One 2021; 16:e0261633. [PMID: 34936674 PMCID: PMC8694476 DOI: 10.1371/journal.pone.0261633] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/06/2021] [Indexed: 11/19/2022] Open
Abstract
We investigated the contributions of platelet microRNAs (miRNAs) to the rate of growth and regulation of gene expression in primary ectopic tumors using mouse models. We previously identified an inhibitory role for platelets in solid tumor growth, mediated by tumor infiltration of platelet microvesicles (microparticles) which are enriched in platelet-derived miRNAs. To investigate the specific roles of platelet miRNAs in tumor growth models, we implanted pancreatic ductal adenocarcinoma cells as a bolus into mice with megakaryocyte-/platelet-specific depletion of mature miRNAs. We observed an ~50% increase in the rate of growth of ectopic primary tumors in these mice compared to controls including at early stages, associated with reduced apoptosis in the tumors, in particular in tumor cells associated with platelet microvesicles-which were depleted of platelet-enriched miRNAs-demonstrating a specific role for platelet miRNAs in modulation of primary tumor growth. Differential expression RNA sequencing of tumor cells isolated from advanced primary tumors revealed a broad cohort of mRNAs modulated in the tumor cells as a function of host platelet miRNAs. Altered genes comprised 548 up-regulated transcripts and 43 down-regulated transcripts, mostly mRNAs altogether spanning a variety of growth signaling pathways-notably pathways related to epithelial-mesenchymal transition-in tumor cells from platelet miRNA-deleted mice compared with those from control mice. Tumors in platelet miRNA-depleted mice showed more sarcomatoid growth and more advanced tumor grade, indicating roles for host platelet miRNAs in tumor plasticity. We further validated increased protein expression of selected genes associated with increased cognate mRNAs in the tumors due to platelet miRNA depletion in the host animals, providing proof of principle of widespread effects of platelet miRNAs on tumor cell functional gene expression in primary tumors in vivo. Together, these data demonstrate that platelet-derived miRNAs modulate solid tumor growth in vivo by broad-spectrum restructuring of the tumor cell transcriptome.
Collapse
Affiliation(s)
- Jeremy G. T. Wurtzel
- Division of Hematology, Department of Medicine, Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Sophia Lazar
- Division of Hematology, Department of Medicine, Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Sonali Sikder
- Molecular Therapeutics Program and The Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, United States of America
| | - Kathy Q. Cai
- Cancer Biology Program and Histopathology Facility, Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, PA, United States of America
| | - Igor Astsaturov
- Molecular Therapeutics Program and The Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Philadelphia, PA, United States of America
| | - Andrew S. Weyrich
- Molecular Medicine Program, Pathology Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States of America
| | - Jesse W. Rowley
- Molecular Medicine Program, Pulmonary Division, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States of America
| | - Lawrence E. Goldfinger
- Division of Hematology, Department of Medicine, Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States of America
| |
Collapse
|
14
|
Wu J, Piao Y, Liu Q, Yang X. Platelet-rich plasma-derived extracellular vesicles: A superior alternative in regenerative medicine? Cell Prolif 2021; 54:e13123. [PMID: 34609779 PMCID: PMC8666280 DOI: 10.1111/cpr.13123] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/13/2021] [Accepted: 08/31/2021] [Indexed: 02/06/2023] Open
Abstract
Platelet-rich plasma (PRP), due to its promising therapeutic properties, has been used in regenerative medicine for more than 30 years and numerous encouraging outcomes have been obtained. Currently, by benefiting from new insights into PRP mechanisms and the excellent performance of extracellular vesicles (EVs) in the field of tissue repair and regeneration, studies have found that a large number of EVs released from activated platelets also participate in the regulation of tissue repair. A growing number of preclinical studies are exploring the functions of PRP-derived EVs (PRP-EVs), especially in tissue regeneration. Here, we summarize the latest progress in PRP-EVs as a superior alternative cell-free therapeutic strategy in regenerative medicine, clarify their underlying molecular mechanisms, and discuss the advantages and limitations of the upcoming clinical applications. This review highlights the potential of PRP-EVs to replace the application of PRP or even become a superior alternative in regenerative medicine.
Collapse
Affiliation(s)
- Jiuping Wu
- Department of Orthopaedics, The Second Hospital, Jilin University, Changchun, China
| | - Yingxin Piao
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Qinyi Liu
- Department of Orthopaedics, The Second Hospital, Jilin University, Changchun, China
| | - Xiaoyu Yang
- Department of Orthopaedics, The Second Hospital, Jilin University, Changchun, China
| |
Collapse
|
15
|
Božič D, Vozel D, Hočevar M, Jeran M, Jan Z, Pajnič M, Pađen L, Iglič A, Battelino S, Kralj-Iglič V. Enrichment of plasma in platelets and extracellular vesicles by the counterflow to erythrocyte settling. Platelets 2021; 33:592-602. [PMID: 34384320 DOI: 10.1080/09537104.2021.1961716] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
In order to prepare optimal platelet and extracellular vesicle (EV)-rich plasma for the treatment of chronic temporal bone inflammation, we studied effects of centrifugation parameters on redistribution of blood constituents in blood samples of 23 patients and 20 volunteers with no record of disease. Concentrations of blood cells and EVs were measured by flow cytometry. Sample content was inspected by scanning electron microscopy. A mathematical model was constructed to interpret the experimental results. The observed enrichment of plasma in platelets and EVs after a single spin of blood depended on the erythrocyte sedimentation rate, thereby indicating the presence of a flow of plasma that carried platelets and EVs in the direction opposite to settling of erythrocytes. Prolonged handling time correlated with the decrease of concentration of platelets and larger EVs in platelet and EV-rich plasma (PVRP), R = -0.538, p = 0.003, indicating cell fragmentation during the processing of samples. In further centrifugation of the obtained plasma, platelet and EV enrichment depended on the average distance of the sample from the centrifuge rotor axis. Based on the agreement of the model predictions with observations, we propose the centrifugation protocol optimal for platelet and EV enrichment and recovery in an individual sample, adjusted to the dimensions of the centrifuge rotor, volume of blood and erythrocyte sedimentation rate.[Figure: see text].
Collapse
Affiliation(s)
- Darja Božič
- University of Ljubljana, Faculty of Health Sciences, Laboratory of Clinical Biophysics, Ljubljana, Slovenia.,University of Ljubljana, Faculty of Electrical Engineering, Laboratory of Physics, Ljubljana, Slovenia
| | - Domen Vozel
- University Medical Centre Ljubljana, Department of Otorhinolaryngology and Cervicofacial Surgery, Ljubljana, Slovenia.,University of Ljubljana, Faculty of Medicine, Department of Otorhinolaryngology, Ljubljana, Slovenia
| | - Matej Hočevar
- Department of Physics and Chemistry of Materials, Institute of Metals and Technology, Ljubljana, Slovenia
| | - Marko Jeran
- University of Ljubljana, Faculty of Health Sciences, Laboratory of Clinical Biophysics, Ljubljana, Slovenia.,University of Ljubljana, Faculty of Electrical Engineering, Laboratory of Physics, Ljubljana, Slovenia
| | - Zala Jan
- University of Ljubljana, Faculty of Health Sciences, Laboratory of Clinical Biophysics, Ljubljana, Slovenia
| | - Manca Pajnič
- University of Ljubljana, Faculty of Health Sciences, Laboratory of Clinical Biophysics, Ljubljana, Slovenia
| | - Ljubiša Pađen
- University of Ljubljana, Faculty of Health Sciences, Laboratory of Clinical Biophysics, Ljubljana, Slovenia
| | - Aleš Iglič
- University of Ljubljana, Faculty of Electrical Engineering, Laboratory of Physics, Ljubljana, Slovenia.,University of Ljubljana, Faculty of Medicine, Chair of Orthopedics, Laboratory of Clinical Biophysics, Ljubljana, Slovenia
| | - Saba Battelino
- University Medical Centre Ljubljana, Department of Otorhinolaryngology and Cervicofacial Surgery, Ljubljana, Slovenia.,University of Ljubljana, Faculty of Medicine, Department of Otorhinolaryngology, Ljubljana, Slovenia
| | - Veronika Kralj-Iglič
- University of Ljubljana, Faculty of Health Sciences, Laboratory of Clinical Biophysics, Ljubljana, Slovenia
| |
Collapse
|
16
|
The Molecular Aspects of Disturbed Platelet Activation through ADP/P2Y 12 Pathway in Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms22126572. [PMID: 34207429 PMCID: PMC8234174 DOI: 10.3390/ijms22126572] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 12/22/2022] Open
Abstract
Epidemiological studies confirm a high risk of ischemic events in secondary-progressive multiple sclerosis (SP MS) patients, directly associated with an increased level of pro-thrombotic activity of platelets. Our work aimed to verify potential molecular abnormalities of the platelet P2Y12 receptor expression and functionality as a cause of an increased risk of thromboembolism observed in the course of MS. We have demonstrated an enhanced platelet reactivity in response to adenosine diphosphate (ADP) in SP MS relative to controls. We have also shown an increased mRNA expression for the P2RY12 gene in both platelets and megakaryocytes, as well as enhanced density of these receptors on the platelet surface. We postulate that one of the reasons for the elevated risk of ischemic events observed in MS may be a genetically or phenotypically reinforced expression of the platelet P2Y12 receptor. In order to analyze the effect of the PAR1 (protease activated receptor type 1) signaling pathway on the expression level of P2Y12, we also analyzed the correlation parameters between P2Y12 expression and the markers of platelet activation in MS induced by selective PAR1 agonist (thrombin receptor activating peptide-6, TRAP-6). Identifying the molecular base responsible for the enlarged pro-thrombotic activity of platelets in SP MS could contribute to the implementation of prevention and targeted treatment, reducing the development of cardiovascular disorders in the course of the disease.
Collapse
|
17
|
Zahran AM, El-Badawy O, Ali WA, Mahran ZG, Mahran EEMO, Rayan A. Circulating microparticles and activated platelets as novel prognostic biomarkers in COVID-19; relation to cancer. PLoS One 2021; 16:e0246806. [PMID: 33617530 PMCID: PMC7899358 DOI: 10.1371/journal.pone.0246806] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/26/2021] [Indexed: 01/08/2023] Open
Abstract
Background and aim The study aimed to determine whether the MPs levels and platelet activation are affected by the COVID-19 infection in both malignant and non-malignant patients compared to healthy individuals and define their contribution to the COVID-19 associated coagulopathy and the relation of these MPs to other hematologic parameters. Patients and methods We recruited 23 malignant patients with reverse transcription polymerase chain reaction (RT-PCR) positive COVID-19, also, 19 COVID-19 non-malignant patients, and 20 healthy volunteers were also enrolled for comparison. Blood samples were collected from patients and healthy donors into 5 mL vacutainer tube containing 3.5% buffered sodium citrate solution for measurement of total microparticles (TMPs), platelet microparticles (PMPs), endothelial microparticles (EMPs), CD62 activated platelets, and CD41 platelet marker. Results COVID-19 malignant patients had significantly lower hemoglobin and platelets compared to COVID non-malignant ones, while they had significantly higher C-reactive protein, LDH, AST, Albunim, creatinine, and prognostic index (PI) compared to COVID-19 non-malignant patients. significant accumulations of TMPs, PMPs, EMPs, and activated platelets in COVID-19 affected patients compared to healthy controls. TMPs, and EMPs were significantly accumulated in COVID-19 malignant compared to COVID-19 non-malignant patients with no significant difference in PMPs between both. Conclusion Circulating MPs and activated platelets may be promising novel prognostic biomarkers capable of identifying potentially severe COVID-19 patients who require immediate care especially in cancer patients.
Collapse
Affiliation(s)
- Asmaa M. Zahran
- Department of Clinical Pathology, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Omnia El-Badawy
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Wageeh A. Ali
- Diagnostic and Interventional Radiology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Zainab Gaber Mahran
- Department of Tropical Medicine and Gastroenterology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Essam Eldeen M. O. Mahran
- Department of Tropical Medicine and Gastroenterology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Amal Rayan
- Clinical Oncology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
- * E-mail: ,
| |
Collapse
|
18
|
Alfeky M, Abdelmaksoud M, Abdelmaksoud S, Abdelsamee H, Ezzelregal H. Platelets derived microparticles in COVID-19: Correlation to inflammatory and coagulation State. JOURNAL OF APPLIED HEMATOLOGY 2021. [DOI: 10.4103/joah.joah_102_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
19
|
Li SS, Gao S, Chen Y, Bao H, Li ZT, Yao QP, Liu JT, Wang Y, Qi YX. Platelet-derived microvesicles induce calcium oscillations and promote VSMC migration via TRPV4. Theranostics 2021; 11:2410-2423. [PMID: 33500733 PMCID: PMC7797689 DOI: 10.7150/thno.47182] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: Abnormal migration of vascular smooth muscle cells (VSMCs) from the media to the interior is a critical process during the intimal restenosis caused by vascular injury. Here, we determined the role of platelet-derived microvesicles (PMVs) released by activated platelets in VSMC migration. Methods: A percutaneous transluminal angioplasty balloon dilatation catheter was used to establish vascular intimal injury. Collagen I was used to activate PMVs, mimicking collagen exposure during intimal injury. To determine the effects of PMVs on VSMC migration in vitro, scratch wound healing assays were performed. Fluorescence resonance energy transfer was used to detect variations of calcium dynamics in VSMCs. Results: Morphological results showed that neointimal hyperplasia was markedly increased after balloon injury of the carotid artery in rats, and the main component was VSMCs. PMVs significantly promoted single cell migration and wound closure in vitro. Fluorescence resonance energy transfer revealed that PMVs induced temporal and dynamic calcium oscillations in the cytoplasms of VSMCs. The influx of extracellular calcium, but not calcium from intracellular stores, was involved in the process described above. The channel antagonist GSK219 and specific siRNA revealed that a membrane calcium channel, transient receptor potential vanilloid 4 (TRPV4), participated in the calcium oscillations and VSMC migration induced by PMVs. Conclusions: TRPV4 participated in the calcium oscillations and VSMC migration induced by PMVs. PMVs and the related molecules might be novel therapeutic targets for vascular remodeling during vascular injury.
Collapse
Affiliation(s)
- Shan-Shan Li
- Institute of Mechanobiology& Medical Engineering, School of Life Sciences &Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, China
| | - Shuang Gao
- Institute of Mechanobiology& Medical Engineering, School of Life Sciences &Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Chen
- Institute of Mechanobiology& Medical Engineering, School of Life Sciences &Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Han Bao
- Institute of Mechanobiology& Medical Engineering, School of Life Sciences &Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zi-Tong Li
- Institute of Mechanobiology& Medical Engineering, School of Life Sciences &Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Qing-Ping Yao
- Institute of Mechanobiology& Medical Engineering, School of Life Sciences &Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ji-Ting Liu
- Institute of Mechanobiology& Medical Engineering, School of Life Sciences &Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yingxiao Wang
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, San Diego, United States
| | - Ying-Xin Qi
- Institute of Mechanobiology& Medical Engineering, School of Life Sciences &Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100083, China
| |
Collapse
|
20
|
Kyselova A, Elgheznawy A, Wittig I, Heidler J, Mann AW, Ruf W, Fleming I, Randriamboavonjy V. Platelet-derived calpain cleaves the endothelial protease-activated receptor 1 to induce vascular inflammation in diabetes. Basic Res Cardiol 2020; 115:75. [PMID: 33258989 PMCID: PMC7716944 DOI: 10.1007/s00395-020-00833-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/11/2020] [Indexed: 12/30/2022]
Abstract
Diabetes mellitus is a major risk factor for cardiovascular disease. Platelets from diabetic patients are hyperreactive and release microparticles that carry activated cysteine proteases or calpains. Whether platelet-derived calpains contribute to the development of vascular complications in diabetes is unknown. Here we report that platelet-derived calpain1 (CAPN1) cleaves the protease-activated receptor 1 (PAR-1) on the surface of endothelial cells, which then initiates a signaling cascade that includes the activation of the tumor necrosis factor (TNF)-α converting enzyme (TACE). The latter elicits the shedding of the endothelial protein C receptor and the generation of TNF-α, which in turn, induces intracellular adhesion molecule (ICAM)-1 expression to promote monocyte adhesion. All of the effects of CAPN1 were mimicked by platelet-derived microparticles from diabetic patients or from wild-type mice but not from CAPN1−/− mice, and were not observed in PAR-1-deficient endothelial cells. Importantly, aortae from diabetic mice expressed less PAR-1 but more ICAM-1 than non-diabetic mice, effects that were prevented by treating diabetic mice with a calpain inhibitor as well as by the platelet specific deletion of CAPN1. Thus, platelet-derived CAPN1 contributes to the initiation of the sterile vascular inflammation associated with diabetes via the cleavage of PAR-1 and the release of TNF-α from the endothelial cell surface.
Collapse
Affiliation(s)
- Anastasia Kyselova
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Amro Elgheznawy
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany
| | - Ilka Wittig
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Functional Proteomics, SFB 815 Core Unit, Goethe University, Frankfurt am Main, Germany
| | - Juliana Heidler
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Functional Proteomics, SFB 815 Core Unit, Goethe University, Frankfurt am Main, Germany
| | | | - Wolfram Ruf
- German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Center for Thrombosis and Hemostasis, University Medical Center, Mainz, Germany.,Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany.,German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Voahanginirina Randriamboavonjy
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany. .,German Center for Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.
| |
Collapse
|
21
|
Sheikh Hosseini M, Parhizkar Roudsari P, Gilany K, Goodarzi P, Payab M, Tayanloo-Beik A, Larijani B, Arjmand B. Cellular Dust as a Novel Hope for Regenerative Cancer Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1288:139-160. [DOI: 10.1007/5584_2020_537] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
Taus F, Meneguzzi A, Castelli M, Minuz P. Platelet-Derived Extracellular Vesicles as Target of Antiplatelet Agents. What Is the Evidence? Front Pharmacol 2019; 10:1256. [PMID: 31780927 PMCID: PMC6857039 DOI: 10.3389/fphar.2019.01256] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022] Open
Abstract
Platelet-derived large extracellular vesicles (often referred to as microparticles in the field of cardiovascular disease) have been identified as effector in the atherothrombotic process, therefore representing a target of pharmacological intervention of potential interest. Despite that, limited evidence is so far available concerning the effects of antiplatelet agents on the release of platelet-derived extracellular vesicles. In the present narrative review, the mechanisms leading to vesiculation in platelets and the pathophysiological processes implicated will be discussed. This will be followed by a summary of the present evidence concerning the effects of antiplatelet agents under experimental conditions and in clinical settings.
Collapse
Affiliation(s)
- Francesco Taus
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| | - Alessandra Meneguzzi
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| | - Marco Castelli
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| | - Pietro Minuz
- Department of Medicine, Section of Internal Medicine C, University of Verona, Verona, Italy
| |
Collapse
|
23
|
Smith JD, Narayanan P, Li N. Biomarkers of platelet dysfunction in non-clinical safety studies and humans. CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2019.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
24
|
Żmigrodzka M, Witkowska-Piłaszewicz O, Rzepecka A, Cywińska A, Jagielski D, Winnicka A. Extracellular Vesicles in the Blood of Dogs with Cancer-A Preliminary Study. Animals (Basel) 2019; 9:ani9080575. [PMID: 31430895 PMCID: PMC6720862 DOI: 10.3390/ani9080575] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/12/2019] [Accepted: 08/14/2019] [Indexed: 12/25/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous population of submicron-sized structures released during the activation, proliferation, or apoptosis of various types of cells. Due to their size, their role in cell-to-cell communication in cancer is currently being discussed. In blood, the most abundant population of EVs is platelet-derived EVs (PEVs). The aim of this study was to estimate the absolute number and the origin of EVs in the blood of healthy dogs and of dogs with various types of cancer. The EV absolute number and cellular origin were examined by flow cytometry technique. EVs were classified on the basis of surface annexin V expression (phosphatidylserine PS+) and co-expression of specific cellular markers (CD61, CD45, CD3, CD21). The number of PEVs was significantly higher in dogs with cancer (median: 409/µL, range: 42-2748/µL vs. median: 170/µL, range: 101-449/µL in controls). The numbers of EVs derived from leukocytes (control median: 86/µL, range: 40-240/µL; cancer median: 443/µL, range: 44-3 352/µL) and T cells (control median: 5/µL, range: 2-66/µL; cancer median: 108/µL, range: 3-1735/µL) were higher in dogs with neoplasia compared to healthy controls. The estimation of PEV and leukocyte-derived EV counts may provide a useful biological marker in dogs with cancer.
Collapse
Affiliation(s)
- Magdalena Żmigrodzka
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, 02-787 Warsaw, Poland.
| | - Olga Witkowska-Piłaszewicz
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, 02-787 Warsaw, Poland
| | - Alicja Rzepecka
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, 02-787 Warsaw, Poland
| | - Anna Cywińska
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, 02-787 Warsaw, Poland
| | - Dariusz Jagielski
- Veterinary Clinic BIALOBRZESKA, Częstochowska 20, 02-344 Warsaw, Poland
| | - Anna Winnicka
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, 02-787 Warsaw, Poland
| |
Collapse
|
25
|
Guo J, Feng C, Zhang B, Zhang S, Shen X, Zhu J, Zhao XX. Extraction and identification of platelet‑derived microparticles. Mol Med Rep 2019; 20:2916-2921. [PMID: 31322221 DOI: 10.3892/mmr.2019.10484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 06/07/2019] [Indexed: 11/06/2022] Open
Abstract
Microparticles are carriers of signals for intracellular signal transduction. These carriers include proteins, mRNAs, microRNAs and other bioactive substances. Platelets are a major source of circulating microparticles, and microparticles are closely associated with the development of certain cardiovascular diseases. In the present study, a method for separating, extracting and identifying platelet‑derived microparticles was developed and differences in the expression of surface proteins on microparticles harvested from platelets stimulated by vortexing or treatment with thrombin was investigated. The counts, composition, sizes and inner structures of microparticles were determined using flow cytometry and transmission electron microscopy. Additionally, it was demonstrated that platelets could be readily activated, and a large quantity of microparticles with varying complex compositions, structures and sizes were derived from activated platelets. High purity platelet‑derived microparticles were obtained by gradient centrifugation. However, the microparticles derived from platelets stimulated by thrombin treatment or vortexing differed significantly in the levels of CD63. The present study aimed to provide improved options for the extraction and identification of microparticles.
Collapse
Affiliation(s)
- Jun Guo
- Department of Geriatrics, Anhui Provincial Hospital, Hefei, Anhui 230000, P.R. China
| | - Can Feng
- Department of Cardiology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200433, P.R. China
| | - Bili Zhang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Shiyang Zhang
- Department of Geriatrics, Anhui Provincial Hospital, Hefei, Anhui 230000, P.R. China
| | - Xiaxian Shen
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Jiaqi Zhu
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Xian-Xian Zhao
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
26
|
Sluijter JPG, Davidson SM, Boulanger CM, Buzás EI, de Kleijn DPV, Engel FB, Giricz Z, Hausenloy DJ, Kishore R, Lecour S, Leor J, Madonna R, Perrino C, Prunier F, Sahoo S, Schiffelers RM, Schulz R, Van Laake LW, Ytrehus K, Ferdinandy P. Extracellular vesicles in diagnostics and therapy of the ischaemic heart: Position Paper from the Working Group on Cellular Biology of the Heart of the European Society of Cardiology. Cardiovasc Res 2019; 114:19-34. [PMID: 29106545 PMCID: PMC5852624 DOI: 10.1093/cvr/cvx211] [Citation(s) in RCA: 252] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/01/2017] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs)—particularly exosomes and microvesicles (MVs)—are attracting considerable interest in the cardiovascular field as the wide range of their functions is recognized. These capabilities include transporting regulatory molecules including different RNA species, lipids, and proteins through the extracellular space including blood and delivering these cargos to recipient cells to modify cellular activity. EVs powerfully stimulate angiogenesis, and can protect the heart against myocardial infarction. They also appear to mediate some of the paracrine effects of cells, and have therefore been proposed as a potential alternative to cell-based regenerative therapies. Moreover, EVs of different sources may be useful biomarkers of cardiovascular disease identities. However, the methods used for the detection and isolation of EVs have several limitations and vary widely between studies, leading to uncertainties regarding the exact population of EVs studied and how to interpret the data. The number of publications in the exosome and MV field has been increasing exponentially in recent years and, therefore, in this ESC Working Group Position Paper, the overall objective is to provide a set of recommendations for the analysis and translational application of EVs focussing on the diagnosis and therapy of the ischaemic heart. This should help to ensure that the data from emerging studies are robust and repeatable, and optimize the pathway towards the diagnostic and therapeutic use of EVs in clinical studies for patient benefit.
Collapse
Affiliation(s)
- Joost Petrus Gerardus Sluijter
- Experimental Cardiology Laboratory, UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, University Utrecht, 3508GA Utrecht, The Netherlands
| | | | | | - Edit Iren Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary.,MTA-SE Immunoproteogenomics Research Group, Budapest, Hungary
| | - Dominique Paschalis Victor de Kleijn
- Department of Vascular Surgery, UMC Utrecht, Utrecht University, Utrecht, the Netherlands.,Netherlands Heart Institute, Utrecht, the Netherlands
| | - Felix Benedikt Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Derek J Hausenloy
- Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857.,National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609.,Yong Loo Lin School of Medicine, National University Singapore, 1E Kent Ridge Road, Singapore 119228.,The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK.,The National Institute of Health Research University College London Hospitals Biomedical Research Centre, Research & Development, Maple House 1st floor, 149 Tottenham Court Road, London W1T 7DN, UK.,Department of Cardiology, Barts Heart Centre, St Bartholomew's Hospital, W Smithfield, London EC1A 7BE, UK
| | - Raj Kishore
- Department of Pharmacology, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Sandrine Lecour
- Hatter Institute for Cardiovascular Research in Africa and Lionel Opie Preclinical Imaging Core Facility, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Jonathan Leor
- Neufeld Cardiac Research Institute, Sackler Faculty of Medicine, Tel-Aviv University, Tel Hashomer, Israel; Tamman Cardiovascular Research Institute, Heart Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Rosalinda Madonna
- Center of Aging Science and Regenerative Medicine, CESI-Met and Institute of Cardiology, "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy.,Department of Internal Medicine, University of Texas Medical School in Houston, TX, USA.,Texas Heart Institute, Houston, TX, USA
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Fabrice Prunier
- Institut Mitovasc, CHU d'Angers, Université d'Angers, Angers, France
| | - Susmita Sahoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ray Michel Schiffelers
- Laboratory Clinical Chemistry and Hematology Division, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University of Giessen, Aulweg 129, 35392, Giessen, Germany
| | - Linda Wilhelmina Van Laake
- Division Heart and Lungs, and Hubrecht Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kirsti Ytrehus
- Cardiovascular Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest 1089, Hungary and.,Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
27
|
Balaphas A, Meyer J, Sadoul K, Fontana P, Morel P, Gonelle-Gispert C, Bühler LH. Platelets and Platelet-Derived Extracellular Vesicles in Liver Physiology and Disease. Hepatol Commun 2019; 3:855-866. [PMID: 31304449 PMCID: PMC6601322 DOI: 10.1002/hep4.1358] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/12/2019] [Indexed: 02/06/2023] Open
Abstract
Beyond their role in hemostasis, platelets are proposed as key mediators of several physiological and pathophysiological processes of the liver, such as liver regeneration, toxic or viral acute liver injury, liver fibrosis, and carcinogenesis. The effects of platelets on the liver involve interactions with sinusoidal endothelial cells and the release of platelet‐contained molecules following platelet activation. Platelets are the major source of circulating extracellular vesicles, which are suggested to play key roles in platelet interactions with endothelial cells in several clinical disorders. In the present review, we discuss the implications of platelet‐derived extracellular vesicles in physiological and pathophysiological processes of the liver.
Collapse
Affiliation(s)
- Alexandre Balaphas
- Division of Digestive Surgery Geneva University Hospitals Geneva Switzerland.,Surgical Research Unit Geneva University Hospitals Geneva Switzerland.,Geneva Medical School University of Geneva Geneva Switzerland
| | - Jeremy Meyer
- Division of Digestive Surgery Geneva University Hospitals Geneva Switzerland.,Surgical Research Unit Geneva University Hospitals Geneva Switzerland.,Geneva Medical School University of Geneva Geneva Switzerland
| | - Karin Sadoul
- Regulation and Pharmacology of the Cytoskeleton, Institute for Advanced Biosciences Université Grenoble Alpes Grenoble France
| | - Pierre Fontana
- Division of Angiology and Hemostasis Geneva University Hospitals Geneva Switzerland.,Geneva Platelet Group University of Geneva Geneva Switzerland
| | - Philippe Morel
- Division of Digestive Surgery Geneva University Hospitals Geneva Switzerland.,Surgical Research Unit Geneva University Hospitals Geneva Switzerland.,Geneva Medical School University of Geneva Geneva Switzerland
| | - Carmen Gonelle-Gispert
- Surgical Research Unit Geneva University Hospitals Geneva Switzerland.,Geneva Medical School University of Geneva Geneva Switzerland
| | - Leo H Bühler
- Division of Digestive Surgery Geneva University Hospitals Geneva Switzerland.,Surgical Research Unit Geneva University Hospitals Geneva Switzerland.,Geneva Medical School University of Geneva Geneva Switzerland
| |
Collapse
|
28
|
Rossaint J, Margraf A, Zarbock A. Role of Platelets in Leukocyte Recruitment and Resolution of Inflammation. Front Immunol 2018; 9:2712. [PMID: 30515177 PMCID: PMC6255980 DOI: 10.3389/fimmu.2018.02712] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/02/2018] [Indexed: 12/30/2022] Open
Abstract
Platelets are most often recognized for their crucial role in the control of acute hemorrhage. However, current research has greatly expanded the appreciation of platelets beyond their contribution to primary hemostasis, indicating that platelets also actively participate in leukocyte recruitment and the regulation of the host defense in response to exogenous pathogens and sterile injury. Early recruitment of leukocytes, especially neutrophils, is the evolutionary stronghold of the innate immune response to successfully control exogenous infections. Platelets have been shown to physically interact with different leukocyte subsets during inflammatory processes. This interaction holds far-reaching implications for the leukocyte recruitment into peripheral tissues as well as the regulation of leukocyte cell autonomous functions, including the formation and liberation of neutrophil extracellular traps. These functions critically depend on the interaction of platelets with leukocytes. The host immune response and leukocyte recruitment must be tightly regulated to avoid excessive tissue and organ damage and to avoid chronification of inflammation. Thus, platelet-leukocyte interactions and the resulting leukocyte activation and recruitment also underlies tight regulation by several inherited feedback mechanisms to limit the extend of vascular inflammation and to protect the host from collateral damage caused by overshooting immune system activation. After the acute inflammatory phase has been overcome the host defense response must eventually be terminated to allow for resolution from inflammation and restoration of tissue and organ function. Besides their essential role for leukocyte recruitment and the initiation and propagation of vascular inflammation, platelets have lately also been implicated in the resolution process. Here, their contribution to phagocyte clearance, T cell recruitment and macrophage reprogramming is also of outmost importance. This review will focus on the role of platelets in leukocyte recruitment during the initiation of the host defense and we will also discuss the participation of platelets in the resolution process after acute inflammation.
Collapse
Affiliation(s)
- Jan Rossaint
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| | - Andreas Margraf
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany.,Interdisciplinary Centre for Clinical Research, University Hospital Münster, Münster, Germany
| | - Alexander Zarbock
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
29
|
Wilhelm EN, Mourot L, Rakobowchuk M. Exercise-Derived Microvesicles: A Review of the Literature. Sports Med 2018; 48:2025-2039. [PMID: 29868992 DOI: 10.1007/s40279-018-0943-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Initially suggested as simple cell debris, cell-derived microvesicles (MVs) have now gained acceptance as recognized players in cellular communication and physiology. Shed by most, and perhaps all, human cells, these tiny lipid-membrane vesicles carry bioactive agents, such as proteins, lipids and microRNA from their cell source, and are produced under orchestrated events in response to a myriad of stimuli. Physical exercise introduces systemic physiological challenges capable of acutely disrupting cell homeostasis and stimulating the release of MVs into the circulation. The novel and promising field of exercise-derived MVs is expanding quickly, and the following work provides a review of the influence of exercise on circulating MVs, considering both acute and chronic aspects of exercise and training. Potential effects of the MV response to exercise are highlighted and future directions suggested as exercise and sports sciences extend the realm of extracellular vesicles.
Collapse
Affiliation(s)
- Eurico N Wilhelm
- School of Physical Education, UFPel, Rua Luís de Camões, 625, Três Vendas, Pelotas, RS, 96055-630, Brazil.
| | - Laurent Mourot
- EA3920 Prognostic Factors and Regulatory Factors of Cardiac and Vascular Pathologies, (Exercise Performance Health Innovation-EPHI), University of Bourgogne Franche-Comté, 25000, Besançon, France.,Tomsk Polytechnic University, Tomsk, Russia
| | - Mark Rakobowchuk
- Department of Biological Sciences, Faculty of Science, Thompson Rivers University, Kamloops, BC, Canada
| |
Collapse
|
30
|
Iba T, Ogura H. Role of extracellular vesicles in the development of sepsis-induced coagulopathy. J Intensive Care 2018; 6:68. [PMID: 30377532 PMCID: PMC6194680 DOI: 10.1186/s40560-018-0340-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022] Open
Abstract
Background The advances of research on extracellular vesicles (EVs) are of particular interest to the clinicians as well as the researchers who are studying coagulation disorder in sepsis. Here, we intend to update the latest knowledge and currently unsolved problems that should be addressed. Main body Secreted membrane-enclosed vesicles including apoptotic bodies, exosomes, ectosomes, microvesicles, and microparticles are generically called EVs. Though the basic structure of these vesicles is the same, i.e., originating from the plasma membrane, their characteristics differ significantly depending on their surface structures and interior components. Numerous studies have shown elevated levels of circulating EVs that exhibit proinflammatory and procoagulant properties during sepsis. These EVs are known to play important roles in the development of coagulation disorder and organ dysfunction in sepsis. Coagulation disorder in sepsis is characterized by activated coagulation, disrupted anticoagulant systems, and imbalanced fibrinolytic systems. These processes collaborate with one another and contribute to the development of disseminated intravascular coagulation (DIC), with devastating consequences. As part of this pathogenesis, the membrane-exposed tissue factor, phosphatidylserine and bioactive substances contained within the vesicles, such as histones, nucleosomes, and high-mobility group box 1, contribute to the development of DIC. EVs not only upregulate the procoagulant systems by themselves, but they also disseminate prothrombotic activities by transferring their procoagulant properties to distant target cells. Though the basic concept behind the role of procoagulant properties, EVs in the development of sepsis-induced coagulopathy has started to be unveiled, knowledge of the actual status is far from satisfactory, mainly because of the lack of standardized assay procedures. Recent advances and current problems that remain to be resolved are introduced in this review. Conclusion The recent studies succeeded to elucidate the important roles of EVs in the progress of coagulation disorder in sepsis. However, further harmonization in terminology, methodology, and evaluation methods is required for future studies.
Collapse
Affiliation(s)
- Toshiaki Iba
- 1Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo Bunkyo-ku, Tokyo, 113-8421 Japan
| | - Hiroshi Ogura
- 2Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
31
|
Microparticles from β-thalassaemia/HbE patients induce endothelial cell dysfunction. Sci Rep 2018; 8:13033. [PMID: 30158562 PMCID: PMC6115342 DOI: 10.1038/s41598-018-31386-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/16/2018] [Indexed: 12/18/2022] Open
Abstract
Thromboembolic complication occurs frequently in β-thalassaemia/HbE patients, particularly in splenectomised patients. Endothelial cells play an important role in thrombosis. There is strong evidence of endothelial cell activation and dysfunction in β-thalassaemia. Microparticles (MPs) are associated with thrombosis and endothelial cell dysfunction in many diseases including β-thalassaemia. However, the effect of thalassaemic-MPs on endothelial cells mediating thrombus formation has not been elucidated. In this study, the effects of circulating MPs from β-thalassaemia/HbE patients on endothelial cell functions were investigated. The results showed that MPs directly induce tissue factor, interleukin (IL)-6, IL-8, intracellular adhesion molecule-1, vascular cell adhesion molecule-1 and E-selectin expression in human umbilical vein endothelial cells (HUVECs). Notably, the levels of these endothelial cell activation markers were significantly increased in HUVECs treated with MPs obtained from splenectomised β-thalassaemia/HbE patients when compared to MPs from non-splenectomised patients or normal subjects. The increased endothelial cell activation ultimately lead to increased monocyte-endothelial cell adhesion. THP-1 and HUVECs adhesion induced by MPs from normal subjects, non-splenectomised and splenectomised patients increased to 2.0 ± 0.4, 2.3 ± 0.4 and 3.8 ± 0.4 fold, respectively when compared to untreated cells. This finding suggests that MPs play an important role on thrombosis and vascular dysfunction in β-thalassaemia/HbE disease, especially in splenectomised cases.
Collapse
|
32
|
Abstract
Platelet-derived microvesicles (pMVs) are small, heterogeneous vesicles released from platelet membranes as a result of activation. These microvesicles possess a wide range of properties, including prothrombotic, proatherogenic, proinflammatory, immunomodulatory, and even anticoagulant activity. The elevated release of these microvesicles has been observed in various metabolic, inflammatory, thrombotic, and vascular diseases, including ischemic heart disease, stroke, hypertension, diabetes, and connective tissue disease. Modulation of both pMV generation and the expression of their surface molecules may have beneficial clinical implications and could become a novel therapeutic target. However, mechanisms by which pharmacological agents can modify pMV formation are elusive. The purpose of this review is to discuss the effects of drugs routinely used in primary and secondary prevention of vascular disease on the release of pMV and expression of their surface procoagulant and proinflammatory molecules.
Collapse
Affiliation(s)
- Justyna Rosińska
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland.
| | - Maria Łukasik
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland
| |
Collapse
|
33
|
Xia L, Zeng Z, Tang WH. The Role of Platelet Microparticle Associated microRNAs in Cellular Crosstalk. Front Cardiovasc Med 2018; 5:29. [PMID: 29670887 PMCID: PMC5893844 DOI: 10.3389/fcvm.2018.00029] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/15/2018] [Indexed: 01/11/2023] Open
Abstract
Platelet is an anucleate cell containing abundant messenger RNAs and microRNAs (miRNAs), and their functional roles in hemostasis and inflammation remain elusive. Accumulating evidence has suggested that platelets can actively transfer RNAs to hepatocytes, vascular cells, macrophages, and tumor cells. The incorporated mRNAs are translated into proteins, and miRNAs were found to regulate the gene expression, resulting in the functional change of the recipient cells. This novel intercellular communication opens up a new avenue for the pathophysiological role of platelet in platelet-associated vascular diseases. Therefore, understanding the underlying mechanism and identification of the platelet miRNAs involved in this biological process would provide novel diagnostic and therapeutic targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Luoxing Xia
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Zhi Zeng
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Wai Ho Tang
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
34
|
|
35
|
Ay C, Pabinger I, Thaler J. Clinical significance of circulating microparticles for venous thrombo - embolism in cancer patients. Hamostaseologie 2017; 32:127-31. [DOI: 10.5482/ha-1164] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Accepted: 06/20/2011] [Indexed: 12/13/2022] Open
Abstract
SummaryCancer patients have a four-to seven-fold increased risk to develop a venous thromboembolic event. Accumulating evidence from experimental and clinical studies indicates that microparticles (MPs), small procoagulant membrane vesicles that are defined by size and a negatively charged phosphatidylserine rich surface, play an important role in the pathogenesis of cancer-related venous thromboembolism (VTE). However, the clinical significance of MPs as a predictive biomarker for VTE in cancer patients has not been fully elucidated yet. This might be due to unresolved methodological problems and a lack of data from large prospective clinical studies that investigate the role of MPs in cancer-related VTE.It is the aim of this review to give an overview on the most important characteristics of MPs and studies dealing with the role of MPs in cancer-related VTE. Also recent progresses, unresolved problems and future perspectives in this research field will be discussed. In the conclusion we will assess the clinical significance of MPs in cancer-related VTE.
Collapse
|
36
|
Schwarz V, Düsing P, Liman T, Werner C, Herm J, Bachelier K, Krüll M, Brechtel L, Jungehulsing GJ, Haverkamp W, Böhm M, Endres M, Haeusler KG, Laufs U. Marathon running increases circulating endothelial- and thrombocyte-derived microparticles. Eur J Prev Cardiol 2017; 25:317-324. [PMID: 29183152 DOI: 10.1177/2047487317744364] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background Acute vascular effects of high intensity physical activity are incompletely characterized. Circulating microparticles are cellular markers for vascular activation and damage. Methods Microparticles were analysed in 99 marathon runners (49 ± 6 years, 22% female) of the prospective Berlin Beat of Running study. Blood samples were taken within three days before, immediately after and within two days after the marathon run. Endothelial-derived microparticles were labelled with CD144, CD31 and CD62E, platelet-derived microparticles with CD62P and CD42b, leukocyte-derived microparticles with CD45 and monocyte-derived microparticles with CD14. Results Marathon running induced leukocytosis (5.9 ± 0.1 to 14.8 ± 0.3 109/l, p < 0.0001) and increased platelet counts (239 ± 4.6 to 281 ± 5.9 109/l, p < 0.0001) immediately after the marathon. Blood monocytes increased and lymphocytes decreased after the run ( p < 0.0001). Endothelial-derived microparticles were acutely increased ( p = 0.008) due to a 23% increase of apoptotic endothelial-derived microparticles ( p = 0.007) and returned to baseline within two days after the marathon. Thrombocyte-derived microparticles acutely increased by 38% accompanied by an increase in activated and apoptotic thrombocyte-derived microparticles ( p ≤ 0.0001) each. Both monocyte- and leukocyte-derived microparticles were decreased immediately after marathon run ( p < 0.0001) and remained below baseline until day 2. Troponin T increased from 12 to 32 ng/l ( p < 0.0001) immediately after the run and returned to baseline after two days. Conclusion Circulating apoptotic endothelial- and thrombocyte-derived microparticles increased after marathon running consistent with an acute pro-thrombotic and pro-inflammatory state. Exercise-induced vascular damage reflected by microparticles could indicate potential mechanisms of post-exertional cardiovascular complications. Further studies are warranted to investigate microparticles as markers to identify individuals prone to such complications.
Collapse
Affiliation(s)
- Viktoria Schwarz
- 1 Department for Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, Saarland University, Homburg/Saar, Germany
| | - Philip Düsing
- 1 Department for Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, Saarland University, Homburg/Saar, Germany
| | - Thomas Liman
- 2 Department of Neurology, Charité - Universitätsmedizin Berlin, Germany
| | - Christian Werner
- 1 Department for Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, Saarland University, Homburg/Saar, Germany
| | - Juliane Herm
- 2 Department of Neurology, Charité - Universitätsmedizin Berlin, Germany.,3 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Germany
| | - Katrin Bachelier
- 1 Department for Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, Saarland University, Homburg/Saar, Germany
| | - Matthias Krüll
- 4 SMS Sports Medicine Berlin, Medical Institute of the BMW BERLIN-MARATHON, Germany
| | | | | | - Wilhelm Haverkamp
- 7 Department of Cardiology, Charité - Universitätsmedizin Berlin Campus Virchow-Klinikum, Germany
| | - Michael Böhm
- 1 Department for Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, Saarland University, Homburg/Saar, Germany
| | - Matthias Endres
- 2 Department of Neurology, Charité - Universitätsmedizin Berlin, Germany.,3 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Germany.,8 German Center for Neurodegenerative Diseases (DZNE) & German Center for Cardiovascular Diseases (DZHK), Partner Site, Berlin, Germany
| | - Karl Georg Haeusler
- 2 Department of Neurology, Charité - Universitätsmedizin Berlin, Germany.,3 Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Germany
| | - Ulrich Laufs
- 9 Department of Cardiology, Universitätsklinikum Leipzig, Germany
| |
Collapse
|
37
|
Ball S, Nugent K. Microparticles in Hematological Malignancies: Role in Coagulopathy and Tumor Pathogenesis. Am J Med Sci 2017; 355:207-214. [PMID: 29549921 DOI: 10.1016/j.amjms.2017.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/09/2017] [Accepted: 11/25/2017] [Indexed: 12/12/2022]
Abstract
Microparticles (MP) are submicron vesicles released from various cells in response to activation, injury or apoptosis. They contain different structural and functional proteins and RNAs, which contribute to physiological intercellular "crosstalk" and to the pathogenesis of various diseases including cancer. In hematological malignancies, these MPs participate in the initiation and propagation of thrombosis through different pathways. They have a role in the angiogenesis, malignant cell survival and metastasis. MPs act as a mediator of resistance of leukemic cells to chemotherapy. The number of MPs is one of the prognostic factors following stem cell transplant, and studies have also found they contribute to the pathogenesis of graft versus host disease. MPs are being tested as therapeutic options in leukemias and graft versus host disease. Future studies should help us understand the interactions between MPs and cancer cells better, thereby opening new approaches for treatment of hematological malignancies.
Collapse
Affiliation(s)
- Somedeb Ball
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas.
| | - Kenneth Nugent
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas
| |
Collapse
|
38
|
Menter DG, Kanikarla-Marie P, Lam M, Davis JS, Kopetz S. Platelet microparticles: small payloads with profound effects on tumor growth. ACTA ACUST UNITED AC 2017; 1. [PMID: 31218277 DOI: 10.21037/ncri.2017.11.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- David G Menter
- Departments of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Preeti Kanikarla-Marie
- Departments of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Lam
- Departments of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer S Davis
- Departments of Epidemiology, MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Departments of Gastrointestinal Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
39
|
Fricke A, Ullrich PV, Cimniak AFV, Becherer C, Follo M, Heinz J, Scholber J, Herget GW, Hauschild O, Wittel UA, Stark GB, Bannasch H, Braig D, Eisenhardt SU. Levels of activated platelet-derived microvesicles in patients with soft tissue sarcoma correlate with an increased risk of venous thromboembolism. BMC Cancer 2017; 17:527. [PMID: 28784104 PMCID: PMC5547532 DOI: 10.1186/s12885-017-3515-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 07/31/2017] [Indexed: 12/21/2022] Open
Abstract
Background Microvesicles are small vesicles expressing specific antigens from their cells of origin. Elevated levels of microvesicles have been shown to be associated with coagulation disorders as well as with different types of malignancies. This study aims to evaluate a possible correlation of different microvesicle subpopulations with a positive history of venous thromboembolism (VTE) in patients with soft tissue sarcoma. Methods Annexin V - positive microvesicles, leukocyte (CD45-positive), platelet (CD61-positive), activated platelet (CD62P-, CD63-positive), endothelium-derived (CD62E-positive) and tissue-factor (CD142-positive) microvesicles were identified in the peripheral blood of patients with soft tissue sarcoma (n = 39) and healthy controls (n = 17) using fluorescence-activated cell sorting (FACS). Results Both the total amount of Annexin V-positive microvesicles and levels of endothelium-derived (CD62E-positive) microvesicles were shown to decrease significantly after tumor resection (n = 18, p = 0.0395 and p = 0.0109, respectively). Furthermore, the total amount of Annexin V – positive microvesicles as well as leukocyte (CD45-positive) and endothelium-derived (CD62E-positive) microvesicles were significantly higher in patients with grade 3 (G3) soft tissue sarcoma (n = 9) compared to healthy controls (n = 17) (p = 0.0304, p = 0.0254 and p = 0.0357, respectively). Moreover, patients with G3 soft tissue sarcoma (n = 9) presented higher levels of Annexin V-positive and endothelium-derived (CD62E-positive) microvesicles compared to patients with grade 2 (G2) soft tissue sarcoma (n = 8) (p = 0.0483 and p = 0.0045). Patients with grade 1 (G1) soft tissue sarcoma (n = 3) presented with significantly lower levels of platelet (CD61-positive) microvesicles than patients with G3 soft tissue sarcoma (n = 9) (p = 0.0150). In patients with a positive history of VTE (n = 11), significantly higher levels of activated platelet (CD62P- and CD63-positive) microvesicles (p = 0.0078 and p = 0.0450, respectively) were found compared to patients without a history of VTE (n = 28). Conclusion We found significantly higher levels of Annexin V-positive and endothelium-derived (CD62E-positive) microvesicles to be circulating in the peripheral blood of patients with G3 soft tissue sarcoma compared to patients with G2 soft tissue sarcoma. Furthermore, we showed that high counts of activated platelet-derived microvesicles correlate with the occurrence of VTE. Thus, the detection of these microvesicles might be an interesting new tool for early diagnosis of soft tissue sarcoma patients with increased risk for VTE, possibly facilitating VTE prevention by earlier use of thromboprophylaxis. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3515-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- A Fricke
- Department of Plastic and Hand Surgery, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - P V Ullrich
- Department of Plastic and Hand Surgery, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - A F V Cimniak
- Department of Plastic and Hand Surgery, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - C Becherer
- Department of Plastic and Hand Surgery, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - M Follo
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - J Heinz
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - J Scholber
- Department of Radiation Oncology, Medical Center - Faculty of Medicine, University of Freiburg, Robert-Koch-Straße 3, 79106, Freiburg, Germany
| | - G W Herget
- Department of Orthopedics and Traumatology, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - O Hauschild
- Department of Orthopedics and Traumatology, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - U A Wittel
- Department of General and Visceral Surgery, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - G B Stark
- Department of Plastic and Hand Surgery, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - H Bannasch
- Department of Plastic and Hand Surgery, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - D Braig
- Department of Plastic and Hand Surgery, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - S U Eisenhardt
- Department of Plastic and Hand Surgery, Medical Center - Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany.
| |
Collapse
|
40
|
Ponomareva AA, Nevzorova TA, Mordakhanova ER, Andrianova IA, Rauova L, Litvinov RI, Weisel JW. Intracellular origin and ultrastructure of platelet-derived microparticles. J Thromb Haemost 2017; 15:1655-1667. [PMID: 28561434 PMCID: PMC5657319 DOI: 10.1111/jth.13745] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Indexed: 12/18/2022]
Abstract
Essentials Platelet microparticles play a major role in pathologies, including hemostasis and thrombosis. Platelet microparticles have been analyzed and classified based on their ultrastructure. The structure and intracellular origin of microparticles depend on the cell-activating stimulus. Thrombin-treated platelets fall apart and form microparticles that contain cellular organelles. SUMMARY Background Platelet-derived microparticles comprise the major population of circulating blood microparticles that play an important role in hemostasis and thrombosis. Despite numerous studies on the (patho)physiological roles of platelet-derived microparticles, mechanisms of their formation and structural details remain largely unknown. Objectives Here we studied the formation, ultrastructure and composition of platelet-derived microparticles from isolated human platelets, either quiescent or stimulated with one of the following activators: arachidonic acid, ADP, collagen, thrombin or calcium ionophore A23187. Methods Using flow cytometry, transmission and scanning electron microscopy, we analyzed the intracellular origin, structural diversity and size distributions of the subcellular particles released from platelets. Results The structure, dimensions and intracellular origin of microparticles depend on the cell-activating stimulus. The main structural groups include a vesicle surrounded by one thin membrane or multivesicular structures. Thrombin, unlike other stimuli, induced formation of microparticles not only from the platelet plasma membrane and cytoplasm but also from intracellular structures. A fraction of these vesicular particles having an intracellular origin contained organelles, such as mitochondria, glycogen granules and vacuoles. The size of platelet-derived microparticles depended on the nature of the cell-activating stimulus. Conclusion The results obtained provide a structural basis for the qualitative differences of various platelet activators, for specific physiological and pathological effects of microparticles, and for development of advanced assays.
Collapse
Affiliation(s)
- A A Ponomareva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- Kazan Institute of Biochemistry and Biophysics, Russian Academy of Sciences, Kazan, Russia Federation
| | - T A Nevzorova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - E R Mordakhanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - I A Andrianova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - L Rauova
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - R I Litvinov
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - J W Weisel
- University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
41
|
Greven J, Pfeifer R, Zhi Q, Pape HC. Update on the role of endothelial cells in trauma. Eur J Trauma Emerg Surg 2017; 44:667-677. [PMID: 28674817 DOI: 10.1007/s00068-017-0812-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 06/21/2017] [Indexed: 12/23/2022]
Abstract
PURPOSE This review gives an overview of physiological processes, mainly regarding vascular endothelial cells and their important role in hemostasis, information processing, and communication during trauma. An insight is given into molecules and cells involved in the first innate immune response through to the behavior of endothelial cells in developing trauma. The goal of this review is to show the overlap of crucial factors related to the endothelium and the development of trauma. METHODS A systemic literature search was performed using Google scholar and PubMed. RESULTS The results of the literature search showed that the endothelium, especially the vascular endothelium, is involved in various cellular and subcellular pathways of activation, suppression, and transfer of information. A variety of molecules and cells are orchestrated, subsequently the endothelium gets in contact with a traumatizing event. CONCLUSION The endothelium is one of the first barriers that comes into contact with exo- and endogenous trauma-related signals and is a pivotal point in activating subsequent pathways and cascades by transfer of information.
Collapse
Affiliation(s)
- J Greven
- Department of Trauma and Reconstructive Surgery, University of Aachen Medical Center, Pauwelsstr 30, 52074, Aachen, Germany.
| | - R Pfeifer
- Department for Traumatology, University of Zürich Medical Center, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Q Zhi
- Department of Trauma and Reconstructive Surgery, University of Aachen Medical Center, Pauwelsstr 30, 52074, Aachen, Germany
| | - H C Pape
- Department for Traumatology, University of Zürich Medical Center, Rämistrasse 100, 8091, Zurich, Switzerland
| |
Collapse
|
42
|
Kim HJ, Choi EH, Lim YJ, Kil HR. The Usefulness of Platelet-derived Microparticle as Biomarker of Antiplatelet Therapy in Kawasaki Disease. J Korean Med Sci 2017; 32:1147-1153. [PMID: 28581272 PMCID: PMC5461319 DOI: 10.3346/jkms.2017.32.7.1147] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 04/09/2017] [Indexed: 11/20/2022] Open
Abstract
Little is known about platelet dynamics and the effect of antiplatelet therapy in Kawasaki disease (KD). This study sought to define platelet activation dynamics in KD patients by assaying platelet-derived microparticles (PDMPs). We measured plasma PDMPs levels in 46 patients with KD using an enzyme-linked immunosorbent assay (ELISA). Blood samples were collected before, at 2-5 days, and 9-15 days after intravenous immunoglobulin (IVIG) infusion, 2 months and 4-5 months after the onset of KD. We measured PDMP levels in 23 febrile and 10 afebrile control patients. In the acute phase of KD patients, PDMP levels increased significantly after IVIG treatment (12.04 ± 5.58 nmol before IVIG infusion vs. 19.81 ± 13.21 nmol at 2-5 days after IVIG infusion, P = 0.006). PDMP levels were negatively correlated with age and positively correlated with procalcitonin levels in the acute phase of KD. No significant difference was found in PDMP levels between KD patients with and without coronary artery lesion (CAL). Elevated PDMP levels after IVIG therapy significantly decreased below the pre-IVIG level in subacute phase (19.81 ± 13.21 nmol at 2-5 days after IVIG infusion vs. 8.33 ± 2.02 nmol at 9-15 days after IVIG infusion, P < 0.001), and PDMP levels stayed below the pre-IVIG level in the convalescent phase, during which antiplatelet therapy was given. However, PDMP levels rebounded after discontinuing aspirin in 17 patients. In conclusion, enhanced platelet activation was noted before treatment of KD and peaked immediately after IVIG treatment. Recurrent rising of PDMP levels was observed after discontinuing aspirin, although there were no significant differences between the PDMP levels at 2 months after the onset of KD and those at 4-5 months after the onset of the disease.
Collapse
Affiliation(s)
- Hyun Jung Kim
- Department of Pediatrics, Eulji University School of Medicine, Daejeon, Korea
| | - Eun Hye Choi
- Eulji Medi-Bio Research Institute, Eulji University School of Medicine, Daejeon, Korea
| | - Yeon Jung Lim
- Department of Pediatrics, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hong Ryang Kil
- Department of Pediatrics, Chungnam National University School of Medicine, Daejeon, Korea.
| |
Collapse
|
43
|
Kanikarla-Marie P, Lam M, Menter DG, Kopetz S. Platelets, circulating tumor cells, and the circulome. Cancer Metastasis Rev 2017; 36:235-248. [DOI: 10.1007/s10555-017-9681-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
44
|
Circulating microparticles are prognostic biomarkers in advanced non-small cell lung cancer patients. Oncotarget 2017; 8:75952-75967. [PMID: 29100283 PMCID: PMC5652677 DOI: 10.18632/oncotarget.18372] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 04/25/2017] [Indexed: 12/13/2022] Open
Abstract
We investigated whether circulating microparticles (MPs) could serve as prognostic biomarkers in non-small cell lung cancer (NSCLC) patients. We enrolled 25 control subjects and 136 NSCLC patients categorized into disease-progression (DP, n=42) and disease-control (DC, n=94) groups. Flow cytometric analysis showed that levels of four types of circulating microparticles (EDAc-MPs, EDAp-MPs, PDAc-MPs and PDAp-MPs) were higher in the study patients than the control subjects (P < 0.04). DP patients showed poor initially performance status and more non-adenocarcinomas than DC patients. DC patients showed more EGFR mutations and poorer performance to targeted therapy than DP patients (P < 0.01). Three months after therapy, the levels of all four types of circulating MPs were lower in DC than DP patients (P < 0.02), and were comparable to the levels in control subjects. In addition, the levels of circulating MPs after 3 months accurately predicted one-year prognostic outcomes (P < 0.05). This study showed that circulating MPs are valuable prognostic biomarkers in advanced NSCLC patients.
Collapse
|
45
|
Circulating soluble P-selectin must dimerize to promote inflammation and coagulation in mice. Blood 2017; 130:181-191. [PMID: 28515093 DOI: 10.1182/blood-2017-02-770479] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/14/2017] [Indexed: 11/20/2022] Open
Abstract
Leukocyte adhesion to P-selectin on activated platelets and endothelial cells induces shedding of the P-selectin ectodomain into the circulation. Plasma soluble P-selectin (sP-selectin) is elevated threefold to fourfold in patients with cardiovascular disease. Circulating sP-selectin is thought to trigger signaling in leukocytes that directly contributes to inflammation and thrombosis. However, sP-selectin likely circulates as a monomer, and in vitro studies suggest that sP-selectin must dimerize to induce signaling in leukocytes. To address this discrepancy, we expressed the entire ectodomain of mouse P-selectin as a monomer (sP-selectin) or as a disulfide-linked dimer fused to the Fc portion of mouse immunoglobulin G (sP-selectin-Fc). Dimeric sP-selectin-Fc, but not monomeric sP-selectin, triggered integrin-dependent adhesion of mouse leukocytes in vitro. Antibody-induced oligomerization of sP-selectin or sP-selectin-Fc was required to trigger formation of neutrophil extracellular traps. Injecting sP-selectin-Fc, but not sP-selectin, into mice augmented integrin-dependent adhesion of neutrophils in venules, generated tissue factor-bearing microparticles, shortened plasma-clotting times, and increased thrombus frequency in the inferior vena cava. Furthermore, transgenic mice that overexpressed monomeric sP-selectin did not exhibit increased inflammation or thrombosis. We conclude that elevated plasma sP-selectin is a consequence rather than a cause of cardiovascular disease.
Collapse
|
46
|
Platelet microparticles infiltrating solid tumors transfer miRNAs that suppress tumor growth. Blood 2017; 130:567-580. [PMID: 28500171 DOI: 10.1182/blood-2016-11-751099] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 05/08/2017] [Indexed: 12/13/2022] Open
Abstract
Platelet-derived microparticles (PMPs) are associated with enhancement of metastasis and poor cancer outcomes. Circulating PMPs transfer platelet microRNAs (miRNAs) to vascular cells. Solid tumor vasculature is highly permeable, allowing the possibility of PMP-tumor cell interaction. Here, we show that PMPs infiltrate solid tumors in humans and mice and transfer platelet-derived RNA, including miRNAs, to tumor cells in vivo and in vitro, resulting in tumor cell apoptosis. MiR-24 was a major species in this transfer. PMP transfusion inhibited growth of both lung and colon carcinoma ectopic tumors, whereas blockade of miR-24 in tumor cells accelerated tumor growth in vivo, and prevented tumor growth inhibition by PMPs. Conversely, Par4-deleted mice, which had reduced circulating microparticles (MPs), supported accelerated tumor growth which was halted by PMP transfusion. PMP targeting was associated with tumor cell apoptosis in vivo. We identified direct RNA targets of platelet-derived miR-24 in tumor cells, which included mitochondrial mt-Nd2, and Snora75, a noncoding small nucleolar RNA. These RNAs were suppressed in PMP-treated tumor cells, resulting in mitochondrial dysfunction and growth inhibition, in an miR-24-dependent manner. Thus, platelet-derived miRNAs transfer in vivo to tumor cells in solid tumors via infiltrating MPs, regulate tumor cell gene expression, and modulate tumor progression. These findings provide novel insight into mechanisms of horizontal RNA transfer and add multiple layers to the regulatory roles of miRNAs and PMPs in tumor progression. Plasma MP-mediated transfer of regulatory RNAs and modulation of gene expression may be a common feature with important outcomes in contexts of enhanced vascular permeability.
Collapse
|
47
|
Tafelmeier M, Fischer A, Orsó E, Konovalova T, Böttcher A, Liebisch G, Matysik S, Schmitz G. Mildly oxidized HDL decrease agonist-induced platelet aggregation and release of pro-coagulant platelet extracellular vesicles. J Steroid Biochem Mol Biol 2017; 169:176-188. [PMID: 27163393 DOI: 10.1016/j.jsbmb.2016.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 05/01/2016] [Accepted: 05/04/2016] [Indexed: 01/17/2023]
Abstract
Stored platelet concentrates (PLCs) for therapeutic purpose, develop a platelet storage lesion (PSL), characterized by impaired platelet (PLT) viability and function, platelet extracellular vesicle (PL-EV) release and profound lipidomic changes. Whereas oxidized low-density lipoprotein (oxLDL) activates PLTs and promotes atherosclerosis, effects linked to oxidized high-density lipoprotein (oxHDL) are poorly characterized. PLCs from blood donors were treated with native (nHDL) or mildly oxidized HDL (moxHDL) for 5days under blood banking conditions. Flow cytometry, nanoparticle tracking analysis (NTA), aggregometry, immunoblot analysis and mass spectrometry were carried out to analyze PL-EV and platelet exosomes (PL-EX) release, PLT aggregation, protein expression, and PLT and plasma lipid composition. In comparison to total nHDL, moxHDL significantly decreased PL-EV release by -36% after 5days of PLT storage and partially reversed agonist-induced PLT aggregation. PL-EV release positively correlated with PLT aggregation. MoxHDL improved PLT membrane lipid homeostasis through enhanced uptake of lysophospholipids and their remodeling to corresponding phospholipid species. This also appeared for sphingomyelin (SM) and d18:0/d18:1 sphingosine-1-phosphate (S1P) at the expense of ceramide (Cer) and hexosylceramide (HexCer) leading to reduced Cer/S1P ratio as PLT-viability indicator. This membrane remodeling was associated with increased content of CD36 and maturation of scavenger receptor-B1 (SR-B1) protein in secreted PL-EVs. MoxHDL, more potently than nHDL, improves PLT-membrane lipid homeostasis, partially antagonizes PL-EV release and agonist-induced PLT aggregation. Altogether, this may be the result of more efficient phospho- and sphingolipid remodeling mediated by CD36 and SR-B1 in the absence of ABCA1 on PLTs. As in vitro supplement in PLCs, moxHDL has the potential to improve PLC quality and to prolong storage.
Collapse
Affiliation(s)
- M Tafelmeier
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic of Regensburg Franz-Josef-Strauss-Allee 11, D-93052 Regensburg, Germany
| | - A Fischer
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic of Regensburg Franz-Josef-Strauss-Allee 11, D-93052 Regensburg, Germany
| | - E Orsó
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic of Regensburg Franz-Josef-Strauss-Allee 11, D-93052 Regensburg, Germany
| | - T Konovalova
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic of Regensburg Franz-Josef-Strauss-Allee 11, D-93052 Regensburg, Germany
| | - A Böttcher
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic of Regensburg Franz-Josef-Strauss-Allee 11, D-93052 Regensburg, Germany
| | - G Liebisch
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic of Regensburg Franz-Josef-Strauss-Allee 11, D-93052 Regensburg, Germany
| | - S Matysik
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic of Regensburg Franz-Josef-Strauss-Allee 11, D-93052 Regensburg, Germany
| | - G Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic of Regensburg Franz-Josef-Strauss-Allee 11, D-93052 Regensburg, Germany.
| |
Collapse
|
48
|
Spinelli SL, Lannan KL, Loelius SG, Phipps RP. In Vitro and Ex Vivo Approaches to Evaluate Next-Generation Tobacco and Non-Tobacco Products on Human Blood Platelets. ACTA ACUST UNITED AC 2017; 3:110-120. [PMID: 28337466 PMCID: PMC5338183 DOI: 10.1089/aivt.2016.0034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human blood platelets are major hemostatic regulators in the circulation and important in the mediation of chronic inflammation and immunomodulation. They are key elements that promote cardiovascular pathogenesis that leads to atherosclerosis, thrombosis, myocardial infarction, and stroke. New information on tobacco use and platelet dysregulation shows that these highly understudied vascular cells are dysregulated by tobacco smoke. Thus, platelet function studies should be an important consideration for the evaluation of existing and next-generation tobacco and non-tobacco products. Novel in vitro approaches are being sought to investigate these products and their influence on platelet function. Platelets are ideally suited for product assessment, as robust and novel in vitro translational methods are available to assess platelet function. Furthermore, the use of human biological systems has the advantage that risk predictions will better reflect the human condition.
Collapse
Affiliation(s)
- Sherry L Spinelli
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry , Rochester, New York
| | - Katie L Lannan
- Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry , Rochester, New York
| | - Shannon G Loelius
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry , Rochester, New York
| | - Richard P Phipps
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York.; Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York.; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York
| |
Collapse
|
49
|
Laroche M, Dunois C, Vissac AM, Amiral J. Update on functional and genetic laboratory assays for the detection of platelet microvesicles. Platelets 2017; 28:235-241. [PMID: 28102740 DOI: 10.1080/09537104.2016.1265925] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Functional and genetic assays for measuring platelet microvesicles (PMVs) are presented and discussed. Functional assays concern two groups of methods: a) homogeneous assays using the cofactor activity of phospholipids (PPLs) contained in PMVs and present in assayed plasmas, and a coagulation or a thrombin generation assay (TGA) as "end points"; b) capture-based assays, in which PMVs bind to an immobilized ligand, such as Annexin V in the presence of calcium, or monoclonal antibodies (MoAbs) specific for membrane proteins. Genetic assays aim to detect micro-RNA (miRNA) present in PMVs: miRNA must be extracted from plasma, and the expression pattern can be determined by various methods such as quantitative real-time PCR, microarray or sequencing. All these technical approaches introduce new exploration tools for measuring or quantitating PMVs or their associated activities, as biomarkers for disease evolution, their diagnosis or prognosis, and for monitoring of some antithrombotic or anti-inflammatory therapies. They offer invaluable analytical tools for research, drug discovery and epidemiological studies and have a strong potential as diagnostic tests.
Collapse
Affiliation(s)
- Maxime Laroche
- a R&D Department , HYPHEN BioMed , Neuville sur Oise , France
| | - Claire Dunois
- a R&D Department , HYPHEN BioMed , Neuville sur Oise , France
| | | | - Jean Amiral
- b Scientific Consultant for HYPHEN BioMed , Neuville sur Oise , France
| |
Collapse
|
50
|
Brisson AR, Tan S, Linares R, Gounou C, Arraud N. Extracellular vesicles from activated platelets: a semiquantitative cryo-electron microscopy and immuno-gold labeling study. Platelets 2017; 28:263-271. [DOI: 10.1080/09537104.2016.1268255] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Alain R. Brisson
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France
| | - Sisareuth Tan
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France
| | - Romain Linares
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France
| | - Céline Gounou
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France
| | - Nicolas Arraud
- Extracellular Vesicles and Membrane Repair, UMR-5248-CBMN CNRS-University of Bordeaux-IPB, Allée Geoffroy Saint-Hilaire, Pessac, France
| |
Collapse
|