1
|
Lou J, Zhang J, Deng Q, Chen X. Neutrophil extracellular traps mediate neuro-immunothrombosis. Neural Regen Res 2024; 19:1734-1740. [PMID: 38103239 PMCID: PMC10960287 DOI: 10.4103/1673-5374.389625] [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: 03/29/2023] [Revised: 07/29/2023] [Accepted: 10/14/2023] [Indexed: 12/18/2023] Open
Abstract
Neutrophil extracellular traps are primarily composed of DNA and histones and are released by neutrophils to promote inflammation and thrombosis when stimulated by various inflammatory reactions. Neutrophil extracellular trap formation occurs through lytic and non-lytic pathways that can be further classified by formation mechanisms. Histones, von Willebrand factor, fibrin, and many other factors participate in the interplay between inflammation and thrombosis. Neuro-immunothrombosis summarizes the intricate interplay between inflammation and thrombosis during neural development and the pathogenesis of neurological diseases, providing cutting-edge insights into post-neurotrauma thrombotic events. The blood-brain barrier defends the brain and spinal cord against external assaults, and neutrophil extracellular trap involvement in blood-brain barrier disruption and immunothrombosis contributes substantially to secondary injuries in neurological diseases. Further research is needed to understand how neutrophil extracellular traps promote blood-brain barrier disruption and immunothrombosis, but recent studies have demonstrated that neutrophil extracellular traps play a crucial role in immunothrombosis, and identified modulators of neuro-immunothrombosis. However, these neurological diseases occur in blood vessels, and the mechanisms are unclear by which neutrophil extracellular traps penetrate the blood-brain barrier to participate in immunothrombosis in traumatic brain injury. This review discusses the role of neutrophil extracellular traps in neuro-immunothrombosis and explores potential therapeutic interventions to modulate neutrophil extracellular traps that may reduce immunothrombosis and improve traumatic brain injury outcomes.
Collapse
Affiliation(s)
- Jianbo Lou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Quanjun Deng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| |
Collapse
|
2
|
Sun DS, Chang HH. Extracellular vesicles: Function, resilience, biomarker, bioengineering, and clinical implications. Tzu Chi Med J 2024; 36:251-259. [PMID: 38993825 PMCID: PMC11236075 DOI: 10.4103/tcmj.tcmj_28_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 07/13/2024] Open
Abstract
Extracellular vesicles (EVs) have emerged as key players in intercellular communication, disease pathology, and therapeutic innovation. Initially overlooked as cellular debris, EVs are now recognized as vital mediators of cell-to-cell communication, ferrying a cargo of proteins, nucleic acids, and lipids, providing cellular resilience in response to stresses. This review provides a comprehensive overview of EVs, focusing on their role as biomarkers in disease diagnosis, their functional significance in physiological and pathological processes, and the potential of bioengineering for therapeutic applications. EVs offer a promising avenue for noninvasive disease diagnosis and monitoring, reflecting the physiological state of originating cells. Their diagnostic potential spans a spectrum of diseases, including cancer, cardiovascular disorders, neurodegenerative diseases, and infectious diseases. Moreover, their presence in bodily fluids such as blood, urine, and cerebrospinal fluid enhances their diagnostic utility, presenting advantages over traditional methods. Beyond diagnostics, EVs mediate crucial roles in intercellular communication, facilitating the transfer of bioactive molecules between cells. This communication modulates various physiological processes such as tissue regeneration, immune modulation, and neuronal communication. Dysregulation of EV-mediated communication is implicated in diseases such as cancer, immune disorders, and neurodegenerative diseases, highlighting their therapeutic potential. Bioengineering techniques offer avenues for manipulating EVs for therapeutic applications, from isolation and purification to engineering cargo and targeted delivery systems. These approaches hold promise for developing novel therapeutics tailored to specific diseases, revolutionizing personalized medicine. However, challenges such as standardization, scalability, and regulatory approval need addressing for successful clinical translation. Overall, EVs represent a dynamic frontier in biomedical research with vast potential for diagnostics, therapeutics, and personalized medicine.
Collapse
Affiliation(s)
- Der-Shan Sun
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
3
|
Mackman N. Tissue Factor and COVID-19 Associated Thrombosis. Arterioscler Thromb Vasc Biol 2024; 44:523-529. [PMID: 38381854 PMCID: PMC10883617 DOI: 10.1161/atvbaha.123.320144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024]
Abstract
Microbial infections activate the innate and adaptive immune systems.1 Pathogen-associated molecular patterns produced by microbes, such as double-stranded RNA, are detected by PRRs (pattern-recognition receptors), such as toll-like receptor 3, and this leads to the expression of interferons and cytokines.1,2.
Collapse
Affiliation(s)
- Nigel Mackman
- Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill
| |
Collapse
|
4
|
Nappi F. To Gain Insights into the Pathophysiological Mechanisms of the Thrombo-Inflammatory Process in the Atherosclerotic Plaque. Int J Mol Sci 2023; 25:47. [PMID: 38203218 PMCID: PMC10778759 DOI: 10.3390/ijms25010047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/17/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024] Open
Abstract
Thromboinflammation, the interplay between thrombosis and inflammation, is a significant pathway that drives cardiovascular and autoimmune diseases, as well as COVID-19. SARS-CoV-2 causes inflammation and blood clotting issues. Innate immune cells have emerged as key modulators of this process. Neutrophils, the most predominant white blood cells in humans, are strategically positioned to promote thromboinflammation. By releasing decondensed chromatin structures called neutrophil extracellular traps (NETs), neutrophils can initiate an organised cell death pathway. These structures are adorned with histones, cytoplasmic and granular proteins, and have cytotoxic, immunogenic, and prothrombotic effects that can hasten disease progression. Protein arginine deiminase 4 (PAD4) catalyses the citrullination of histones and is involved in the release of extracellular DNA (NETosis). The neutrophil inflammasome is also required for this process. Understanding the link between the immunological function of neutrophils and the procoagulant and proinflammatory activities of monocytes and platelets is important in understanding thromboinflammation. This text discusses how vascular blockages occur in thromboinflammation due to the interaction between neutrophil extracellular traps and ultra-large VWF (von Willebrand Factor). The activity of PAD4 is important for understanding the processes that drive thromboinflammation by linking the immunological function of neutrophils with the procoagulant and proinflammatory activities of monocytes and platelets. This article reviews how vaso-occlusive events in thrombo-inflammation occur through the interaction of neutrophil extracellular traps with von Willebrand factor. It highlights the relevance of PAD4 in neutrophil inflammasome assembly and neutrophil extracellular traps in thrombo-inflammatory diseases such as atherosclerosis and cardiovascular disease. Interaction between platelets, VWF, NETs and inflammasomes is critical for the progression of thromboinflammation in several diseases and was recently shown to be active in COVID-19.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| |
Collapse
|
5
|
Ling T, Liu J, Dong L, Liu J. The roles of P-selectin in cancer cachexia. Med Oncol 2023; 40:338. [PMID: 37870739 DOI: 10.1007/s12032-023-02207-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 09/30/2023] [Indexed: 10/24/2023]
Abstract
P-selectin, a cell adhesion molecule of the selectin family, is expressed on the surface of activated endothelial cells (ECs) and platelets. Binding of P-selectin to P-selectin glycoprotein ligand-1 (PSGL-1) supports the leukocytes capture and rolling on stimulated ECs and increases the aggregation of leukocytes and activated platelets. Cancer cachexia is a systemic inflammation disorder characterized by metabolic disturbances, reduced body weight, loss of appetite, fat depletion, and progressive muscle atrophy. Cachexia status is associated with increased pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), which activates ECs to release P-selectin. Single-nucleotide polymorphisms (SNPs) loci of P-selectin encoding gene SELP are associated with higher level of plasma P-selectin and increase the susceptibility to cachexia in cancer patients. Elevated P-selectin expression has been observed in the hypothalamus, liver, and gastrocnemius muscle in animal models with cancer cachexia. Increased P-selectin may cause excessive inflammatory processes, muscle atrophy, and blood hypercoagulation, thus facilitating the development of cancer cachexia. In this review, physiological functions of P-selectin and its potential roles in cancer cachexia have been summarized. We also discuss the therapeutic potential of P-selectin inhibitors for the treatment of cancer cachexia.
Collapse
Affiliation(s)
- Tingting Ling
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, School of Clinical Medicine, Weifang Medical College, Weifang, 261053, China
| | - Jing Liu
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, 16766 Jingshi Road, Jinan, 250014, China
| | - Liang Dong
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Ju Liu
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, 16766 Jingshi Road, Jinan, 250014, China.
| |
Collapse
|
6
|
Nair S, Nova-Lamperti E, Labarca G, Kulasinghe A, Short KR, Carrión F, Salomon C. Genomic communication via circulating extracellular vesicles and long-term health consequences of COVID-19. J Transl Med 2023; 21:709. [PMID: 37817137 PMCID: PMC10563316 DOI: 10.1186/s12967-023-04552-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023] Open
Abstract
COVID-19 continues to affect an unprecedented number of people with the emergence of new variants posing a serious challenge to global health. There is an expansion of knowledge in understanding the pathogenesis of Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the impact of the acute disease on multiple organs. In addition, growing evidence reports that the impact of COVID-19 on different organs persists long after the recovery phase of the disease, leading to long-term consequences of COVID-19. These long-term consequences involve pulmonary as well as extra-pulmonary sequelae of the disease. Noteably, recent research has shown a potential association between COVID-19 and change in the molecular cargo of extracellular vesicles (EVs). EVs are vesicles released by cells and play an important role in cell communication by transfer of bioactive molecules between cells. Emerging evidence shows a strong link between EVs and their molecular cargo, and regulation of metabolism in health and disease. This review focuses on current knowledge about EVs and their potential role in COVID-19 pathogenesis, their current and future implications as tools for biomarker and therapeutic development and their possible effects on long-term impact of COVID-19.
Collapse
Affiliation(s)
- Soumyalekshmi Nair
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4072, Australia
| | - Estefania Nova-Lamperti
- Molecular and Translational Immunology Laboratory, Clinical Biochemistry and Immunology Department, Pharmacy Faculty, Universidad de Concepción, Concepción, Chile
| | - Gonzalo Labarca
- Molecular and Translational Immunology Laboratory, Clinical Biochemistry and Immunology Department, Pharmacy Faculty, Universidad de Concepción, Concepción, Chile
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4102, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Australia
| | - Flavio Carrión
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, UQ Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, Qld, 4072, Australia.
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago, Chile.
| |
Collapse
|
7
|
Della Corte V, Riolo R, Scaglione S, Pecoraro R, Tuttolomondo A. The Role of Biomarkers, Metabolomics, and COVID-19 in Venous Thromboembolism-A Review of Literature. Int J Mol Sci 2023; 24:13411. [PMID: 37686216 PMCID: PMC10488048 DOI: 10.3390/ijms241713411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/22/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
In recent years, the field of venous thromboembolism has undergone numerous innovations, starting from the recent discoveries on the role of biomarkers, passing through the role of metabolomics in expanding our knowledge on pathogenic mechanisms, which have opened up new therapeutic targets. A variety of studies have contributed to characterizing the metabolic phenotype that occurs in venous thromboembolism, identifying numerous pathways that are altered in this setting. Among these pathways are the metabolism of carnitine, tryptophan, purine, and fatty acids. Furthermore, new evidence has emerged with the recent COVID-19 pandemic. Hypercoagulability phenomena induced by this viral infection appear to be related to altered von Willebrand factor activity, alteration of the renin-angiotensin-aldosterone system, and dysregulation of both innate and adaptive immunity. This is the first literature review that brings together the most recent evidence regarding biomarkers, metabolomics, and COVID-19 in the field of venous thromboembolism, while also mentioning current therapeutic protocols.
Collapse
Affiliation(s)
- Vittoriano Della Corte
- Internal Medicine and Stroke Care Ward, Department of Health Promotion, Maternal and Infant Care, Internal Medicine and Medical Specialities (PROMISE) “G. D’Alessandro”, University of Palermo, Piazza delle Cliniche n.2, 90127 Palermo, Italy
| | | | | | | | | |
Collapse
|
8
|
Madkhali AM, Mobarki AA, Ghzwani AH, Al-Mekhlafi HM, Zhranei A, Osais A, Sohel A, Othman B, Dobie G, Hamali HA. Elevated Levels of Procoagulant Microvesicles and Tissue-Factor Bearing Microvesicles in Malaria Patients. Int J Gen Med 2023; 16:1205-1215. [PMID: 37041799 PMCID: PMC10083009 DOI: 10.2147/ijgm.s402212] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/30/2023] [Indexed: 04/08/2023] Open
Abstract
Background Procoagulant microvesicles (MVs) are submicron membrane fragments released from activated cells and cells undergoing apoptosis. The procoagulant activity of MVs is enhanced in the presence of tissue factor (TF). MVs and TF are active mediators that induce pro-inflammatory response and prothrombotic tendency and have been linked to the severity of several disorders, including malaria infection. The current study aimed to measure the levels of circulating procoagulant MVs and TF-bearing MVs in malaria patients and correlate these levels with other hematological parameters and parasitemia. Materials and Methods Levels of MVs and TF-bearing MVs in the plasma of children and adult patients infected with Plasmodium falciparum were measured alongside matched healthy controls. Results Patients with Plasmodium falciparum infection had ~3.8 times MVs (p < 0.0001) and ~13.0 times TF-bearing MVs compared to the matched healthy controls. MVs showed inverse significant correlation with platelet count (p = 0.0055), hemoglobin (p = 0.0004) and parasitemia. Conclusion Elevated levels of MVs and TF-bearing MVs could be useful biomarkers to evaluate the procoagulant activity, inflammatory response and parasitemia levels in malaria infection, aiding in better management of the disease.
Collapse
Affiliation(s)
- Aymen M Madkhali
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| | - Abdullah A Mobarki
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| | - Ahmad H Ghzwani
- Medical Research Center, Jazan University, Gizan, Saudi Arabia
| | | | - Ahmed Zhranei
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| | - Abdulrahman Osais
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| | - Ahmed Sohel
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| | - Basim Othman
- Department of Public Health, Faculty of Applied Medical Sciences, Albaha University, Albaha, Saudi Arabia
| | - Gasim Dobie
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
| | - Hassan A Hamali
- Department of Medical Laboratory Technology, College of Applied Medical Sciences, Jazan University, Gizan, Saudi Arabia
- Correspondence: Hassan A Hamali, Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, Jazan University, P.O. Box 1906, Gizan, 45142, Saudi Arabia, Email
| |
Collapse
|
9
|
Humic Acids Inhibit Platelet Activation to Reduce Venous Thromboembolism in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6606423. [PMID: 36588591 PMCID: PMC9797308 DOI: 10.1155/2022/6606423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 11/17/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
Objective We aimed to investigate the effects of the natural product humic acids (HA) on platelet activation and development of venous thromboembolism (VTE) in mice and further explore the relevant mechanism. Methods Eight-week C57BL/6 mice were randomly assigned to three groups: sham operation group (n = 7), VTE group (n = 8), and VTE + HA group (n = 10). Thrombi were harvested to hematoxylin-eosin staining to evaluate the thrombolysis and recanalization of the thrombus. In addition, flow cytometry was performed to detect the expression levels of protein disulfide isomerase on endothelial-derived exosomes and glycoprotein IIb/IIIa on the surface of the activated platelets surface in plasma. Furthermore, the protein expression level of glycoprotein IIb/IIIa in thrombus was determined by immunohistochemistry and immunofluorescence. Results The length of thrombosis in the VTE + HA group was significantly shorter than that in the VTE group (P = 0.040). No significant differences were observed in thrombolysis and recanalization between the VTE + HA group and the VTE group (P > 0.05). The content of protein disulfide isomerase carried by endothelial-derived exosomes was significantly increased in the VTE group (P = 0.008) but significantly reduced by native humic acids (P = 0.012). Compared with the VTE group, the expression of glycoprotein IIb/IIIa on activated platelet surface in the VTE + HA group was significantly decreased (P = 0.002). The concentration of plasmatic P-selectin in the VTE group was significantly higher than that in the VTE + HA group (P < 0.001). Conclusion We demonstrate that HA possess a pharmacological property that decreases venous thrombus formation in mice. The underlying mechanism is that HA could inhibit the expression of glycoprotein IIb/IIIa on the activated platelets surface by suppressing endothelial-derived exosomes carrying on protein disulfide isomerase, thereby blocking platelet activation.
Collapse
|
10
|
Majumder R. Phosphatidylserine Regulation of Coagulation Proteins Factor IXa and Factor VIIIa. J Membr Biol 2022; 255:733-737. [PMID: 36098799 DOI: 10.1007/s00232-022-00265-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 08/09/2022] [Indexed: 12/24/2022]
Abstract
Blood coagulation is an intricate process, and it requires precise control of the activities of pro- and anticoagulant factors and sensitive signaling systems to monitor and respond to blood vessel insults. These requirements are fulfilled by phosphatidylserine, a relatively miniscule-sized lipid molecule amid the myriad of large coagulation proteins. This review limelight the role of platelet membrane phosphatidylserine (PS) in regulating a key enzymatic reaction of blood coagulation; conversion of factor X to factor Xa by the enzyme factor IXa and its cofactor factor VIIIa. PS is normally located on the inner leaflet of the resting platelet membrane but appears on the outer leaflet surface of the membrane surface after an injury happens. Human platelet activation leads to exposure of buried PS molecules on the surface of the platelet-derived membranes and the exposed PS binds to discrete and specific sites on factors IXa and VIIIa. PS binding to these sites allosterically regulates both factors IXa and VIIIa. The exposure of PS and its binding to factors IXa/VIIIa is a vital step during clotting. Insufficient exposure or a defective binding of PS to these clotting proteins is responsible for various hematologic diseases which are discussed in this review.
Collapse
Affiliation(s)
- Rinku Majumder
- Department of Biochemistry, Louisiana State University Health Sciences Center, MEB-7114, New Orleans, LA, 70112, USA.
| |
Collapse
|
11
|
Abstract
The activating interplay of thrombosis and inflammation (thromboinflammation) has been established as a major underlying pathway, driving not only cardiovascular disease but also autoimmune disease and most recently, COVID-19. Throughout the years, innate immune cells have emerged as important modulators of this process. As the most abundant white blood cell in humans, neutrophils are well-positioned to propel thromboinflammation. This includes their ability to trigger an organized cell death pathway with the release of decondensed chromatin structures called neutrophil extracellular traps. Decorated with histones and cytoplasmic and granular proteins, neutrophil extracellular traps exert cytotoxic, immunogenic, and prothrombotic effects accelerating disease progression. Distinct steps leading to extracellular DNA release (NETosis) require the activities of PAD4 (protein arginine deiminase 4) catalyzing citrullination of histones and are supported by neutrophil inflammasome. By linking the immunologic function of neutrophils with the procoagulant and proinflammatory activities of monocytes and platelets, PAD4 activity holds important implications for understanding the processes that fuel thromboinflammation. We will also discuss mechanisms whereby vascular occlusion in thromboinflammation depends on the interaction of neutrophil extracellular traps with ultra-large VWF (von Willebrand Factor) and speculate on the importance of PAD4 in neutrophil inflammasome assembly and neutrophil extracellular traps in thromboinflammatory diseases including atherosclerosis and COVID-19.
Collapse
Affiliation(s)
- Denisa D Wagner
- Program in Cellular and Molecular Medicine, Division of Hematology and Oncology, Boston Children's Hospital/Harvard Medical School, MA (D.D.W., L.A.H.)
| | - Lukas A Heger
- Program in Cellular and Molecular Medicine, Division of Hematology and Oncology, Boston Children's Hospital/Harvard Medical School, MA (D.D.W., L.A.H.)
| |
Collapse
|
12
|
Zhang X, Tang J, Kou X, Huang W, Zhu Y, Jiang Y, Yang K, Li C, Hao M, Qu Y, Ma L, Chen C, Shi S, Zhou Y. Proteomic analysis of MSC-derived apoptotic vesicles identifies Fas inheritance to ameliorate haemophilia a via activating platelet functions. J Extracell Vesicles 2022; 11:e12240. [PMID: 36856683 PMCID: PMC9927920 DOI: 10.1002/jev2.12240] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/12/2022] Open
Abstract
Apoptotic vesicles (apoVs) are apoptotic cell-derived nanosized vesicles that play a crucial role in multiple pathophysiological settings. However, their detailed characteristics, specific surface markers, and biological properties are not fully elucidated. In this study, we compared mesenchymal stem cell (MSC)-derived apoVs and exosomes from three different types of MSCs including human bone marrow MSCs (hBMSCs), human adipose MSCs (hASCs), and mouse bone marrow MSCs (mBMSCs). We established a unique protein map of MSC-derived apoVs and identified the differences between apoVs and exosomes in terms of functional protein cargo and surface markers. Furthermore, we identified 13 proteins specifically enriched in apoVs compared to exosomes, which can be used as apoV-specific biomarkers. In addition, we showed that apoVs inherited apoptotic imprints such as Fas to ameliorate haemophilia A in factor VIII knockout mice via binding to the platelets' FasL to activate platelet functions, and therefore rescuing the blood clotting disorder. In summary, we systemically characterized MSC-derived apoVs and identified their therapeutic role in haemophilia A treatment through a previously unknown Fas/FasL linkage mechanism.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of ProsthodonticsPeking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081China
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Jianxia Tang
- Hunan Key Laboratory of Oral Health Research & Hunan Clinical Research Center of Oral Major Diseases and Oral HealthXiangya School of Stomatology, Xiangya Stomatological Hospital, Central South UniversityChangsha410000China
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Xiaoxing Kou
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat‐Sen University)Ministry of EducationGuangzhou510080China
| | - Weiying Huang
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Yuan Zhu
- Department of ProsthodonticsPeking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081China
| | - Yuhe Jiang
- Department of ProsthodonticsPeking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081China
| | - Kunkun Yang
- Department of ProsthodonticsPeking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081China
| | - Can Li
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Meng Hao
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Yan Qu
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Lan Ma
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and PharmacologyUniversity of Pennsylvania, School of Dental MedicinePhiladelphiaPA 19104USA
| | - Songtao Shi
- South China Center of Craniofacial Stem Cell ResearchHospital of Stomatology, Guanghua School of Stomatology, Sun Yat‐sen UniversityGuangzhou510080China
- Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat‐Sen University)Ministry of EducationGuangzhou510080China
| | - Yongsheng Zhou
- Department of ProsthodonticsPeking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental MaterialsBeijing100081China
| |
Collapse
|
13
|
Zhang YY, Ning BT. Signaling pathways and intervention therapies in sepsis. Signal Transduct Target Ther 2021; 6:407. [PMID: 34824200 PMCID: PMC8613465 DOI: 10.1038/s41392-021-00816-9] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by dysregulated host systemic inflammatory and immune response to infection. Over decades, advanced understanding of host-microorganism interaction has gradually unmasked the genuine nature of sepsis, guiding toward new definition and novel therapeutic approaches. Diverse clinical manifestations and outcomes among infectious patients have suggested the heterogeneity of immunopathology, while systemic inflammatory responses and deteriorating organ function observed in critically ill patients imply the extensively hyperactivated cascades by the host defense system. From focusing on microorganism pathogenicity, research interests have turned toward the molecular basis of host responses. Though progress has been made regarding recognition and management of clinical sepsis, incidence and mortality rate remain high. Furthermore, clinical trials of therapeutics have failed to obtain promising results. As far as we know, there was no systematic review addressing sepsis-related molecular signaling pathways and intervention therapy in literature. Increasing studies have succeeded to confirm novel functions of involved signaling pathways and comment on efficacy of intervention therapies amid sepsis. However, few of these studies attempt to elucidate the underlining mechanism in progression of sepsis, while other failed to integrate preliminary findings and describe in a broader view. This review focuses on the important signaling pathways, potential molecular mechanism, and pathway-associated therapy in sepsis. Host-derived molecules interacting with activated cells possess pivotal role for sepsis pathogenesis by dynamic regulation of signaling pathways. Cross-talk and functions of these molecules are also discussed in detail. Lastly, potential novel therapeutic strategies precisely targeting on signaling pathways and molecules are mentioned.
Collapse
Affiliation(s)
- Yun-Yu Zhang
- Department of Pediatric Intensive Care Unit, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Bo-Tao Ning
- Department of Pediatric Intensive Care Unit, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China.
| |
Collapse
|
14
|
Haschemi R, Gockel LM, Bendas G, Schlesinger M. A Combined Activity of Thrombin and P-Selectin Is Essential for Platelet Activation by Pancreatic Cancer Cells. Int J Mol Sci 2021; 22:3323. [PMID: 33805059 PMCID: PMC8037188 DOI: 10.3390/ijms22073323] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/15/2021] [Accepted: 03/21/2021] [Indexed: 01/01/2023] Open
Abstract
Pancreatic cancer patients have an elevated risk of suffering from venous thrombosis. Among several risk factors that contribute to hypercoagulability of this malignancy, platelets possess a key role in the initiation of clot formation. Although single mechanisms of platelet activation are well-known in principle, combinations thereof and their potential synergy to mediate platelet activation is, in the case of pancreatic cancer, far from being clear. Applying an inhibitor screening approach using light transmission aggregometry, dense granule release, and thrombin formation assays, we provide evidence that a combination of tissue factor-induced thrombin formation by cancer cells and their platelet P-selectin binding is responsible for AsPC-1 and Capan-2 pancreatic cancer cell-mediated platelet activation. While the blockade of one of these pathways leads to a pronounced inhibition of platelet aggregation and dense granule release, the simultaneous blockade of both pathways is inevitable to prevent platelet aggregation completely and minimize ATP release. In contrast, MIA PaCa-2 pancreatic cancer cells express reduced levels of tissue factor and P-selectin ligands and thus turn out to be poor platelet activators. Consequently, a simultaneous blockade of thrombin and P-selectin binding seems to be a powerful approach, as mediated by heparin to crucially reduce the hypercoagulable state of pancreatic cancer patients.
Collapse
Affiliation(s)
| | | | | | - Martin Schlesinger
- Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany; (R.H.); (L.M.G.); (G.B.)
| |
Collapse
|
15
|
Shet AS, Lizarralde-Iragorri MA, Naik RP. The molecular basis for the prothrombotic state in sickle cell disease. Haematologica 2020; 105:2368-2379. [PMID: 33054077 PMCID: PMC7556662 DOI: 10.3324/haematol.2019.239350] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/22/2020] [Indexed: 12/15/2022] Open
Abstract
The genetic and molecular basis of sickle cell disease (SCD) has long since been characterized but the pathophysiological basis is not entirely defined. How a red cell hemolytic disorder initiates inflammation, endothelial dysfunction, coagulation activation and eventually leads to vascular thrombosis, is yet to be elucidated. Recent evidence has demonstrated a high frequency of unprovoked/recurrent venous thromboembolism (VTE) in SCD, with an increased risk of mortality among patients with a history of VTE. Here, we thoroughly review the molecular basis for the prothrombotic state in SCD, specifically highlighting emerging evidence for activation of overlapping inflammation and coagulation pathways, that predispose to venous thromboembolism. We share perspectives in managing venous thrombosis in SCD, highlighting innovative therapies with the potential to influence the clinical course of disease and reduce thrombotic risk, while maintaining an acceptable safety profile.
Collapse
Affiliation(s)
- Arun S. Shet
- Laboratory of Sickle Thrombosis and Vascular Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda
| | | | - Rakhi P. Naik
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
16
|
Lizarralde-Iragorri MA, Shet AS. Sickle Cell Disease: A Paradigm for Venous Thrombosis Pathophysiology. Int J Mol Sci 2020; 21:ijms21155279. [PMID: 32722421 PMCID: PMC7432404 DOI: 10.3390/ijms21155279] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023] Open
Abstract
Venous thromboembolism (VTE) is an important cause of vascular morbidity and mortality. Many risk factors have been identified for venous thrombosis that lead to alterations in blood flow, activate the vascular endothelium, and increase the propensity for blood coagulation. However, the precise molecular and cellular mechanisms that cause blood clots in the venous vasculature have not been fully elucidated. Patients with sickle cell disease (SCD) demonstrate all the risk factors for venous stasis, activated endothelium, and blood hypercoagulability, making them particularly vulnerable to VTE. In this review, we will discuss how mouse models have elucidated the complex vascular pathobiology of SCD. We review the dysregulated pathways of inflammation and coagulation in SCD and how the resultant hypercoagulable state can potentiate thrombosis through down-regulation of vascular anticoagulants. Studies of VTE pathogenesis using SCD mouse models may provide insight into the intersection between the cellular and molecular processes involving inflammation and coagulation and help to identify novel mechanistic pathways.
Collapse
|
17
|
Abstract
Neutrophils are critical to innate immunity, including host defense against bacterial and fungal infections. They achieve their host defense role by phagocytosing pathogens, secreting their granules full of cytotoxic enzymes, or expelling neutrophil extracellular traps (NETs) during the process of NETosis. NETs are weblike DNA structures decorated with histones and antimicrobial proteins released by activated neutrophils. Initially described as a means for neutrophils to neutralize pathogens, NET release also occurs in sterile inflammation, promotes thrombosis, and can mediate tissue damage. To effectively manipulate this double-edged sword to fight a particular disease, researchers must work toward understanding the mechanisms driving NETosis. Such understanding would allow the generation of new drugs to promote or prevent NETosis as needed. While knowledge regarding the (patho)physiological roles of NETosis is accumulating, little is known about the cellular and biophysical bases of this process. In this review, we describe and discuss our current knowledge of the molecular, cellular, and biophysical mechanisms mediating NET release as well as open questions in the field.
Collapse
Affiliation(s)
- Hawa Racine Thiam
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland 20892, USA; ,
| | - Siu Ling Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232
| | - Denisa D Wagner
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Clare M Waterman
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland 20892, USA; ,
| |
Collapse
|
18
|
Holstein K, Matysiak A, Witt L, Sievers B, Beckmann L, Haddad M, Renné T, Voigtlaender M, Langer F. LPS-induced expression and release of monocyte tissue factor in patients with haemophilia. Ann Hematol 2020; 99:1531-1542. [PMID: 32430703 PMCID: PMC7316670 DOI: 10.1007/s00277-020-04075-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022]
Abstract
In haemophilia, thrombin generation and fibrin deposition upon vascular injury critically depend on the tissue factor (TF)-driven coagulation pathway. TF expression by monocytes/macrophages and circulating microvesicles contributes to haemostasis, thrombosis and inflammation. Inflammation is a hallmark of blood-induced joint disease. The aim of this study is to correlate TF production by whole-blood monocytes with inflammatory markers and clinical parameters in patients with moderate-to-severe haemophilia A or B (n = 43) in comparison to healthy males (n = 23). Monocyte TF antigen and microvesicle-associated TF procoagulant activity (MV TF PCA) were measured immediately after blood draw (baseline) and following incubation of whole blood with buffer or lipopolysaccharide (LPS) using two-colour flow cytometry and chromogenic FXa generation assay, respectively. Patients with HIV or uncontrolled HBV/HCV infections were excluded. TF was hardly detectable and not different in baseline and buffer-treaded samples from both groups. Stimulation with LPS, however, induced monocyte TF production, with increased TF-specific mean fluorescence intensity (P = 0.08) and MV TF PCA (P < 0.05) in patients compared to controls. Patients also had elevated hs-CRP and IL-6 serum levels (P < 0.001), which correlated with LPS-induced TF parameters. Further exploratory analyses revealed that the presence of systemic (low-grade) inflammation and boosted LPS-induced monocyte TF production were mainly restricted to patients with clinically controlled HBV and/or HCV infection (n = 16), who were older and also had a significantly worse orthopaedic joint score than patients with no history of viral hepatitis (P < 0.01). Our study delineates a previously unrecognised link between systemic inflammation and inducible monocyte TF production in patients with haemophilia A or B.
Collapse
Affiliation(s)
- Katharina Holstein
- Department of Haematology and Oncology, University Cancer Centre Hamburg (UCCH), University Medical Centre Eppendorf, Hamburg, Germany
| | - Anna Matysiak
- Department of Haematology and Oncology, University Cancer Centre Hamburg (UCCH), University Medical Centre Eppendorf, Hamburg, Germany
| | - Leonora Witt
- Department of Haematology and Oncology, University Cancer Centre Hamburg (UCCH), University Medical Centre Eppendorf, Hamburg, Germany
| | - Bianca Sievers
- Department of Haematology and Oncology, University Cancer Centre Hamburg (UCCH), University Medical Centre Eppendorf, Hamburg, Germany
| | - Lennart Beckmann
- Department of Haematology and Oncology, University Cancer Centre Hamburg (UCCH), University Medical Centre Eppendorf, Hamburg, Germany
| | - Munif Haddad
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre Eppendorf, Hamburg, Germany
| | - Minna Voigtlaender
- Department of Haematology and Oncology, University Cancer Centre Hamburg (UCCH), University Medical Centre Eppendorf, Hamburg, Germany
| | - Florian Langer
- Department of Haematology and Oncology, University Cancer Centre Hamburg (UCCH), University Medical Centre Eppendorf, Hamburg, Germany.
| |
Collapse
|
19
|
Phosphatidylserine positive microparticles improve hemostasis in in-vitro hemophilia A plasma models. Sci Rep 2020; 10:7871. [PMID: 32398812 PMCID: PMC7217932 DOI: 10.1038/s41598-020-64686-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 04/17/2020] [Indexed: 12/19/2022] Open
Abstract
Circulating microparticles (MPs) are procoagulant due to the surface containing phosphatidylserine (PS), which facilitates coagulation. We investigated if MPs improve hemostasis in HA plasma models. MPs isolated from pooled normal human plasma were added to severe, moderate and mild HA plasma models (0%, 2.5%, 20% FVIII). The MPs’ effect on hemostasis was evaluated by calibrated automated thrombogram (CAT) and overall hemostasis potential (OHP) assays, while fibrin structure was imaged by standard confocal, stimulated emission depletion (STED) microscopy and scanning electron microscopy (SEM). MPs partially restored thrombin generation and fibrin formation in all HA plasma models. The procoagulant effect of MPs requires PS exposure, to a less extent of contact pathway activation, but not tissue factor exposure or in vitro stimulation of MPs. MPs partially normalized the fibrin structure, and using super-resolution STED, MPs attached to fibrin were clearly resolved. In summary, our results demonstrate that PS positive MPs could improve hemostasis in HA plasma models.
Collapse
|
20
|
Soni P, Shanmukhaiah C, Patil R, Shetty S. Differential response to FEIBA is strongly associated with the prothrombotic microparticles. Blood Cells Mol Dis 2020; 84:102441. [PMID: 32388201 DOI: 10.1016/j.bcmd.2020.102441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Treatment of patients with hemophilia with an inhibitor is generally done using bypassing agents (BPA), wherein variability in response is observed. Due to lack of validated laboratory assays, monitoring is being carried out by clinical response only. Emerging biomarkers like procoagulant microparticles (MPs) may prove to be promising. AIM To analyze whether procoagulant MP levels correlate with clinical response to FEIBA therapy. MATERIALS AND METHODS Total phosphatidylserine (PS) expressing MPs along with different cell derived MPs were measured in blood samples obtained prior and 2 hour post-FEIBA infusion in 64 bleeding episodes associated with 43 severe hemophilia patients. RESULTS AND DISCUSSION Patients with excellent response showed statistically significant increase in %MP of PS-MPs (p < 0.0001; 95.0% CI Range: -64.33 to -24.42) when compared to those with moderate response; platelet %MP change was also found significantly associated (p < 0.05) with clinical response. In search of an assay for monitoring FEIBA, results though preliminary seem to be promising with increase in %PS-MP correlating well with the clinical response. Coagulation being multifactorial process involves multiple factors for balanced hemostasis, which needs to be accounted. Larger studies in this line may provide indications for usage of MPs as monitoring and dose adjustment tool of FEIBA therapy.
Collapse
Affiliation(s)
- Puja Soni
- National Institute of Immunohematology (NIIH-ICMR), 13th floor, KEM Hospital, Parel, Mumbai 400012, India
| | | | - Rucha Patil
- National Institute of Immunohematology (NIIH-ICMR), 13th floor, KEM Hospital, Parel, Mumbai 400012, India
| | - Shrimati Shetty
- National Institute of Immunohematology (NIIH-ICMR), 13th floor, KEM Hospital, Parel, Mumbai 400012, India.
| |
Collapse
|
21
|
NETosis proceeds by cytoskeleton and endomembrane disassembly and PAD4-mediated chromatin decondensation and nuclear envelope rupture. Proc Natl Acad Sci U S A 2020; 117:7326-7337. [PMID: 32170015 PMCID: PMC7132277 DOI: 10.1073/pnas.1909546117] [Citation(s) in RCA: 216] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Neutrophils are white blood cells specialized as the first line of host defense in the immune system. One way they protect organisms is through NETosis, in which they expel their DNA to form a web-like trap that ensnares pathogens and promotes clotting. However, NETs also mediate sterile inflammation, causing damage to the body. We used high-resolution live-cell microscopy to characterize the timing of dynamic cellular events leading to NETosis in human and mouse neutrophils and a neutrophil-like cell line. We discovered that NETosis proceeds by a stepwise sequence of cellular events that is conserved across species and requires the activity of the PAD4 enzyme for DNA to be released from the nucleus and cell membrane. Neutrophil extracellular traps (NETs) are web-like DNA structures decorated with histones and cytotoxic proteins that are released by activated neutrophils to trap and neutralize pathogens during the innate immune response, but also form in and exacerbate sterile inflammation. Peptidylarginine deiminase 4 (PAD4) citrullinates histones and is required for NET formation (NETosis) in mouse neutrophils. While the in vivo impact of NETs is accumulating, the cellular events driving NETosis and the role of PAD4 in these events are unclear. We performed high-resolution time-lapse microscopy of mouse and human neutrophils and differentiated HL-60 neutrophil-like cells (dHL-60) labeled with fluorescent markers of organelles and stimulated with bacterial toxins or Candida albicans to induce NETosis. Upon stimulation, cells exhibited rapid disassembly of the actin cytoskeleton, followed by shedding of plasma membrane microvesicles, disassembly and remodeling of the microtubule and vimentin cytoskeletons, ER vesiculation, chromatin decondensation and nuclear rounding, progressive plasma membrane and nuclear envelope (NE) permeabilization, nuclear lamin meshwork and then NE rupture to release DNA into the cytoplasm, and finally plasma membrane rupture and discharge of extracellular DNA. Inhibition of actin disassembly blocked NET release. Mouse and dHL-60 cells bearing genetic alteration of PAD4 showed that chromatin decondensation, lamin meshwork and NE rupture and extracellular DNA release required the enzymatic and nuclear localization activities of PAD4. Thus, NETosis proceeds by a stepwise sequence of cellular events culminating in the PAD4-mediated expulsion of DNA.
Collapse
|
22
|
Myers D, Lester P, Adili R, Hawley A, Durham L, Dunivant V, Reynolds G, Crego K, Zimmerman Z, Sood S, Sigler R, Fogler W, Magnani J, Holinstat M, Wakefield T. A new way to treat proximal deep venous thrombosis using E-selectin inhibition. J Vasc Surg Venous Lymphat Disord 2020; 8:268-278. [PMID: 32067728 PMCID: PMC9006622 DOI: 10.1016/j.jvsv.2019.08.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 08/16/2019] [Indexed: 11/23/2022]
Abstract
OBJECTIVE There is an inter-relationship between thrombosis and inflammation. Previously, we have shown the importance of P-selectin in thrombogenesis and thrombus resolution in many preclinical animal models. The role of E-selectin has been explored in rodent models and in a small pilot study of clinical calf vein deep venous thrombosis. The purpose of this study was to determine the role of E-selectin in thrombosis in a primate model of proximal iliac vein thrombosis, a model close to the human condition. METHODS Iliac vein thrombosis was induced with a well-characterized primate model. Through a transplant incision, the hypogastric vein and iliac vein branches were ligated. Thrombus was induced by balloon occlusion of the proximal and distal iliac vein for 6 hours. The balloons were then deflated, and the primates recovered. Starting on postocclusion day 2, animals were treated with the E-selectin inhibitor GMI-1271, 25 mg/kg subcutaneously, once daily until day 21 (n = 4). Nontreated control animals received no treatment (n = 5). All animals were evaluated by magnetic resonance venography (MRV); evaluation of vessel area by ultrasound, protein analysis, hematology (complete blood count), and coagulation tests (bleeding time, prothrombin time, activated partial thromboplastin time, fibrinogen, and thromboelastography) were performed at baseline, day 2, day 7, day 14, and day 21 with euthanasia. In addition, platelet function and CD44 expression on leukocytes were determined. RESULTS E-selectin inhibition by GMI-1271 significantly increased vein recanalization by MRV vs control animals on day 14 (P < .05) and day 21 (P < .0001). GMI-1271 significantly decreased vein wall inflammation by MRV with gadolinium vein wall enhancement vs control also on day 14 (P < .0001) and day 21 (P < .0001). The thromboelastographic measure of clot strength (maximum amplitude) showed significant decreases in animals treated with GMI-1271 vs controls at day 2 (P < .05) and day 7 (P < .05). Animals treated with GMI-1271 had significant vessel area increase by day 21 vs controls (P < .05) by ultrasound. Vein wall intimal thickening (P < .001) and intimal fibrosis (P < .05) scores were significantly decreased in GMI-1271-treated animals vs controls. Importantly, no significant differences in hematology or coagulation test results were noted between all groups, suggesting that E-selectin inhibition carries no bleeding potential. GMI-1271 did not affect platelet function or aggregation or CD44 expression on leukocytes. In addition, no episodes of bleeding were noted in either group. CONCLUSIONS This study suggests that E-selectin modulates venous thrombus progression and that its inhibition will increase thrombus recanalization and decrease vein wall inflammation, without affecting coagulation. The use of an E-selectin inhibitor such as GMI-1271 could potentially change how we treat deep venous thrombosis.
Collapse
Affiliation(s)
- Daniel Myers
- Conrad Jobst Vascular Research Laboratories, Section of Vascular Surgery, University of Michigan, Ann Arbor, Mich; Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Mich.
| | - Patrick Lester
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Mich
| | - Reheman Adili
- Department of Pharmacology, University of Michigan, Ann Arbor, Mich
| | - Angela Hawley
- Conrad Jobst Vascular Research Laboratories, Section of Vascular Surgery, University of Michigan, Ann Arbor, Mich
| | - Laura Durham
- Conrad Jobst Vascular Research Laboratories, Section of Vascular Surgery, University of Michigan, Ann Arbor, Mich
| | - Veronica Dunivant
- Conrad Jobst Vascular Research Laboratories, Section of Vascular Surgery, University of Michigan, Ann Arbor, Mich
| | - Garrett Reynolds
- Conrad Jobst Vascular Research Laboratories, Section of Vascular Surgery, University of Michigan, Ann Arbor, Mich
| | - Kiley Crego
- Conrad Jobst Vascular Research Laboratories, Section of Vascular Surgery, University of Michigan, Ann Arbor, Mich
| | - Zoe Zimmerman
- Department of Pharmacology, University of Michigan, Ann Arbor, Mich
| | - Suman Sood
- Division of Hematology/Oncology, University of Michigan, Ann Arbor, Mich
| | - Robert Sigler
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Mich
| | | | | | | | - Thomas Wakefield
- Conrad Jobst Vascular Research Laboratories, Section of Vascular Surgery, University of Michigan, Ann Arbor, Mich
| |
Collapse
|
23
|
Macia L, Nanan R, Hosseini-Beheshti E, Grau GE. Host- and Microbiota-Derived Extracellular Vesicles, Immune Function, and Disease Development. Int J Mol Sci 2019; 21:ijms21010107. [PMID: 31877909 PMCID: PMC6982009 DOI: 10.3390/ijms21010107] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/14/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) are blebs of either plasma membrane or intracellular membranes carrying a cargo of proteins, nucleic acids, and lipids. EVs are produced by eukaryotic cells both under physiological and pathological conditions. Genetic and environmental factors (diet, stress, etc.) affecting EV cargo, regulating EV release, and consequences on immunity will be covered. EVs are found in virtually all body fluids such as plasma, saliva, amniotic fluid, and breast milk, suggesting key roles in immune development and function at different life stages from in utero to aging. These will be reviewed here. Under pathological conditions, plasma EV levels are increased and exacerbate immune activation and inflammatory reaction. Sources of EV, cells targeted, and consequences on immune function and disease development will be discussed. Both pathogenic and commensal bacteria release EV, which are classified as outer membrane vesicles when released by Gram-negative bacteria or as membrane vesicles when released by Gram-positive bacteria. Bacteria derived EVs can affect host immunity with pathogenic bacteria derived EVs having pro-inflammatory effects of host immune cells while probiotic derived EVs mostly shape the immune response towards tolerance.
Collapse
Affiliation(s)
- Laurence Macia
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia;
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW 2006, Australia;
- Correspondence: (L.M.); (G.E.G.); Tel.: +61-2-8627-6525 (L.M.); +61-2-9036-3260 (G.E.G.)
| | - Ralph Nanan
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia;
- The University of Sydney, Sydney Medical School Nepean, Penrith 2751, Australia
| | - Elham Hosseini-Beheshti
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW 2006, Australia;
- Vascular Immunology Unit, The University of Sydney, NSW 2006, Australia
| | - Georges E. Grau
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW 2006, Australia;
- Vascular Immunology Unit, The University of Sydney, NSW 2006, Australia
- Correspondence: (L.M.); (G.E.G.); Tel.: +61-2-8627-6525 (L.M.); +61-2-9036-3260 (G.E.G.)
| |
Collapse
|
24
|
Borgel D, Bianchini E, Lasne D, Pascreau T, Saller F. Inflammation in deep vein thrombosis: a therapeutic target? Hematology 2019; 24:742-750. [DOI: 10.1080/16078454.2019.1687144] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Delphine Borgel
- Laboratoire d’Hématologie, AP-HP, Hôpital Necker-Enfants malades, Paris, France
- INSERM UMR-S1176, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Elsa Bianchini
- INSERM UMR-S1176, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Dominique Lasne
- Laboratoire d’Hématologie, AP-HP, Hôpital Necker-Enfants malades, Paris, France
- INSERM UMR-S1176, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Tiffany Pascreau
- Laboratoire d’Hématologie, AP-HP, Hôpital Necker-Enfants malades, Paris, France
- INSERM UMR-S1176, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - François Saller
- INSERM UMR-S1176, Université Paris Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
25
|
|
26
|
Romana M, Connes P, Key NS. Microparticles in sickle cell disease. Clin Hemorheol Microcirc 2018; 68:319-329. [DOI: 10.3233/ch-189014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Marc Romana
- Unité Biologie Intégrée du Globule Rouge, Université des Antilles, Inserm 1134, laboratoire d’Excellence GR-Ex, Paris, France
| | - Philippe Connes
- Unité Biologie Intégrée du Globule Rouge, Université des Antilles, Inserm 1134, laboratoire d’Excellence GR-Ex, Paris, France
- Laboratoire LIBM EA7424, Equipe « Biologie Vasculaire et du Globule Rouge», Université Claude Bernard Lyon 1, Villeurbanne, France
- Institut Universitaire de France, Paris, France
| | - Nigel S. Key
- Department of Medicine, Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
27
|
Hosseini-Beheshti E, Grau GER. Extracellular vesicles as mediators of immunopathology in infectious diseases. Immunol Cell Biol 2018; 96:694-703. [PMID: 29577413 DOI: 10.1111/imcb.12044] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 03/19/2018] [Accepted: 03/19/2018] [Indexed: 12/12/2022]
Abstract
In the last decades, extracellular vesicles have emerged as important elements in cell-cell communication and as key players in disease pathogenesis via transmission of their cargo between different cells. Various works have described different subpopulations of these membrane structures, based on their cell of origin, biogenesis, size, biophysical properties and cargo. In addition to their pathophysiological role in the development and progression of different diseases including infectious diseases, neurodegenerative disorders and cancer, extracellular vesicles are now recognized for their potential as novel therapeutic targets and intelligent drug delivery system. Here, we have reviewed the most recent data on different subtypes of extracellular vesicles, focusing on microvesicles and exosomes and their subpopulations, their involvement in immune-mediated pathogenesis of various infectious diseases and their role as potential therapeutic targets.
Collapse
Affiliation(s)
- Elham Hosseini-Beheshti
- Vascular Immunology Unit, Department of Pathology, School of Medical Sciences, Marie Bashir Institute and The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, Australia
| | - Georges Emile Raymond Grau
- Vascular Immunology Unit, Department of Pathology, School of Medical Sciences, Marie Bashir Institute and The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
28
|
Laberge A, Arif S, Moulin VJ. Microvesicles: Intercellular messengers in cutaneous wound healing. J Cell Physiol 2018; 233:5550-5563. [DOI: 10.1002/jcp.26426] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/02/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Alexandra Laberge
- Centre de recherche en organogenese experimentale de l'Université Laval/LOEXCentre de recherche du CHU de QuebecQuebecCanada
| | - Syrine Arif
- Centre de recherche en organogenese experimentale de l'Université Laval/LOEXCentre de recherche du CHU de QuebecQuebecCanada
| | - Véronique J. Moulin
- Centre de recherche en organogenese experimentale de l'Université Laval/LOEXCentre de recherche du CHU de QuebecQuebecCanada
- Department of SurgeryFaculty of MedicineUniversite LavalQuebecCanada
| |
Collapse
|
29
|
Kapupara K, Wen YT, Tsai RK, Huang SP. Soluble P-selectin promotes retinal ganglion cell survival through activation of Nrf2 signaling after ischemia injury. Cell Death Dis 2017; 8:e3172. [PMID: 29144506 PMCID: PMC5775414 DOI: 10.1038/cddis.2017.566] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 09/17/2017] [Accepted: 09/20/2017] [Indexed: 12/21/2022]
Abstract
Retinal ischemic injuries play an important role in the pathogenesis of several eye disorders. Inflammation and oxidative stress are key players in ischemic injuries. Following retinal ischemia, vascular endothelial cells and leukocytes express several inflammatory adhesion receptors, such as selectins and cell adhesion molecules. P-selectin stimulates leukocyte recruitment to platelet aggregates and has an important role in vascular homeostasis and inflammatory leukocyte extravasation. Soluble P-selectin can be neuroprotective through competitive binding to the receptors of endogenous P-selectin molecules. Here, we demonstrate the neuroprotective effect of a recombinant P-selectin immunoglobin G (P-sel-IgG) chimeric fusion protein in a rat anterior ischemic optic neuropathy (rAION) model. rAION was induced by photodynamic therapy. P-sel-IgG treatment reduced optic nerve edema and stabilized the blood-optic nerve barrier (BONB) in the acute phase of rAION. Further, P-sel-IgG increased the retinal ganglion cell (RGC) survival rate, reduced RGC apoptosis, preserved visual function, maintained retinal nerve fiber layer thickness, and reduced macrophage infiltration in optic nerve tissue in the chronic phase (day 28). Increased NAD(P)H quinone dehydrogenase 1 (NQO1) and heme oxygenase 1(HO-1) expression levels, along with increased transcription factor Nrf2, suggesting an antioxidant role of P-sel-IgG via the Nrf2 signaling pathway. In conclusion, this study is the first to demonstrate that P-sel-IgG treatment promotes RGC survival by stabilizing the BONB and activating the Nrf2 signaling pathway in a rAION model.
Collapse
Affiliation(s)
- Kishan Kapupara
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | - Yao-Tseng Wen
- Institute of Eye Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Rong-Kung Tsai
- Institute of Eye Research, Buddhist Tzu Chi General Hospital, Hualien, Taiwan.,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Shun-Ping Huang
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
30
|
Campbell JD. Self-defense against Bacillus anthracis toxins: Is P-selectin the key? Virulence 2017; 8:1059-1061. [PMID: 28281892 DOI: 10.1080/21505594.2017.1304344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Affiliation(s)
- John D Campbell
- a Discovery Research, Dynavax Technologies , Berkeley , CA , USA
| |
Collapse
|
31
|
Bern MM. Extracellular vesicles: how they interact with endothelium, potentially contributing to metastatic cancer cell implants. Clin Transl Med 2017; 6:33. [PMID: 28933058 PMCID: PMC5607152 DOI: 10.1186/s40169-017-0165-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/13/2017] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EV) are blebs of cellular membranes, which entrap small portions of subjacent cytosol. They are released from a variety of cells, circulate in the blood for an unknown length of time and come to rest on endothelial surfaces. They contribute to an array of physiologic pathways, the complexity of which is still being investigated. They contribute to metastatic malignant cell implants and tumor-related angiogenesis, possibly abetted by the tissue factor that they carry. It is thought that the adherence of the EV to endothelium is dependent upon a combination of their P-selectin glycoprotein ligand-1 and exposed phosphatidylserine, the latter of which is normally hidden on the inner bilayer of the intact cellular membrane. This manuscript reviews what is known about EV origins, their clearance from the circulation and how they contribute to malignant cell implants upon endothelium surfaces and subsequent tumor growth.
Collapse
Affiliation(s)
- Murray M Bern
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud, Albuquerque, NM, 87131, USA.
| |
Collapse
|
32
|
Thrombo-hemorrhagic liability in children with congenital heart diseases. Hematol Oncol Stem Cell Ther 2017; 11:123-128. [PMID: 28867175 DOI: 10.1016/j.hemonc.2017.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/27/2017] [Accepted: 07/19/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The precise mechanisms of the increased incidence of hemostatic abnormalities in congenital heart disease (CHD) have not been determined. The aim of the study was to evaluate some indicators of activation of platelets and vascular endothelial cells in patients with CHD, evaluation of bleeding liability of these patients, and correlation with the clinical presentation of these patients. METHODS This work was carried out on 20 patients with cyanotic congenital heart diseases (CCHD), 20 patients with acyanotic congenital heart diseases (ACHD), and 20 healthy children who served as the control group, aged between 1 and 10years. All were subjected to full clinical examination, complete blood count, oxygen saturation, echocardiography, bleeding and coagulation times, PT, PTT, FDPs, plasma soluble P-selectin, E-selectin, and platelet factor 4 (PF4). RESULTS There was significant prolongation of PT and PTT, and there was a significant lowering of platelet counts. These results were obtained in CCHD and ACHD, but were more significant in CCHD patients. There was a significant elevation in PF4 (55.0±25.5ng/mL), P-selectin (128.9±42.44ng/dL), and E-selectin (9461.5±1701.24pg/mL) levels in children with CCHD as compared to those with ACHD (PF4, 21±7.94ng/mL; P-selectin, 80.1±13.2ng/mL; E-selectin, 7969.6±2127.5pg/mL), and significant increase in both groups when compared to the control group (PF4, 8.1±4.7ng/mL; P-selectin, 27.83±9.73ng/mL; E-selectin, 6750.00±3204.00pg/mL). There was a significant negative correlation between oxygen saturation, plasma P-selectin (r=-0.865), E-selectin (r=-0.401), and PF4 (r=-0.792) in patients with CCHD. CONCLUSION Patients with CHD-both cyanotic and acyanotic-have variable degrees of increased liability for both thrombosis and hemorrhage that represents some sort of adaptation to preserve hemostasis and to protect these patients against the clinical presentation of both thrombosis and bleeding. This is to say that CHD patients have their own point of balance between thrombogenicity and bleeding liability. Wide-scale studies are needed to detect the normal levels of different thrombohemorrhagic parameters of these patients.
Collapse
|
33
|
Jardim LL, Chaves DG, Silveira-Cassette ACO, Simões e Silva AC, Santana MP, Cerqueira MH, Prezotti A, Lorenzato C, Franco V, van der Bom JG, Rezende SM. Immune status of patients with haemophilia A before exposure to factor VIII: first results from the HEMFIL study. Br J Haematol 2017; 178:971-978. [DOI: 10.1111/bjh.14799] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/01/2017] [Indexed: 01/22/2023]
Affiliation(s)
- Letícia L. Jardim
- Faculty of Medicine; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
| | | | | | | | | | - Monica H. Cerqueira
- Instituto de Hematologia Arthur de Siqueira Cavalcanti (HEMORIO); Rio de Janeiro Brazil
| | - Alessandra Prezotti
- Centro de Hematologia e Hemoterapia do Espírito Santo (HEMOES); Vitória Espírito Santo Brazil
| | - Claudia Lorenzato
- Centro de Hematologia e Hemoterapia do Paraná (HEMEPAR); Curitiba Paraná Brazil
| | - Vivian Franco
- Centro de Hematologia e Hemoterapia de Santa Catarina (HEMOSC); Florianópolis Santa Catarina Brazil
| | - Johanna G. van der Bom
- Centre for Clinical Transfusion Research; Sanquin & Department of Clinical Epidemiology; Leiden University Medical Centre (LUMC); Leiden the Netherlands
| | - Suely M. Rezende
- Faculty of Medicine; Universidade Federal de Minas Gerais; Belo Horizonte Minas Gerais Brazil
| |
Collapse
|
34
|
The potential role of platelets in the consensus molecular subtypes of colorectal cancer. Cancer Metastasis Rev 2017; 36:273-288. [DOI: 10.1007/s10555-017-9678-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
35
|
Riedl J, Ay C, Pabinger I. Platelets and hemophilia: A review of the literature. Thromb Res 2017; 155:131-139. [DOI: 10.1016/j.thromres.2017.05.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 05/10/2017] [Accepted: 05/11/2017] [Indexed: 10/19/2022]
|
36
|
Lai JCY, Rocha-Ferreira E, Ek CJ, Wang X, Hagberg H, Mallard C. Immune responses in perinatal brain injury. Brain Behav Immun 2017; 63:210-223. [PMID: 27865947 DOI: 10.1016/j.bbi.2016.10.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/28/2016] [Accepted: 10/30/2016] [Indexed: 12/13/2022] Open
Abstract
The perinatal period has often been described as immune deficient. However, it has become clear that immune responses in the neonate following exposure to microbes or as a result of tissue injury may be substantial and play a role in perinatal brain injury. In this article we will review the immune cell composition under normal physiological conditions in the perinatal period, both in the human and rodent. We will summarize evidence of the inflammatory responses to stimuli and discuss how neonatal immune activation, both in the central nervous system and in the periphery, may contribute to perinatal hypoxic-ischemic brain injury.
Collapse
Affiliation(s)
- Jacqueline C Y Lai
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Eridan Rocha-Ferreira
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - C Joakim Ek
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Xiaoyang Wang
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Henrik Hagberg
- Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden
| | - Carina Mallard
- Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Box 432, 405 30 Gothenburg, Sweden.
| |
Collapse
|
37
|
The Interaction of Selectins and PSGL-1 as a Key Component in Thrombus Formation and Cancer Progression. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6138145. [PMID: 28680883 PMCID: PMC5478826 DOI: 10.1155/2017/6138145] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/12/2017] [Accepted: 04/23/2017] [Indexed: 12/11/2022]
Abstract
Cellular interaction is inevitable in the pathomechanism of human disease. Formation of heterotypic cellular aggregates, between distinct cells of hematopoietic and nonhematopoietic origin, may be involved in events leading to inflammation and the complex process of cancer progression. Among adhesion receptors, the family of selectins with their ligands have been considered as one of the major contributors to cell-cell interactions. Consequently, the inhibition of the interplay between selectins and their ligands may have potential therapeutic benefits. In this review, we focus on the current evidence on the selectins as crucial modulators of inflammatory, thrombotic, and malignant disorders. Knowing that there is promiscuity in selectin binding, we outline the importance of a key protein that serves as a ligand for all selectins. This dimeric mucin, the P-selectin glycoprotein ligand 1 (PSGL-1), has emerged as a major player in inflammation, thrombus, and cancer development. We discuss the interaction of PSGL-1 with various selectins in physiological and pathological processes with particular emphasis on mechanisms that lead to severe disease.
Collapse
|
38
|
Ridger VC, Boulanger CM, Angelillo-Scherrer A, Badimon L, Blanc-Brude O, Bochaton-Piallat ML, Boilard E, Buzas EI, Caporali A, Dignat-George F, Evans PC, Lacroix R, Lutgens E, Ketelhuth DFJ, Nieuwland R, Toti F, Tunon J, Weber C, Hoefer IE. Microvesicles in vascular homeostasis and diseases. Position Paper of the European Society of Cardiology (ESC) Working Group on Atherosclerosis and Vascular Biology. Thromb Haemost 2017; 117:1296-1316. [PMID: 28569921 DOI: 10.1160/th16-12-0943] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/27/2017] [Indexed: 12/15/2022]
Abstract
Microvesicles are members of the family of extracellular vesicles shed from the plasma membrane of activated or apoptotic cells. Microvesicles were initially characterised by their pro-coagulant activity and described as "microparticles". There is mounting evidence revealing a role for microvesicles in intercellular communication, with particular relevance to hemostasis and vascular biology. Coupled with this, the potential of microvesicles as meaningful biomarkers is under intense investigation. This Position Paper will summarise the current knowledge on the mechanisms of formation and composition of microvesicles of endothelial, platelet, red blood cell and leukocyte origin. This paper will also review and discuss the different methods used for their analysis and quantification, will underline the potential biological roles of these vesicles with respect to vascular homeostasis and thrombosis and define important themes for future research.
Collapse
Affiliation(s)
| | - Chantal M Boulanger
- Victoria Ridger, PhD, Department of Infection, Immunity and Cardiovascular Disease, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK, E-mail: , or, Chantal M. Boulanger, PhD, INSERM UMR-S 970, Paris Cardiovascular Research Center - PARCC, 56 rue Leblanc, 75015 Paris, France, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
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
|
40
|
Mosaad SM, Zaitone SA, Ahmed AAM, Abo-Elmatty DM, El-Baz AA, Moustafa YM. Evening primrose oil or forskolin ameliorates celecoxib-enhanced upregulation of tissue factor expression in mice subjected to lipopolysaccharide-induced endotoxemia. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:483-492. [PMID: 28124089 DOI: 10.1007/s00210-017-1342-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/12/2017] [Indexed: 11/30/2022]
Abstract
Celecoxib, a selective cyclooxygenase-2 inhibitor, produces thrombotic events in patients predisposed to cardiovascular risk factors. One theory reported an increase in endothelial expression of tissue factor (TF) as a predisposing factor. This work explored the effect of evening primrose oil (EPO), a source of prostaglandin E1, and forskolin (a cyclic adenosine monophosphate stimulator) against the prothrombotic effect of celecoxib in mice. Lipopolysaccharide mouse model of endotoxemia was used to induce an upregulation of TF activity. Male mice received celecoxib (25 mg/kg), celecoxib plus EPO, or celecoxib plus forskolin for 4 weeks and then subjected to a prothrombotic challenge in the form of an intraperitoneal injection of lipopolysaccharide. Results showed an increase in plasma TF activity, endothelial TF expression, and thrombin-antithrombin (TAT) but lower antithrombin III (ATIII) level in mice that received celecoxib in comparison to those that received the vehicle. Adding EPO or forskolin to celecoxib regimen significantly decreased the prothrombotic effect of celecoxib. A positive correlation (r = 0.8501) was found between TF activity and TAT. Co-administration of EPO or forskolin decreased the activity of TF and mitigated the prothrombotic effect of celecoxib. Therefore, these combinations may have the utility to abrogate the prothrombotic adverse effect of celecoxib in clinical setting.
Collapse
Affiliation(s)
- Sarah M Mosaad
- Department of Pharmaceutical Inspection, Ministry of Health, Ismailia, 41111, Egypt
| | - Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt. .,Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk City, Kingdom of Saudi Arabia.
| | - Amal A M Ahmed
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Dina M Abo-Elmatty
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Amani A El-Baz
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Yasser M Moustafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
41
|
Sun DS, Chang YW, Kau JH, Huang HH, Ho PH, Tzeng YJ, Chang HH. Soluble P-selectin rescues mice from anthrax lethal toxin-induced mortality through PSGL-1 pathway-mediated correction of hemostasis. Virulence 2017; 8:1216-1228. [PMID: 28102766 DOI: 10.1080/21505594.2017.1282027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
As one of the virulence factors of Bacillus anthracis, lethal toxin (LT) induces various pathogenic responses including the suppression of the coagulation system. In this study, we observed that LT markedly increased the circulating soluble P-selectin (sP-sel) levels and microparticle (MP) count in wild-type but not P-selectin (P-sel, Selp-/-) or P-sel ligand-1 (PSGL-1, Selplg-/-) knockout mice. Because sP-sel induces a hypercoagulable state through PSGL-1 pathway to generate tissue factor-positive MPs, we hypothesized that the increase in plasma sP-sel levels can be a self-rescue response in hosts against the LT-mediated suppression of the coagulation system. In agreement with our hypothesis, our results indicated that compared with wild-type mice, Selp-/- and Selplg-/- mice were more sensitive to LT. In addition, the recombinant sP-sel treatment markedly ameliorated LT-mediated pathogenesis and reduced mortality. As a result, elicitation of circulating sP-sel is potentially a self-rescue response, which is beneficial to host recovery from an LT-induced hypocoagulation state. These results suggest that the administration of sP-sel is likely to be useful in the development of a new strategy to treat anthrax.
Collapse
Affiliation(s)
- Der-Shan Sun
- a Department of Molecular Biology and Human Genetics , Tzu-Chi University , Hualien , Taiwan.,b Center for Vascular Medicine , Tzu-Chi University , Hualien , Taiwan
| | - Yao-Wen Chang
- a Department of Molecular Biology and Human Genetics , Tzu-Chi University , Hualien , Taiwan
| | - Jyh-Hwa Kau
- c Institute of Microbiology and Immunology, National Defense Medical Center , Taipei , Taiwan.,d Institute of Preventive Medicine, National Defense Medical Center , Taipei , Taiwan
| | - Hsin-Hsien Huang
- d Institute of Preventive Medicine, National Defense Medical Center , Taipei , Taiwan
| | - Pei-Hsun Ho
- a Department of Molecular Biology and Human Genetics , Tzu-Chi University , Hualien , Taiwan
| | - Yin-Jeh Tzeng
- a Department of Molecular Biology and Human Genetics , Tzu-Chi University , Hualien , Taiwan
| | - Hsin-Hou Chang
- a Department of Molecular Biology and Human Genetics , Tzu-Chi University , Hualien , Taiwan.,b Center for Vascular Medicine , Tzu-Chi University , Hualien , Taiwan
| |
Collapse
|
42
|
Pagel O, Walter E, Jurk K, Zahedi RP. Taking the stock of granule cargo: Platelet releasate proteomics. Platelets 2016; 28:119-128. [PMID: 27928935 DOI: 10.1080/09537104.2016.1254762] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human platelets are key players in a multitude of physiological and pathological processes. Upon activation they release cargo from different types of granules as well as microparticles in an apparently well-regulated and orchestrated manner. The resulting specific platelet releasates create microenvironments of biologically active compounds and proteins during platelet aggregation and thrombus formation, allowing efficient delivery of growth factors and immune modulators to their sites of effect and enhancing the coagulative response in a positive feedback loop. Thus, platelet releasates play a central role in the regulation of platelet homeostasis and heterotypic cell interaction. Additionally, it recently emerged that both the qualitative and quantitative composition of the releasate as well as release dynamics may be stimulus dependent and therefore more complex than expected. Mass spectrometry-based proteomics is an important asset for studying platelet releasates in vitro, as it allows not only (i) identifying released proteins, but moreover (ii) determining their quantities and the dynamics of release as well as (iii) differentially comparing releasates across a variety of conditions. Though owing to the high sensitivity and comprehensiveness of modern proteomic techniques, a thorough experimental design and a standardized and robust sample preparation are essential to obtain highly confident and reliable insights into platelet biology and pathology. Here, we review releasate proteome studies and crucial sample preparation strategies to summarize possible achievements of state-of-the-art technologies and furthermore discuss potential pitfalls and limitations. We provide a future perspective of platelet releasate proteomics including targeted analyses, post-translational modifications and multi-omics approaches that should be adopted by platelet releasate researchers due to their tremendous depth and comprehensiveness.
Collapse
Affiliation(s)
- Oliver Pagel
- a Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V ., Dortmund , Germany
| | - Elena Walter
- b Center for Thrombosis and Hemostasis (CTH) , Universitätsklinikum der Johannes Gutenberg-Universität Mainz , Mainz , Germany
| | - Kerstin Jurk
- b Center for Thrombosis and Hemostasis (CTH) , Universitätsklinikum der Johannes Gutenberg-Universität Mainz , Mainz , Germany
| | - René P Zahedi
- a Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V ., Dortmund , Germany
| |
Collapse
|
43
|
Abstract
Platelets are small, anucleate circulating cells that possess a dynamic repertoire of functions spanning the hemostatic, inflammatory, and immune continuum. Once thought to be merely cell fragments with responses limited primarily to acute hemostasis and vascular wall repair, platelets are now increasingly recognized as key sentinels and effector cells regulating host responses to many inflammatory and infectious cues. Platelet granules, including α-granules and dense-granules, store hundreds of factors and secrete these mediators in response to activating signals. The cargo packaged and stored within platelet granules orchestrates communication between platelets and other circulating cells, augments host defense mechanisms to invading pathogens and tumor cells, and - in some settings - drives dysregulated and injurious responses. This focused review will highlight several of the established and emerging mechanisms and roles of platelet secretion in inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Bhanu K Manne
- a The University of Utah Molecular Medicine Program , Salt Lake City , Utah , USA
| | | | - Matthew T Rondina
- a The University of Utah Molecular Medicine Program , Salt Lake City , Utah , USA.,c Department of Internal Medicine , Salt Lake City , Utah , USA.,d The GRECC, George E. Wahlen Salt Lake City VAMC , Salt Lake City , Utah , USA
| |
Collapse
|
44
|
Sun DS, Ho PH, Chang HH. Soluble P-selectin rescues viper venom-induced mortality through anti-inflammatory properties and PSGL-1 pathway-mediated correction of hemostasis. Sci Rep 2016; 6:35868. [PMID: 27779216 PMCID: PMC5078805 DOI: 10.1038/srep35868] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/03/2016] [Indexed: 01/24/2023] Open
Abstract
Venomous snakebites are lethal and occur frequently worldwide each year, and receiving the antivenom antibody is currently the most effective treatment. However, the specific antivenom might be unavailable in remote areas. Snakebites by Viperidae usually lead to hemorrhage and mortality if untreated. In the present study, challenges of rattlesnake (Crotalus atrox) venom markedly increased the circulating soluble P-selectin (sP-sel) level, but not P-selectin (P-sel, Selp−/−) mutants, in wild-type mice. Because sP-sel enhances coagulation through the P-selectin ligand 1 (PSGL-1, Selplg) pathway to produce tissue factor–positive microparticles, we hypothesized that increasing the plasma sP-sel level can be a self-rescue response in hosts against snake venom–mediated suppression of the coagulation system. Confirming our hypothesis, our results indicated that compared with wild-type mice, Selp−/− and Selplg−/− mice were more sensitive to rattlesnake venom. Additionally, administration of recombinant sP-sel could effectively reduce the mortality rate of mice challenged with venoms from three other Viperidae snakes. The antivenom property of sP-sel is associated with improved coagulation activity in vivo. Our data suggest that the elevation of endogenous sP-sel level is a self-protective response against venom-suppressed coagulation. The administration of recombinant sP-sel may be developed as a new strategy to treat Viperidae snakebites.
Collapse
Affiliation(s)
- Der-Shan Sun
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan.,Center for Vascular Medicine, Tzu-Chi University, Hualien 970, Taiwan
| | - Pei-Hsun Ho
- Center for Vascular Medicine, Tzu-Chi University, Hualien 970, Taiwan
| | - Hsin-Hou Chang
- Department of Molecular Biology and Human Genetics, Tzu-Chi University, Hualien 970, Taiwan.,Center for Vascular Medicine, Tzu-Chi University, Hualien 970, Taiwan
| |
Collapse
|
45
|
Żmigrodzka M, Guzera M, Miśkiewicz A, Jagielski D, Winnicka A. The biology of extracellular vesicles with focus on platelet microparticles and their role in cancer development and progression. Tumour Biol 2016; 37:14391-14401. [PMID: 27629289 PMCID: PMC5126185 DOI: 10.1007/s13277-016-5358-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 09/07/2016] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of structures which can be classified into smaller in size and relatively homogenous exosomes (EXSMs)—spherical fragments of lipid bilayers from inner cell compartments—and bigger in size ectosomes (ECSMs)—a direct consequence of cell-membrane blebbing. EVs can be found in body fluids of healthy individuals. Their number increases in cancer and other pathological conditions. EVs can originate from various cell types, including leukocytes, erythrocytes, thrombocytes, and neoplastic cells. Platelet microparticles (PMPs) are the most abundant population of EVs in blood. It is well documented that PMPs, being a crucial element of EVs signaling, are involved in tumor growth, metastasis, and angiogenesis and may participate in the development of multidrug resistance by tumor cells. The aim of this review is to present the role of PMPs in carcinogenesis. The biology and functions of PMPs with a particular emphasis on the most recent scientific reports on EV properties are also characterized.
Collapse
Affiliation(s)
- M Żmigrodzka
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, Warsaw, Poland
| | - M Guzera
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - A Miśkiewicz
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, Warsaw, Poland.
| | - D Jagielski
- Veterinary Clinic BIALOBRZESKA, Częstochowska 20, Warsaw, Poland
| | - A Winnicka
- Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences (WULS-SGGW), Nowoursynowska 159c, Warsaw, Poland
| |
Collapse
|
46
|
Xu XR, Zhang D, Oswald BE, Carrim N, Wang X, Hou Y, Zhang Q, Lavalle C, McKeown T, Marshall AH, Ni H. Platelets are versatile cells: New discoveries in hemostasis, thrombosis, immune responses, tumor metastasis and beyond. Crit Rev Clin Lab Sci 2016; 53:409-30. [PMID: 27282765 DOI: 10.1080/10408363.2016.1200008] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Platelets are small anucleate blood cells generated from megakaryocytes in the bone marrow and cleared in the reticuloendothelial system. At the site of vascular injury, platelet adhesion, activation and aggregation constitute the first wave of hemostasis. Blood coagulation, which is initiated by the intrinsic or extrinsic coagulation cascades, is the second wave of hemostasis. Activated platelets can also provide negatively-charged surfaces that harbor coagulation factors and markedly potentiate cell-based thrombin generation. Recently, deposition of plasma fibronectin, and likely other plasma proteins, onto the injured vessel wall has been identified as a new "protein wave of hemostasis" that may occur even earlier than the first wave of hemostasis, platelet accumulation. Although no experimental evidence currently exists, it is conceivable that platelets may also contribute to this protein wave of hemostasis by releasing their granule fibronectin and other proteins that may facilitate fibronectin self- and non-self-assembly on the vessel wall. Thus, platelets may contribute to all three waves of hemostasis and are central players in this critical physiological process to prevent bleeding. Low platelet counts in blood caused by enhanced platelet clearance and/or impaired platelet production are usually associated with hemorrhage. Auto- and allo-immune thrombocytopenias such as idiopathic thrombocytopenic purpura and fetal and neonatal alloimmune thrombocytopenia may cause life-threatening bleeding such as intracranial hemorrhage. When triggered under pathological conditions such as rupture of an atherosclerotic plaque, excessive platelet activation and aggregation may result in thrombosis and vessel occlusion. This may lead to myocardial infarction or ischemic stroke, the major causes of mortality and morbidity worldwide. Platelets are also involved in deep vein thrombosis and thromboembolism, another leading cause of mortality. Although fibrinogen has been documented for more than half a century as essential for platelet aggregation, recent studies demonstrated that fibrinogen-independent platelet aggregation occurs in both gene deficient animals and human patients under physiological and pathological conditions (non-anti-coagulated blood). This indicates that other unidentified platelet ligands may play important roles in thrombosis and might be novel antithrombotic targets. In addition to their critical roles in hemostasis and thrombosis, emerging evidence indicates that platelets are versatile cells involved in many other pathophysiological processes such as innate and adaptive immune responses, atherosclerosis, angiogenesis, lymphatic vessel development, liver regeneration and tumor metastasis. This review summarizes the current knowledge of platelet biology, highlights recent advances in the understanding of platelet production and clearance, molecular and cellular events of thrombosis and hemostasis, and introduces the emerging roles of platelets in the immune system, vascular biology and tumorigenesis. The clinical implications of these basic science and translational research findings will also be discussed.
Collapse
Affiliation(s)
- Xiaohong Ruby Xu
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada .,b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,c Department of Medicine , Guangzhou University of Chinese Medicine , Guangzhou , Guangdong , P.R. China
| | - Dan Zhang
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,c Department of Medicine , Guangzhou University of Chinese Medicine , Guangzhou , Guangdong , P.R. China
| | - Brigitta Elaine Oswald
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,d Canadian Blood Services , Toronto , ON , Canada .,e Department of Physiology , University of Toronto , Toronto , ON , Canada
| | - Naadiya Carrim
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada .,b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,d Canadian Blood Services , Toronto , ON , Canada
| | - Xiaozhong Wang
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,f The Second Affiliated Hospital of Nanchang University , Nanchang , Jiangxi , P.R. China
| | - Yan Hou
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,g Jilin Provincial Center for Disease Prevention and Control , Changchun , Jilin , P.R. China
| | - Qing Zhang
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,h State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University , Guangzhou , Guangdong , P.R. China , and
| | - Christopher Lavalle
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,e Department of Physiology , University of Toronto , Toronto , ON , Canada
| | - Thomas McKeown
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada
| | - Alexandra H Marshall
- b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada
| | - Heyu Ni
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , ON , Canada .,b Department of Laboratory Medicine , Keenan Research Centre for Biomedical Science, St. Michael's Hospital, and Toronto Platelet Immunobiology Group, Li Ka Shing Knowledge Institute , Toronto , ON , Canada .,d Canadian Blood Services , Toronto , ON , Canada .,e Department of Physiology , University of Toronto , Toronto , ON , Canada .,i Department of Medicine , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
47
|
Abstract
During apoptosis or activation, cells can release a subcellular structure, called a membrane microvesicle (also known as microparticle) into the extracellular environment. Microvesicles bud-off as a portion of cell membrane with its associated proteins and lipids surrounding a cytosolic core that contains intracellular proteins, lipids, and nucleic acids (DNA, RNA, siRNA, microRNA, lncRNA). Biologically active molecules on the microvesicle surface and encapsulated within can act on recipient cells as a novel mode of intercellular communication. Apoptosis has long been known to be involved in the development of diseases of autoimmunity. Abnormally persistent microvesicles, particularly apoptotic microvesicles, can accelerate autoimmune responses locally in specific organs and tissues as well as systemically. In this review, we focus on studies implicating microvesicles in the pathogenesis of autoimmune diseases and their complications.
Collapse
|
48
|
Microparticles variability in fresh frozen plasma: preparation protocol and storage time effects. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2016; 14:228-37. [PMID: 27136430 DOI: 10.2450/2016.0179-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 10/20/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND Extracellular vesicles or microparticles exhibiting procoagulant and thrombogenic activity may contribute to the haemostatic potential of fresh frozen plasma. MATERIALS AND METHODS Fresh frozen plasma was prepared from platelet-rich plasma at 20 °C (Group-1 donors) or directly from whole blood at 4 °C (Group-2 donors). Each unit was aseptically divided into three parts, stored frozen for specific periods of time, and analysed by flow cytometry for procoagulant activity immediately after thaw or following post-thaw storage for 24 h at 4 °C. Donors' haematologic, biochemical and life-style profiles as well as circulating microparticles were analysed in parallel. RESULTS Circulating microparticles exhibited a considerable interdonor but not intergroup variation. Fresh frozen plasma units were enriched in microparticles compared to plasma in vivo. Duration of storage significantly affected platelet- and red cell-derived microparticles. Fresh frozen plasma prepared directly from whole blood contained more residual platelets and more platelet-derived microparticles compared to fresh frozen plasma prepared from platelet-rich plasma. Consequently, there was a statistically significant difference in total, platelet- and red cell-derived microparticles between the two preparation protocols over storage time in the freezer. Preservation of the thawed units for 24 h at 4 °C did not significantly alter microparticle accumulation. Microparticle accumulation and anti-oxidant capacity of fresh frozen plasma was positively or negatively correlated, respectively, with the level of circulating microparticles in individual donors. DISCUSSION The preparation protocol and the duration of storage in the freezer, independently and in combination, influenced the accumulation of microparticles in fresh frozen plasma units. In contrast, storage of thawed units for 24 h at 4 °C had no significant effect on the concentration of microparticles.
Collapse
|
49
|
Tempo JA, Englyst NA, Holloway JA, Smith DC. Platelet Microvesicles (Microparticles) in Cardiac Surgery. J Cardiothorac Vasc Anesth 2016; 30:222-8. [DOI: 10.1053/j.jvca.2015.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Indexed: 11/11/2022]
|
50
|
Zubairova LD, Nabiullina RM, Nagaswami C, Zuev YF, Mustafin IG, Litvinov RI, Weisel JW. Circulating Microparticles Alter Formation, Structure, and Properties of Fibrin Clots. Sci Rep 2015; 5:17611. [PMID: 26635081 PMCID: PMC4669434 DOI: 10.1038/srep17611] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/03/2015] [Indexed: 01/21/2023] Open
Abstract
Despite the importance of circulating microparticles in haemostasis and thrombosis, there is limited evidence for potential causative effects of naturally produced cell-derived microparticles on fibrin clot formation and its properties. We studied the significance of blood microparticles for fibrin formation, structure, and susceptibility to fibrinolysis by removing them from platelet-free plasma using filtration. Clots made in platelet-free and microparticle-depleted plasma samples from the same healthy donors were analyzed in parallel. Microparticles accelerate fibrin polymerisation and support formation of more compact clots that resist internal and external fibrinolysis. These variations correlate with faster thrombin generation, suggesting thrombin-mediated kinetic effects of microparticles on fibrin formation, structure, and properties. In addition, clots formed in the presence of microparticles, unlike clots from the microparticle-depleted plasma, contain 0.1-0.5-μm size granular and CD61-positive material on fibres, suggesting that platelet-derived microparticles attach to fibrin. Therefore, the blood of healthy individuals contains functional microparticles at the levels that have a procoagulant potential. They affect the structure and stability of fibrin clots indirectly through acceleration of thrombin generation and through direct physical incorporation into the fibrin network. Both mechanisms underlie a potential role of microparticles in haemostasis and thrombosis as modulators of fibrin formation, structure, and resistance to fibrinolysis.
Collapse
Affiliation(s)
- Laily D Zubairova
- Department of General Pathology, Kazan State Medical University, Kazan 420012, Russian Federation
| | - Roza M Nabiullina
- Department of General Pathology, Kazan State Medical University, Kazan 420012, Russian Federation
| | - Chandrasekaran Nagaswami
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Yuriy F Zuev
- Institute of Biochemistry and Biophysics, Russian Academy of Sciences, Kazan 420111, Russian Federation
| | - Ilshat G Mustafin
- Department of General Pathology, Kazan State Medical University, Kazan 420012, Russian Federation
| | - Rustem I Litvinov
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420012, Russian Federation
| | - John W Weisel
- Department of Cell and Developmental Biology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|