151
|
Ghasemzadeh M, Ahmadi J, Hosseini E. Platelet-leukocyte crosstalk in COVID-19: How might the reciprocal links between thrombotic events and inflammatory state affect treatment strategies and disease prognosis? Thromb Res 2022; 213:179-194. [PMID: 35397313 PMCID: PMC8969450 DOI: 10.1016/j.thromres.2022.03.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/11/2022] [Accepted: 03/28/2022] [Indexed: 01/09/2023]
Abstract
Platelet-leukocyte crosstalk is commonly manifested by reciprocal links between thrombosis and inflammation. Platelet thrombus acts as a reactive matrix that recruits leukocytes to the injury site where their massive accumulation, activation and migration promote thrombotic events while triggering inflammatory responses. As a life-threatening condition with the associations between inflammation and thrombosis, COVID-19 presents diffuse alveolar damage due to exaggerated macrophage activity and cytokine storms. These events, together with direct intracellular virus invasion lead to pulmonary vascular endothelialitis, cell membranes disruption, severe endothelial injury, and thrombosis. The developing pre-alveolar thrombus provides a hyper-reactive milieu that recruits circulating leukocytes to the injury site where their activation contributes to thrombus stabilization and thrombosis propagation, primarily through the formation of Neutrophil extracellular trap (NET). NET fragments can also circulate and deposit in further distance where they may disseminate intravascular thrombosis in severe cases of disease. Thrombi may also facilitate leukocytes migration into alveoli where their accumulation and activation exacerbate cytokine storms and tissue damage, further complicating the disease. Based on these mechanisms, whether an effective anti-inflammatory protocol can prevent thrombotic events, or on the other hand; efficient antiplatelet or anticoagulant regimens may be associated with reduced cytokine storms and tissue damage, is now of interests for several ongoing researches. Thus shedding more light on platelet-leukocyte crosstalk, the review presented here discusses the detailed mechanisms by which platelets may contribute to the pathogenesis of COVID-19, especially in severe cases where their interaction with leukocytes can intensify both inflammatory state and thrombosis in a reciprocal manner.
Collapse
Affiliation(s)
- Mehran Ghasemzadeh
- Corresponding authors at: Blood Transfusion Research Centre, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Exp. Way, Next to the Milad Tower, Tehran, Iran
| | | | - Ehteramolsadat Hosseini
- Corresponding authors at: Blood Transfusion Research Centre, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Hemmat Exp. Way, Next to the Milad Tower, Tehran, Iran
| |
Collapse
|
152
|
Vollbracht C, Kraft K. Oxidative Stress and Hyper-Inflammation as Major Drivers of Severe COVID-19 and Long COVID: Implications for the Benefit of High-Dose Intravenous Vitamin C. Front Pharmacol 2022; 13:899198. [PMID: 35571085 PMCID: PMC9100929 DOI: 10.3389/fphar.2022.899198] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/14/2022] [Indexed: 12/25/2022] Open
Abstract
Oxidative stress is a pivotal point in the pathophysiology of COVID-19 and presumably also in Long-COVID. Inflammation and oxidative stress are mutually reinforcing each other, thus contributing to the systemic hyperinflammatory state and coagulopathy which are cardinal pathological mechanisms of severe stages. COVID-19 patients, like other critically ill patients e.g. with pneumonia, very often show severe deficiency of the antioxidant vitamin C. So far, it has not been investigated how long this deficiency lasts or whether patients with long COVID symptoms also suffer from deficiencies. A vitamin C deficit has serious pathological consequences because vitamin C is one of the most effective antioxidants, but also co-factor of many enzymatic processes that affect the immune and nervous system, blood circulation and energy metabolism. Because of its anti-oxidative, anti-inflammatory, endothelial-restoring, and immunomodulatory effects the supportive intravenous (iv) use of supraphysiological doses has been investigated so far in 12 controlled or observational studies with altogether 1578 inpatients with COVID-19. In these studies an improved oxygenation, a decrease in inflammatory markers and a faster recovery were observed. In addition, early treatment with iv high dose vitamin C seems to reduce the risks of severe courses of the disease such as pneumonia and also mortality. Persistent inflammation, thrombosis and a dysregulated immune response (auto-immune phenomena and/or persistent viral load) seem to be major contributors to Long-COVID. Oxidative stress and inflammation are involved in the development and progression of fatigue and neuro-psychiatric symptoms in various diseases by disrupting tissue (e.g. autoantibodies), blood flow (e.g. immune thrombosis) and neurotransmitter metabolism (e.g. excitotoxicity). In oncological diseases, other viral infections and autoimmune diseases, which are often associated with fatigue, cognitive disorders, pain and depression similar to Long-COVID, iv high dose vitamin C was shown to significantly relieve these symptoms. Supportive iv vitamin C in acute COVID-19 might therefore reduce the risk of severe courses and also the development of Long-COVID.
Collapse
Affiliation(s)
- Claudia Vollbracht
- Medical Science Department, Pascoe Pharmazeutische Präparate GmbH, Giessen, Germany
| | - Karin Kraft
- Chair of Naturopathy, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
153
|
Nappi F, Bellomo F, Avtaar Singh SS. Insights into the Role of Neutrophils and Neutrophil Extracellular Traps in Causing Cardiovascular Complications in Patients with COVID-19: A Systematic Review. Biomedicines 2022; 11:2460. [PMID: 35566589 PMCID: PMC9855935 DOI: 10.3390/biomedicines11010113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic caused by the SARS-CoV-2 virus has resulted in significant mortality and burdening of healthcare resources. While initially noted as a pulmonary pathology, subsequent studies later identified cardiovascular involvement with high mortalities reported in specific cohorts of patients. While cardiovascular comorbidities were identified early on, the exact manifestation and etiopathology of the infection remained elusive. This systematic review aims to investigate the role of inflammatory pathways, highlighting several culprits including neutrophil extracellular traps (NETs) which have since been extensively investigated. METHOD A search was conducted using three databases (MEDLINE; MEDLINE In-Process & Other Non-Indexed Citations and EMBASE). Data from randomized controlled trials (RCT), prospective series, meta-analyses, and unmatched observational studies were considered for the processing of the algorithm and treatment of inflammatory response during SARS-CoV-2 infection. Studies without the SARS-CoV-2 Infection period and case reports were excluded. RESULTS A total of 47 studies were included in this study. The role of the acute inflammatory response in the propagation of the systemic inflammatory sequelae of the disease plays a major part in determining outcomes. Some of the mechanisms of activation of these pathways have been highlighted in previous studies and are highlighted. CONCLUSION NETs play a pivotal role in the pathogenesis of the inflammatory response. Despite moving into the endemic phase of the disease in most countries, COVID-19 remains an entity that has not been fully understood with long-term effects remaining uncertain and requiring ongoing monitoring and research.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord of Saint-Denis, 93200 Saint-Denis, France
| | - Francesca Bellomo
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | | |
Collapse
|
154
|
Prével R, Dupont A, Labrouche-Colomer S, Garcia G, Dewitte A, Rauch A, Goutay J, Caplan M, Jozefowicz E, Lanoix JP, Poissy J, Rivière E, Orieux A, Malvy D, Gruson D, Garçon L, Susen S, James C. Plasma Markers of Neutrophil Extracellular Trap Are Linked to Survival but Not to Pulmonary Embolism in COVID-19-Related ARDS Patients. Front Immunol 2022; 13:851497. [PMID: 35371025 PMCID: PMC8968169 DOI: 10.3389/fimmu.2022.851497] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/23/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction Coronavirus disease 2019 (COVID-19) can cause life-threatening acute respiratory distress syndrome (ARDS). Recent data suggest a role for neutrophil extracellular traps (NETs) in COVID-19-related lung damage partly due to microthrombus formation. Besides, pulmonary embolism (PE) is frequent in severe COVID-19 patients, suggesting that immunothrombosis could also be responsible for increased PE occurrence in these patients. Here, we evaluate whether plasma levels of NET markers measured shorty after admission of hospitalized COVID-19 patients are associated with clinical outcomes in terms of clinical worsening, survival, and PE occurrence. Patients and Methods Ninety-six hospitalized COVID-19 patients were included, 50 with ARDS (severe disease) and 46 with moderate disease. We collected plasma early after admission and measured 3 NET markers: total DNA, myeloperoxidase (MPO)–DNA complexes, and citrullinated histone H3. Comparisons between survivors and non-survivors and patients developing PE and those not developing PE were assessed by Mann–Whitney test. Results Analysis in the whole population of hospitalized COVID-19 patients revealed increased circulating biomarkers of NETs in patients who will die from COVID-19 and in patients who will subsequently develop PE. Restriction of our analysis in the most severe patients, i.e., the ones who enter the hospital for COVID-19-related ARDS, confirmed the link between NET biomarker levels and survival but not PE occurrence. Conclusion Our results strongly reinforce the hypothesis that NETosis is an attractive therapeutic target to prevent COVID-19 progression but that it does not seem to be linked to PE occurrence in patients hospitalized with COVID-19.
Collapse
Affiliation(s)
- Renaud Prével
- CHU Bordeaux, Medical Intensive Care Unit, Pessac, France.,Univ. Bordeaux, INSERM, U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Pessac, France
| | - Annabelle Dupont
- Univ. Lille, INSERM, CHU Lille, Department of Hematology and Transfusion, Pôle de Biologie Pathologie Génétique, Institut Pasteur de Lille, UMR1011-EGID, Lille, France
| | - Sylvie Labrouche-Colomer
- Univ. Bordeaux, INSERM, UMR1034, Biology of Cardiovascular Diseases, Pessac, France.,CHU Bordeaux, Laboratory of Hematology, Pessac, France
| | - Geoffrey Garcia
- Laboratoire d'Hématologie, CHU Amiens, EA4666 HEMATIM, UPJV, Amiens, France
| | - Antoine Dewitte
- CHU Bordeaux, Department of Anaesthesia and Critical Care, Magellan Medico-Surgical Centre, Bordeaux, France.,Univ. Bordeaux, CNRS, UMR 5164, INSERM ERL1303, Immunology from Concept and Experiments to Translation (ImmunoConcEpT), Bordeaux, France
| | - Antoine Rauch
- Univ. Lille, INSERM, CHU Lille, Department of Hematology and Transfusion, Pôle de Biologie Pathologie Génétique, Institut Pasteur de Lille, UMR1011-EGID, Lille, France
| | - Julien Goutay
- Centre Hospitalier Universitaire Lille, Intensive Care Department, Pôle de Réanimation, Lille, France
| | - Morgan Caplan
- Centre Hospitalier Universitaire Lille, Intensive Care Department, Pôle de Réanimation, Lille, France
| | - Elsa Jozefowicz
- Centre Hospitalier Universitaire Lille, Surgical Critical Care, Department of Anesthesiology and Critical Care, Lille, France
| | - Jean-Philippe Lanoix
- CHU Amiens-Picardie, Infectious Diseases Department, Amiens, France.,EA4294, Université Picardie Jules Verne, Amiens, France
| | - Julien Poissy
- Univ. Lille, INSERM U1285, CHU Lille, Pôle de réanimation, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Etienne Rivière
- Univ. Bordeaux, INSERM, UMR1034, Biology of Cardiovascular Diseases, Pessac, France.,CHU Bordeaux, Internal Medicine and Infectious Diseases Unit, Pessac, France
| | - Arthur Orieux
- CHU Bordeaux, Medical Intensive Care Unit, Pessac, France
| | - Denis Malvy
- Department for Infectious and Tropical diseases, University Hospital Centre and INSERM 1219, University of Bordeaux, Bordeaux, France
| | - Didier Gruson
- CHU Bordeaux, Medical Intensive Care Unit, Pessac, France.,Univ. Bordeaux, INSERM, U1045, Centre de Recherche Cardio-Thoracique de Bordeaux, Pessac, France
| | - Loic Garçon
- Laboratoire d'Hématologie, CHU Amiens, EA4666 HEMATIM, UPJV, Amiens, France
| | - Sophie Susen
- Univ. Lille, INSERM, CHU Lille, Department of Hematology and Transfusion, Pôle de Biologie Pathologie Génétique, Institut Pasteur de Lille, UMR1011-EGID, Lille, France
| | - Chloé James
- Univ. Bordeaux, INSERM, UMR1034, Biology of Cardiovascular Diseases, Pessac, France.,CHU Bordeaux, Laboratory of Hematology, Pessac, France
| |
Collapse
|
155
|
Thirugnanasambandham I, Radhakrishnan A, Kuppusamy G, Kumar Singh S, Dua K. PEPTIDYLARGININE DEIMINASE-4: MEDICO-FORMULATIVE STRATEGY TOWARDS MANAGEMENT OF RHEUMATOID ARTHRITIS. Biochem Pharmacol 2022; 200:115040. [DOI: 10.1016/j.bcp.2022.115040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/08/2022] [Accepted: 04/08/2022] [Indexed: 12/15/2022]
|
156
|
Wu TH, Hsieh SC, Li TH, Lu CH, Liao HT, Shen CY, Li KJ, Wu CH, Kuo YM, Tsai CY, Yu CL. Molecular Basis for Paradoxical Activities of Polymorphonuclear Neutrophils in Inflammation/Anti-Inflammation, Bactericide/Autoimmunity, Pro-Cancer/Anticancer, and Antiviral Infection/SARS-CoV-II-Induced Immunothrombotic Dysregulation. Biomedicines 2022; 10:biomedicines10040773. [PMID: 35453523 PMCID: PMC9032061 DOI: 10.3390/biomedicines10040773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 02/06/2023] Open
Abstract
Polymorphonuclear neutrophils (PMNs) are the most abundant white blood cells in the circulation. These cells act as the fast and powerful defenders against environmental pathogenic microbes to protect the body. In addition, these innate inflammatory cells can produce a number of cytokines/chemokines/growth factors for actively participating in the immune network and immune homeostasis. Many novel biological functions including mitogen-induced cell-mediated cytotoxicity (MICC) and antibody-dependent cell-mediated cytotoxicity (ADCC), exocytosis of microvesicles (ectosomes and exosomes), trogocytosis (plasma membrane exchange) and release of neutrophil extracellular traps (NETs) have been successively discovered. Furthermore, recent investigations unveiled that PMNs act as a double-edged sword to exhibit paradoxical activities on pro-inflammation/anti-inflammation, antibacteria/autoimmunity, pro-cancer/anticancer, antiviral infection/COVID-19-induced immunothrombotic dysregulation. The NETs released from PMNs are believed to play a pivotal role in these paradoxical activities, especially in the cytokine storm and immunothrombotic dysregulation in the recent SARS-CoV-2 pandemic. In this review, we would like to discuss in detail the molecular basis for these strange activities of PMNs.
Collapse
Affiliation(s)
- Tsai-Hung Wu
- Division of Nephrology, Taipei Veterans General Hospital, National Yang-Ming Chiao-Tung University, Taipei 11217, Taiwan;
| | - Song-Chou Hsieh
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
| | - Tsu-Hao Li
- Division of Allergy, Immunology and Rheumatology, Shin Kong Wu Ho Shi Hospital, Taipei 11101, Taiwan;
- Institute of Clinical Medicine, National Yang-Ming Chiao-Tung University, Taipei 11217, Taiwan
| | - Cheng-Hsun Lu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
- Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Hsien-Tzung Liao
- Division of Allergy, Immunology and Rheumatology, Taipei Veterans General Hospital, National Yang-Ming Chiao-Tung University, Taipei 11217, Taiwan;
| | - Chieh-Yu Shen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
- Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Ko-Jen Li
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
| | - Cheng-Han Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
- Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Yu-Min Kuo
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
- Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 10002, Taiwan
| | - Chang-Youh Tsai
- Division of Allergy, Immunology and Rheumatology, Taipei Veterans General Hospital, National Yang-Ming Chiao-Tung University, Taipei 11217, Taiwan;
- Correspondence: (C.-Y.T.); (C.-L.Y.)
| | - Chia-Li Yu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan; (S.-C.H.); (C.-H.L.); (C.-Y.S.); (K.-J.L.); (C.-H.W.); (Y.-M.K.)
- Correspondence: (C.-Y.T.); (C.-L.Y.)
| |
Collapse
|
157
|
Cheng KH, Contreras GP, Yeh TY. Potential Role of Neutrophil Extracellular Traps in Cardio-Oncology. Int J Mol Sci 2022; 23:ijms23073573. [PMID: 35408933 PMCID: PMC8998890 DOI: 10.3390/ijms23073573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular toxicity has emerged as the leading cause of death in patients undergoing cancer treatment. Thus, cardio-oncology (CO) care must also focus on the prevention and management of related cardiovascular (CV) complications caused by cancer therapy. Neutrophil extracellular traps (NETs)—entities with released DNA, proteases, proinflammatory and prooxidative substances from blasted neutrophils—play an important role in cancer proliferation, propagation metastasis, and incident CV events (acute coronary syndrome, thromboembolic events, and heart failure). Although NETs have been shown to be involved in cancer progression and incident CV events, little is known about their relationship with cardio-oncology, especially on cancer treatment-related cardiovascular toxicity (CTRCT). This review aims to explore the evidence of the impact of NETs on cancer, CV events, and CTRCT, and the possible solutions based on the mechanism of NETs activation and NETs released toxic substances.
Collapse
Affiliation(s)
- Kai-Hung Cheng
- Division of Cardiology, Department of Internal Medicine, E-Da Cancer Hospital, Kaohsiung 82445, Taiwan;
- College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Gregory P. Contreras
- Auxergen Inc., Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, USA;
| | - Ting-Yu Yeh
- Auxergen Inc., Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, USA;
- Correspondence:
| |
Collapse
|
158
|
Cedervall J, Herre M, Dragomir A, Rabelo-Melo F, Svensson A, Thålin C, Rosell A, Hjalmar V, Wallén H, Lindman H, Pejler G, Hagström E, Hultström M, Larsson A, Olsson AK. Neutrophil extracellular traps promote cancer-associated inflammation and myocardial stress. Oncoimmunology 2022; 11:2049487. [PMID: 35309730 PMCID: PMC8928831 DOI: 10.1080/2162402x.2022.2049487] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cancer is associated with systemic pathologies that contribute to mortality, such as thrombosis and distant organ failure. The aim of this study was to investigate the potential role of neutrophil extracellular traps (NETs) in myocardial inflammation and tissue damage in treatment-naïve individuals with cancer. Mice with mammary carcinoma (MMTV-PyMT) had increased plasma levels of NETs measured as H3Cit-DNA complexes, paralleled with elevated coagulation, compared to healthy littermates. MMTV-PyMT mice displayed upregulation of pro-inflammatory markers in the heart, myocardial hypertrophy and elevated cardiac disease biomarkers in the blood, but not echocardiographic heart failure. Moreover, increased endothelial proliferation was observed in hearts from tumor-bearing mice. Removal of NETs by DNase I treatment suppressed the myocardial inflammation, expression of cardiac disease biomarkers and endothelial proliferation. Compared to a healthy control group, treatment-naïve cancer patients with different malignant disorders had increased NET formation, which correlated to plasma levels of the inflammatory marker CRP and the cardiac disease biomarkers NT-proBNP and sTNFR1, in agreement with the mouse data. Altogether, our data indicate that NETs contribute to inflammation and myocardial stress during malignancy. These findings suggest NETs as potential therapeutic targets to prevent cardiac inflammation and dysfunction in cancer patients.
Collapse
Affiliation(s)
- J. Cedervall
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - M. Herre
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - A. Dragomir
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - F. Rabelo-Melo
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - A. Svensson
- Department of Medical Cell Biology, Integrative Physiology, Uppsala University, Uppsala, Sweden
- Department of Surgical Sciences, Anaesthesiology and Intensive Care Medicine, Uppsala University, Uppsala, Sweden
| | - C. Thålin
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - A. Rosell
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - V. Hjalmar
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Diagnostic Centre, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| | - H. Wallén
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - H. Lindman
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, Uppsala, Sweden
| | - G. Pejler
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| | - E. Hagström
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - M. Hultström
- Department of Medical Cell Biology, Integrative Physiology, Uppsala University, Uppsala, Sweden
- Department of Surgical Sciences, Anaesthesiology and Intensive Care Medicine, Uppsala University, Uppsala, Sweden
| | - A. Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - AK. Olsson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Uppsala, Sweden
| |
Collapse
|
159
|
Patil S, Darcourt J, Messina P, Bozsak F, Cognard C, Doyle K. Characterising acute ischaemic stroke thrombi: insights from histology, imaging and emerging impedance-based technologies. Stroke Vasc Neurol 2022; 7:353-363. [PMID: 35241632 PMCID: PMC9453827 DOI: 10.1136/svn-2021-001038] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 02/02/2022] [Indexed: 12/13/2022] Open
Abstract
Treatment of acute ischaemic stroke (AIS) focuses on rapid recanalisation of the occluded artery. In recent years, advent of mechanical thrombectomy devices and new procedures have accelerated the analysis of thrombi retrieved during the endovascular thrombectomy procedure. Despite ongoing developments and progress in AIS imaging techniques, it is not yet possible to conclude definitively regarding thrombus characteristics that could advise on the probable efficacy of thrombolysis or thrombectomy in advance of treatment. Intraprocedural devices with dignostic capabilities or new clinical imaging approaches are needed for better treatment of AIS patients. In this review, what is known about the composition of the thrombi that cause strokes and the evidence that thrombus composition has an impact on success of acute stroke treatment has been examined. This review also discusses the evidence that AIS thrombus composition varies with aetiology, questioning if suspected aetiology could be a useful indicator to stroke physicians to help decide the best acute course of treatment. Furthermore, this review discusses the evidence that current widely used radiological imaging tools can predict thrombus composition. Further use of new emerging technologies based on bioimpedance, as imaging modalities for diagnosing AIS and new medical device tools for detecting thrombus composition in situ has been introduced. Whether bioimpedance would be beneficial for gaining new insights into in situ thrombus composition that could guide choice of optimum treatment approach is also reviewed.
Collapse
Affiliation(s)
- Smita Patil
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland
| | | | | | | | | | - Karen Doyle
- CÚRAM, SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway, Ireland .,Physiology, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
160
|
Aukrust SG, Holte KB, Opstad TB, Seljeflot I, Berg TJ, Helseth R. NETosis in Long-Term Type 1 Diabetes Mellitus and Its Link to Coronary Artery Disease. Front Immunol 2022; 12:799539. [PMID: 35069582 PMCID: PMC8767558 DOI: 10.3389/fimmu.2021.799539] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/15/2021] [Indexed: 12/30/2022] Open
Abstract
Background Neutrophil extracellular traps NETs have been linked to glucose and the pathogenesis of type 1 diabetes mellitus (T1DM). NETs also play a role in vascular inflammation and the development of coronary artery disease (CAD). The role of NETs in CAD progression in patients with long-term T1DM is unclear. We aimed to 1) investigate whether levels of circulating NETs markers were elevated in long-term T1DM subjects compared to controls, and 2) explore whether levels of NETs were related to the presence of CAD. Material and Methods 102 patients with > 45 years of T1DM and 75 age-matched controls were enrolled in a cross-sectional study. Median age was 62 years. Computed tomography coronary angiography (CTCA) was performed in 148 subjects without established coronary heart disease. For the current study, CAD was defined as a coronary artery stenosis >50%. Double-stranded deoxyribonucleic acid (dsDNA) was measured by a nucleic acid stain, myeloperoxidase-DNA (MPO-DNA), citrullinated histone 3 (H3Cit) and peptidylarginine deiminase 4 (PAD4) by ELISAs, while gene expression of PAD4 was measured in leukocytes from PAXgene tubes. Results Circulating MPO-DNA levels were significantly lower in patients with T1DM than in controls (0.17 vs 0.29 OD, p<0.001), while dsDNA, H3Cit, PAD4 and gene expression of PAD4 did not differ with respect to the presence of T1DM. There were no significant associations between NETs markers and HbA1c in the T1DM group. None of the NETs markers differed according to the presence of CAD in patients with T1DM. While all circulating NETs markers correlated significantly with circulating neutrophils in the control group (r=0.292-393, p<0.014), only H3Cit and PAD4 correlated with neutrophils in the T1DM group (r= 0.330-0.449, p ≤ 0.001). Conclusions In this cross-sectional study of patients with long-term T1DM and age-matched controls, circulating NETs levels were not consistently associated with the presence of T1DM or glycemic status, and did not differ according to the presence of CAD in patients with T1DM. Our results entail the possibility of altered neutrophil function and reduced NETosis in T1DM. This warrants further investigation.
Collapse
Affiliation(s)
- Sverre Grøver Aukrust
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | | | - Trine B Opstad
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ingebjørg Seljeflot
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Tore Julsrud Berg
- Department of Endocrinology, Oslo University Hospital Aker, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ragnhild Helseth
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| |
Collapse
|
161
|
Kashir J, Ambia AR, Shafqat A, Sajid MR, AlKattan K, Yaqinuddin A. Scientific premise for the involvement of neutrophil extracellular traps (NETs) in vaccine-induced thrombotic thrombocytopenia (VITT). J Leukoc Biol 2022; 111:725-734. [PMID: 34467562 PMCID: PMC8667645 DOI: 10.1002/jlb.5covr0621-320rr] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/17/2022] Open
Abstract
Following on from the devastating spread of COVID-19, a major global priority has been the production, procurement, and distribution of effective vaccines to ensure that the global pandemic reaches an end. However, concerns were raised about worrying side effects, particularly the occurrence of thrombosis and thrombocytopenia after administration of the Oxford/AstraZeneca and Johnson & Johnson's Janssen COVID-19 vaccine, in a phenomenon being termed vaccine-induced thrombotic thrombocytopenia (VITT). Similar to heparin-induced thrombocytopenia (HIT), this condition has been associated with the development of anti-platelet factor 4 antibodies, purportedly leading to neutrophil-platelet aggregate formation. Although thrombosis has also been a common association with COVID-19, the precise molecular mechanisms governing its occurrence are yet to be established. Recently, increasing evidence highlights the NLRP3 (NOD-like, leucine-rich repeat domains, and pyrin domain-containing protein) inflammasome complex along with IL-1β and effete neutrophils producing neutrophil extracellular traps (NETs) through NETosis. Herein, we propose and discuss that perhaps the incidence of VITT may be due to inflammatory reactions mediated via IL-1β/NLRP3 inflammasome activation and consequent overproduction of NETs, where similar autoimmune mechanisms are observed in HIT. We also discuss avenues by which such modalities could be treated to prevent the occurrence of adverse events and ensure vaccine rollouts remain safe and on target to end the current pandemic.
Collapse
Affiliation(s)
- Junaid Kashir
- Alfaisal UniversityRiyadhKingdom of Saudi Arabia
- Department of Comparative MedicineKing Faisal Specialist Hospital and Research CenterRiyadhKingdom of Saudi Arabia
| | | | | | | | | | | |
Collapse
|
162
|
von Meijenfeldt FA, Stravitz RT, Zhang J, Adelmeijer J, Zen Y, Durkalski V, Lee W, Lisman T. Generation of neutrophil extracellular traps in patients with acute liver failure is associated with poor outcome. Hepatology 2022; 75:623-633. [PMID: 34562318 PMCID: PMC9299791 DOI: 10.1002/hep.32174] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 08/11/2021] [Accepted: 09/09/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS Acute liver failure (ALF) is characterized by significant changes in the hemostatic system and by systemic inflammation. The formation of neutrophil extracellular traps (NETs), in which an activated neutrophil expels its DNA, histones, and granular enzymes, such as myeloperoxidase (MPO), has been associated with immune-mediated and thrombotic diseases. We hypothesized that formation of NETs in patients with ALF contributes to progression of disease. APPROACH AND RESULTS A total of 676 patients with ALF (international normalized ratio [INR], ≥1.5) or severe acute liver injury (ALI; INR, ≥2.0) were recruited from the U.S. ALF Study Group Registry between 2011 and 2018, of whom 308 patients (45.6%) had acetaminophen-induced ALF. Up to 21 days after admission, 483 patients (71.5%) survived without liver transplantation (LT). Levels of cell-free DNA (cfDNA) and the specific NET marker MPO-DNA complexes were measured in plasma samples obtained on admission and compared to levels in healthy controls. In addition, liver tissue obtained at transplantation of 20 ALF patients was stained for NETs. Levels of cfDNA were 7.1-fold, and MPO-DNA complexes 2.5-fold, higher in patients with ALF compared to healthy controls. cfDNA levels were not associated with 21-day transplant-free survival, but were higher in those patients with more-severe disease on admission, as reflected by various laboratory and clinical parameters. MPO-DNA levels were 30% higher in patients with ALF who died or required urgent LT. Liver tissue of ALF patients stained positive for NETs in 12 of 18 evaluable patients. CONCLUSIONS Here, we provide evidence for NET formation in patients with ALF. Elevated plasma levels of MPO-DNA complexes in patients with ALF were associated with poor outcome, which suggests that NET formation contributes to disease progression.
Collapse
Affiliation(s)
- Fien A. von Meijenfeldt
- Surgical Research LaboratoryDepartment of SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - R. Todd Stravitz
- Hume‐Lee Transplant Center of Virginia Commonwealth UniversityRichmondVirginiaUSA
| | - Jingwen Zhang
- Department of Public Health SciencesMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Jelle Adelmeijer
- Surgical Research LaboratoryDepartment of SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Yoh Zen
- Department of PathologyInstitute of Liver StudiesKing’s College HospitalLondonUK
| | - Valerie Durkalski
- Department of Public Health SciencesMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - William M. Lee
- University of Texas‐Southwestern Medical CenterDallasTexasUSA
| | - Ton Lisman
- Surgical Research LaboratoryDepartment of SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
163
|
Zheng J, Qi R, Dai C, Li G, Sang M. Enzyme Catalysis Biomotor Engineering of Neutrophils for Nanodrug Delivery and Cell-Based Thrombolytic Therapy. ACS NANO 2022; 16:2330-2344. [PMID: 35138084 DOI: 10.1021/acsnano.1c08538] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Utilizing neutrophils (NEs) to target and deliver nanodrugs to inflammation sites has received considerable attention. NEs are involved in the formation and development of thrombosis by transforming into neutrophil extracellular traps (NETs); this indicates that NEs may be a natural thrombolytic drug delivery carrier. However, NEs lack an effective power system to overcome blood flow resistance and enhance targeting efficiency. Herein, we report the application of a urease catalysis micromotor powered NEs nanodrug delivery system to promote thrombolysis and suppress rethrombosis. The urease micromotor powered Janus NEs (UM-NEs) were prepared by immobilizing the enzyme asymmetrically onto the surface of natural NEs and then loading urokinase (UK) coupled silver (Ag) nanoparticles (Ag-UK) to obtain the UM-NEs (Ag-UK) system. Urease catalytic endogenous urea is used to generate thrust by producing ammonia and carbon dioxide, which propels NEs actively targeting the thrombus. The UM-NEs (Ag-UK) can be activated by enriched inflammatory cytokines to release NETs at the thrombosis site, resulting in a concomitant release of Ag-UK. Ag-UK induces thrombolysis to restore vascular recanalization. This urease micromotor-driven NEs drug delivery system can significantly reduce the hemorrhagic side effects, promote thrombolysis, and inhibit rethrombosis with high bioavailability and biosafety, which can be used for the treatment of thrombotic diseases.
Collapse
Affiliation(s)
- Jinrong Zheng
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Huli District, Xiamen, China 361006
| | - Ruiqiang Qi
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Huli District, Xiamen, China 361006
| | - Cuilian Dai
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Huli District, Xiamen, China 361006
| | - Gang Li
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Huli District, Xiamen, China 361006
| | - Mangmang Sang
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, 2999 Jinshan Road, Huli District, Xiamen, China 361006
| |
Collapse
|
164
|
Poh XY, Loh FK, Friedland JS, Ong CWM. Neutrophil-Mediated Immunopathology and Matrix Metalloproteinases in Central Nervous System - Tuberculosis. Front Immunol 2022; 12:788976. [PMID: 35095865 PMCID: PMC8789671 DOI: 10.3389/fimmu.2021.788976] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/20/2021] [Indexed: 12/19/2022] Open
Abstract
Tuberculosis (TB) remains one of the leading infectious killers in the world, infecting approximately a quarter of the world’s population with the causative organism Mycobacterium tuberculosis (M. tb). Central nervous system tuberculosis (CNS-TB) is the most severe form of TB, with high mortality and residual neurological sequelae even with effective TB treatment. In CNS-TB, recruited neutrophils infiltrate into the brain to carry out its antimicrobial functions of degranulation, phagocytosis and NETosis. However, neutrophils also mediate inflammation, tissue destruction and immunopathology in the CNS. Neutrophils release key mediators including matrix metalloproteinase (MMPs) which degrade brain extracellular matrix (ECM), tumor necrosis factor (TNF)-α which may drive inflammation, reactive oxygen species (ROS) that drive cellular necrosis and neutrophil extracellular traps (NETs), interacting with platelets to form thrombi that may lead to ischemic stroke. Host-directed therapies (HDTs) targeting these key mediators are potentially exciting, but currently remain of unproven effectiveness. This article reviews the key role of neutrophils and neutrophil-derived mediators in driving CNS-TB immunopathology.
Collapse
Affiliation(s)
- Xuan Ying Poh
- Infectious Diseases Translational Research Programme, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Fei Kean Loh
- Infectious Diseases Translational Research Programme, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jon S Friedland
- Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| | - Catherine W M Ong
- Infectious Diseases Translational Research Programme, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Division of Infectious Diseases, Department of Medicine, National University Hospital, Singapore, Singapore.,Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, Singapore
| |
Collapse
|
165
|
Abstract
Cancer-associated thrombosis (including venous thromboembolism (VTE) and arterial events) is highly consequential for patients with cancer and is associated with worsened survival. Despite substantial improvements in cancer treatment, the risk of VTE has increased in recent years; VTE rates additionally depend on the type of cancer (with pancreas, stomach and primary brain tumours having the highest risk) as well as on individual patient's and cancer treatment factors. Multiple cancer-specific mechanisms of VTE have been identified and can be classified as mechanisms in which the tumour expresses proteins that alter host systems, such as levels of platelets and leukocytes, and in which the tumour expresses procoagulant proteins released into the circulation that directly activate the coagulation cascade or platelets, such as tissue factor and podoplanin, respectively. As signs and symptoms of VTE may be non-specific, diagnosis requires clinical assessment, evaluation of pre-test probability, and objective diagnostic testing with ultrasonography or CT. Risk assessment tools have been validated to identify patients at risk of VTE. Primary prevention of VTE (thromboprophylaxis) has long been recommended in the inpatient and post-surgical settings, and is now an option in the outpatient setting for individuals with high-risk cancer. Anticoagulant therapy is the cornerstone of therapy, with low molecular weight heparin or newer options such as direct oral anticoagulants. Personalized treatment incorporating risk of bleeding and patient preferences is essential, especially as a diagnosis of VTE is often considered by patients even more distressing than their cancer diagnosis, and can severely affect the quality of life. Future research should focus on current knowledge gaps including optimizing risk assessment tools, biomarker discovery, next-generation anticoagulant development and implementation science.
Collapse
|
166
|
Langiu M, Palacios-Acedo AL, Crescence L, Mege D, Dubois C, Panicot-Dubois L. Neutrophils, Cancer and Thrombosis: The New Bermuda Triangle in Cancer Research. Int J Mol Sci 2022; 23:ijms23031257. [PMID: 35163180 PMCID: PMC8836160 DOI: 10.3390/ijms23031257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 12/17/2022] Open
Abstract
Spontaneous venous thrombosis is often the first clinical sign of cancer, and it is linked to a worsened survival rate. Traditionally, tumor-cell induced platelet activation has been the main actor studied in cancer-associated-thrombosis. However, platelet involvement alone does not seem to be sufficient to explain this heightened pro-thrombotic state. Neutrophils are emerging as key players in both thrombus generation and cancer progression. Neutrophils can impact thrombosis through the release of pro-inflammatory cytokines and expression of molecules like P-selectin and Tissue Factor (TF) on their membrane and on neutrophil-derived microvesicles. Their role in cancer progression is evidenced by the fact that patients with high blood-neutrophil counts have a worsened prognosis. Tumors can attract neutrophils to the cancer site via pro-inflammatory cytokine secretions and induce a switch to pro-tumoral (or N2) neutrophils, which support metastatic spread and have an immunosuppressive role. They can also expel their nuclear contents to entrap pathogens forming Neutrophil Extracellular Traps (NETs) and can also capture coagulation factors, enhancing the thrombus formation. These NETs are also known to have pro-tumoral effects by supporting the metastatic process. Here, we strived to do a comprehensive literature review of the role of neutrophils as drivers of both cancer-associated thrombosis (CAT) and cancer progression.
Collapse
Affiliation(s)
- Mélanie Langiu
- Aix Marseille Univ INSERM, INRAE, C2VN, 13005 Marseille, France; (M.L.); (A.-L.P.-A.); (L.C.); (D.M.); (L.P.-D.)
| | - Ana-Luisa Palacios-Acedo
- Aix Marseille Univ INSERM, INRAE, C2VN, 13005 Marseille, France; (M.L.); (A.-L.P.-A.); (L.C.); (D.M.); (L.P.-D.)
| | - Lydie Crescence
- Aix Marseille Univ INSERM, INRAE, C2VN, 13005 Marseille, France; (M.L.); (A.-L.P.-A.); (L.C.); (D.M.); (L.P.-D.)
| | - Diane Mege
- Aix Marseille Univ INSERM, INRAE, C2VN, 13005 Marseille, France; (M.L.); (A.-L.P.-A.); (L.C.); (D.M.); (L.P.-D.)
- Department of Digestive Surgery, La Timone University Hospital, 13005 Marseille, France
| | - Christophe Dubois
- Aix Marseille Univ INSERM, INRAE, C2VN, 13005 Marseille, France; (M.L.); (A.-L.P.-A.); (L.C.); (D.M.); (L.P.-D.)
- Correspondence:
| | - Laurence Panicot-Dubois
- Aix Marseille Univ INSERM, INRAE, C2VN, 13005 Marseille, France; (M.L.); (A.-L.P.-A.); (L.C.); (D.M.); (L.P.-D.)
| |
Collapse
|
167
|
Zhu X, Peng L, Song E, Song Y. Polystyrene Nanoplastics Induce Neutrophil Extracellular Traps in Mice Neutrophils. Chem Res Toxicol 2022; 35:378-382. [DOI: 10.1021/acs.chemrestox.1c00374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xiangyu Zhu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Road, Beibei District, Chongqing 400715, China
| | - Lu Peng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Road, Beibei District, Chongqing 400715, China
| | - Erqun Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Road, Beibei District, Chongqing 400715, China
| | - Yang Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China
| |
Collapse
|
168
|
Alkattan W, Yaqinuddin A, Shafqat A, Kashir J. NET-Mediated Pathogenesis of COVID-19: The Role of NETs in Hepatic Manifestations. JOURNAL OF HEALTH AND ALLIED SCIENCES NU 2022. [DOI: 10.1055/s-0041-1741418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AbstractSome coronavirus disease-2019 (COVID-19) patients exhibit multi-organ failure, which often includes the liver. Indeed, liver disease appears to be an emerging feature of COVID-19 infections. However, the exact mechanism behind this remains unknown. Neutrophil extracellular traps (NETs) have increasingly been attributed as major contributors to various liver pathologies, including sepsis, ischemic-reperfusion (I/R) injury, and portal hypertension in the setting of chronic liver disease. Although vital in normal immunity, excessive NET formation can drive inflammation, particularly of the endothelium. Collectively, we propose that NETs observed to be elevated in severe COVID-19 infection play principal roles in liver injury in addition to acute lung injury. Herein, we discuss the potential mechanisms underlying COVID-induced liver injury including cytopathic effects from direct liver infection, systemic inflammatory response syndrome, and hypoxic injury, encompassing I/R injury and coagulopathy. Further research is required to further elucidate the role of NETs in COVID. This holds potential therapeutic significance, as inhibition of NETosis could alleviate the symptoms of acute respiratory distress syndrome and liver injury, as well as other organs.
Collapse
Affiliation(s)
- Wael Alkattan
- College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| | - Junaid Kashir
- College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
169
|
Afosah DK, Ofori E, Mottamal M, Al-Horani RA. Factor IX(a) inhibitors: an updated patent review (2003-present). Expert Opin Ther Pat 2022; 32:381-400. [PMID: 34991418 DOI: 10.1080/13543776.2022.2026926] [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] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Anticoagulation with no bleeding complications is the current objective of drug discovery programs in the area of treating and/or preventing thromboembolism. Despite the promises of therapeutics targeting factors XI(a) and XII(a), none has been approved thus far. Clinically used thrombin- and/or factor Xa-based anticoagulants continue to be associated with a significant bleeding risk which limits their safe use in a broad range of thrombotic patients. Research findings in animals and humans indicate that it is possible to target factor IX(a) (FIX(a)) to achieve anticoagulation with a limited risk of bleeding. AREAS COVERED A review of patents literature has retrieved >35 patents on the development of molecules targeting FIX(a) since 2003. Small molecules, antibodies, and aptamers have been developed to target FIX(a) to potentially promote effective and safer anticoagulation. Most of these agents are in the pre-clinical development phase and few have been tested in clinical trials. EXPERT OPINION FIX(a) system is being considered to develop new anticoagulants with fewer bleeding complications. Our survey indicates that the number of FIX(a)-targeting agents is mediocre. The agents under development are diverse. Although additional development is essential, moving one or more of these agents to the clinic will facilitate achieving better clinical outcomes.
Collapse
Affiliation(s)
- Daniel K Afosah
- Department of Chemistry and Biochemistry, Washington and Lee University, Lexington, VA, USA
| | - Edward Ofori
- Department of Pharmaceutical Sciences, College of Pharmacy, Chicago State University, Chicago, IL, USA
| | - Madhusoodanan Mottamal
- Department of Chemistry, College of Arts and Sciences, Xavier University of Louisiana, New Orleans, LA, USA
| | - Rami A Al-Horani
- Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, Louisiana, USA
| |
Collapse
|
170
|
Gorog DA, Massberg S. NETs in the infarct-related coronary artery - a marker or mediator of adverse outcome? Thromb Haemost 2022; 122:1251-1254. [PMID: 35008117 DOI: 10.1055/a-1733-9217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
No Abstract.
Collapse
Affiliation(s)
- Diana Adrienne Gorog
- Cardiology, Imperial College, London, United Kingdom of Great Britain and Northern Ireland.,Cardiology, E&N Hertfordshire NHS Trust, United Kingdom of Great Britain and Northern Ireland
| | - Steffen Massberg
- Klinik für Herz- und Kreislauferkrankungen, Deutsches Herzzentrum München, München, Germany
| |
Collapse
|
171
|
Coagulome and the tumor microenvironment: an actionable interplay. Trends Cancer 2022; 8:369-383. [PMID: 35027336 DOI: 10.1016/j.trecan.2021.12.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/19/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022]
Abstract
Human tumors often trigger a hypercoagulable state that promotes hemostatic complications, including venous thromboembolism. The recent application of systems biology to the study of the coagulome highlighted its link to shaping the tumor microenvironment (TME), both within and outside of the vascular space. Addressing this link provides the opportunity to revisit the significance of biomarkers of hemostasis and assess the communication between vasculature and tumor parenchyma, an important topic considering the advent of immune checkpoint inhibitors and vascular normalization strategies. Understanding how the coagulome and TME influence each other offers exciting new prospects for predicting hemostatic complications and boosting the effectiveness of cancer treatment.
Collapse
|
172
|
Louka AM, Sagris D, Ntaios G. Ιmmunity, Vascular Aging, and Stroke. Curr Med Chem 2022; 29:5510-5521. [PMID: 34979888 DOI: 10.2174/0929867329666220103101700] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/04/2021] [Accepted: 11/08/2021] [Indexed: 11/22/2022]
Abstract
Stroke is one of the most devastating manifestations of cardiovascular disease. Growing age, arterial hypertension, and atherosclerosis are identified as independent risk factors for stroke, primarily due to structural and functional alterations in the cerebrovascular tree. Recent data from in vitro and clinical studies have suggested that the immune system influences atherosclerosis, promoting vascular stiffness and vascular aging and contributing to ischemic stroke, intracranial haemorrhage and microbleeds, white matter disease, and cognitive decline. Furthermore, aging is related to a chronic low-grade inflammatory state, in which macrophage, neutrophils, natural killer (NK cells), and B and T lymphocytes act as major effectors of the immune-mediated cell responses. Moreover, oxidative stress and vascular inflammation are correlated with endothelial dysfunction, vascular aging, blood-brain barrier disruption, lacunar lesions, and neurodegenerative disorders. This review discusses the pathophysiological roles of fundamental cellular and molecular mechanisms of aging, including the complex interplay between them and innate immunity, as well as vascular dysfunction, arterial stiffness, atherosclerosis, atherothrombosis, systemic inflammation, and blood-brain barrier dysfunction.
Collapse
Affiliation(s)
- Anna-Maria Louka
- Department of Internal Medicine, School of Health Sciences, Faculty of Medicine, University of Thessaly, Larissa Greece
| | - Dimitrios Sagris
- Department of Internal Medicine, School of Health Sciences, Faculty of Medicine, University of Thessaly, Larissa Greece
| | - George Ntaios
- Department of Internal Medicine, School of Health Sciences, Faculty of Medicine, University of Thessaly, Larissa Greece
| |
Collapse
|
173
|
Bauer AT, Gorzelanny C, Gebhardt C, Pantel K, Schneider SW. Interplay between coagulation and inflammation in cancer: Limitations and therapeutic opportunities. Cancer Treat Rev 2022; 102:102322. [DOI: 10.1016/j.ctrv.2021.102322] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022]
|
174
|
Wolach O, Martinod K. Casting a NET on cancer: the multiple roles for neutrophil extracellular traps in cancer. Curr Opin Hematol 2022; 29:53-62. [PMID: 34854835 DOI: 10.1097/moh.0000000000000690] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW The role of the innate immune system has become widely appreciated in cancer and cancer-associated disorders. Neutrophils, the most abundant circulating leukocytes, have prognostic value in determining cancer progression and survival. One of the ways by which neutrophils negatively impact outcome is by formation of neutrophil extracellular traps (NETs) which result in release of nuclear chromatin and bioactive proteins into the extracellular space. Here, we review the evidence for NETs contributions to cancer progression, metastasis, and cancer-associated thrombosis (CAT). RECENT FINDINGS NETs are increased across several cancer types and predict progression and adverse outcome. Several preclinical and clinical observations implicate NETs in promoting tumor growth, angiogenesis and metastasis via distinct pathways. Furthermore, NETs are shown to contribute to resistance to immunotherapy. NETs also emerge as key players in the prothrombotic phenotype associated with cancer that can result in potentially life-threatening arterial and venous thrombosis. Recent mechanistic insights expose several potential targets to inhibit NET formation and disrupt the interaction between NETs and tumor cells. SUMMARY Clinical and translational insights highlight the central role of NETs in cancer progression and metastasis, disease resistance and CAT. Targeting NETs and NET-associated pathways may represent a novel approach to treat cancer.
Collapse
Affiliation(s)
- Ofir Wolach
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Kimberly Martinod
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
175
|
Caillon A, Trimaille A, Favre J, Jesel L, Morel O, Kauffenstein G. Role of neutrophils, platelets, and extracellular vesicles and their interactions in COVID-19-associated thrombopathy. J Thromb Haemost 2022; 20:17-31. [PMID: 34672094 PMCID: PMC8646423 DOI: 10.1111/jth.15566] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/28/2021] [Accepted: 10/19/2021] [Indexed: 12/14/2022]
Abstract
The COVID-19 pandemic extended all around the world causing millions of deaths. In addition to acute respiratory distress syndrome, many patients with severe COVID-19 develop thromboembolic complications associated to multiorgan failure and death. Here, we review evidence for the contribution of neutrophils, platelets, and extracellular vesicles (EVs) to the thromboinflammatory process in COVID-19. We discuss how the immune system, influenced by pro-inflammatory molecules, EVs, and neutrophil extracellular traps (NETs), can be caught out in patients with severe outcomes. We highlight how the deficient regulation of the innate immune system favors platelet activation and induces a vicious cycle amplifying an immunothrombogenic environment associated with platelet/NET interactions. In light of these considerations, we discuss potential therapeutic strategies underlining the modulation of purinergic signaling as an interesting target.
Collapse
Affiliation(s)
- Antoine Caillon
- Lady Davis Institute for Medical Research, McGill University, Montréal, Quebec, Canada
| | - Antonin Trimaille
- UMR INSERM 1260, CRBS, Strasbourg University, Strasbourg, France
- Division of Cardiovascular Medicine, Nouvel Hôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | - Julie Favre
- INSERM, UMR S 1121, Biomaterials and Bioengineering, CRBS, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
| | - Laurence Jesel
- UMR INSERM 1260, CRBS, Strasbourg University, Strasbourg, France
- Division of Cardiovascular Medicine, Nouvel Hôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | - Olivier Morel
- UMR INSERM 1260, CRBS, Strasbourg University, Strasbourg, France
- Division of Cardiovascular Medicine, Nouvel Hôpital Civil, Strasbourg University Hospital, Strasbourg, France
| | | |
Collapse
|
176
|
Blanch-Ruiz MA, Ortega-Luna R, Gómez-García G, Martínez-Cuesta MÁ, Álvarez Á. Role of Neutrophil Extracellular Traps in COVID-19 Progression: An Insight for Effective Treatment. Biomedicines 2021; 10:31. [PMID: 35052711 PMCID: PMC8772933 DOI: 10.3390/biomedicines10010031] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 12/14/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2, has resulted in a pandemic with over 270 million confirmed cases and 5.3 million deaths worldwide. In some cases, the infection leads to acute respiratory distress syndrome (ARDS), which is triggered by a cytokine storm and multiple organ failure. Clinical hematological, biochemical, coagulation, and inflammatory markers, such as interleukins, are associated with COVID-19 disease progression. In this regard, neutrophilia, neutrophil-to-lymphocyte ratio (NLR), and neutrophil-to-albumin ratio (NAR), have emerged as promising biomarkers of disease severity and progression. In the pathophysiology of ARDS, the inflammatory environment induces neutrophil influx and activation in the lungs, promoting the release of cytokines, proteases, reactive oxygen species (ROS), and, eventually, neutrophil extracellular traps (NETs). NETs components, such as DNA, histones, myeloperoxidase, and elastase, may exert cytotoxic activity and alveolar damage. Thus, NETs have also been described as potential biomarkers of COVID-19 prognosis. Several studies have demonstrated that NETs are induced in COVID-19 patients, and that the highest levels of NETs are found in critical ones, therefore highlighting a correlation between NETs and severity of the disease. Knowledge of NETs signaling pathways, and the targeting of points of NETs release, could help to develop an effective treatment for COVID-19, and specifically for severe cases, which would help to manage the pandemic.
Collapse
Affiliation(s)
- María Amparo Blanch-Ruiz
- Departamento de Farmacología, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.); (G.G.-G.)
| | - Raquel Ortega-Luna
- Departamento de Farmacología, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.); (G.G.-G.)
| | - Guillermo Gómez-García
- Departamento de Farmacología, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.); (G.G.-G.)
| | - Maria Ángeles Martínez-Cuesta
- Departamento de Farmacología, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.); (G.G.-G.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), 46010 Valencia, Spain
| | - Ángeles Álvarez
- Departamento de Farmacología, Facultad de Medicina y Odontología, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.); (G.G.-G.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), 46010 Valencia, Spain
| |
Collapse
|
177
|
Zhou Y, Tao W, Shen F, Du W, Xu Z, Liu Z. The Emerging Role of Neutrophil Extracellular Traps in Arterial, Venous and Cancer-Associated Thrombosis. Front Cardiovasc Med 2021; 8:786387. [PMID: 34926629 PMCID: PMC8674622 DOI: 10.3389/fcvm.2021.786387] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/29/2021] [Indexed: 12/16/2022] Open
Abstract
Neutrophils play a vital role in the formation of arterial, venous and cancer-related thrombosis. Recent studies have shown that in a process known as NETosis, neutrophils release proteins and enzymes complexed to DNA fibers, collectively called neutrophil extracellular traps (NETs). Although NETs were originally described as a way for the host to capture and kill bacteria, current knowledge indicates that NETs also play an important role in thrombosis. According to recent studies, the destruction of vascular microenvironmental homeostasis and excessive NET formation lead to pathological thrombosis. In vitro experiments have found that NETs provide skeletal support for platelets, red blood cells and procoagulant molecules to promote thrombosis. The protein components contained in NETs activate the endogenous coagulation pathway to promote thrombosis. Therefore, NETs play an important role in the formation of arterial thrombosis, venous thrombosis and cancer-related thrombosis. This review will systematically summarize and explain the study of NETs in thrombosis in animal models and in vivo experiments to provide new targets for thrombosis prevention and treatment.
Collapse
Affiliation(s)
- Yilu Zhou
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weimin Tao
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fuyi Shen
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weijia Du
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhendong Xu
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiqiang Liu
- Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
178
|
Mao L, Mostafa R, Ibili E, Fert-Bober J. Role of protein deimination in cardiovascular diseases: potential new avenues for diagnostic and prognostic biomarkers. Expert Rev Proteomics 2021; 18:1059-1071. [PMID: 34929115 DOI: 10.1080/14789450.2021.2018303] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Arginine deimination (citrullination) is a post-translational modification catalyzed by a family of peptidyl arginine deiminase (PAD) enzymes. Cell-based functional studies and animal models have manifested the key role of PADs in various cardiovascular diseases (CVDs). AREA COVERED This review summarizes the latest developments in the role of PADs in CVD pathogenesis. It focuses on the PAD functions and diverse citrullinated proteins in cardiovascular conditions like deep vein thrombosis, ischemia/reperfusion, and atherosclerosis. Identification of PAD isoforms and citrullinated targets are essential for directing diagnosis and clinical intervention. Finally, anti-citrullinated protein antibodies (ACPAs) are addressed as an independent risk factor for cardiovascular events. A search of PubMed biomedical literature from the past ten years was performed with a combination of the following keywords: PAD/PADI, deimination/citrullination, autoimmune, fibrosis, NET, neutrophil, macrophage, inflammation, inflammasome, cardiovascular, heart disease, myocardial infarction, ischemia, atherosclerosis, thrombosis, and aging. Additional papers from retrieved articles were also considered. EXPERT OPINION PADs are unique family of enzymes that converts peptidyl-arginine to -citrulline in protein permanently. Overexpression or increased activity of PAD has been observed in various CVDs with acute and chronic inflammation as the background. Importantly, far beyond being simply involved in forming neutrophil extracellular traps (NETs), accumulating evidence indicated PAD activation as a trigger for numerous processes, such as transcriptional regulation, endothelial dysfunction, and thrombus formation. In summary, the findings so far have testified the important role of deimination in cardiovascular biology, while more basic and translational studies are essential to further exploration.
Collapse
Affiliation(s)
- Liqun Mao
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Advanced Clinical Biosystems Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Rowann Mostafa
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Advanced Clinical Biosystems Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Esra Ibili
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Advanced Clinical Biosystems Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Justyna Fert-Bober
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Advanced Clinical Biosystems Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
179
|
Neutrophil-Derived Extracellular Vesicles Activate Platelets after Pneumolysin Exposure. Cells 2021; 10:cells10123581. [PMID: 34944089 PMCID: PMC8700313 DOI: 10.3390/cells10123581] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Pneumolysin (PLY) is a pore-forming toxin of Streptococcus pneumoniae that contributes substantially to the inflammatory processes underlying pneumococcal pneumonia and lung injury. Host responses against S. pneumoniae are regulated in part by neutrophils and platelets, both individually and in cooperative interaction. Previous studies have shown that PLY can target both neutrophils and platelets, however, the mechanisms by which PLY directly affects these cells and alters their interactions are not completely understood. In this study, we characterize the effects of PLY on neutrophils and platelets and explore the mechanisms by which PLY may induce neutrophil–platelet interactions. In vitro studies demonstrated that PLY causes the formation of neutrophil extracellular traps (NETs) and the release of extracellular vesicles (EVs) from both human and murine neutrophils. In vivo, neutrophil EV (nEV) levels were increased in mice infected with S. pneumoniae. In platelets, treatment with PLY induced the cell surface expression of P-selectin (CD62P) and binding to annexin V and caused a significant release of platelet EVs (pl-EVs). Moreover, PLY-induced nEVs but not NETs promoted platelet activation. The pretreatment of nEVs with proteinase K inhibited platelet activation, indicating that the surface proteins of nEVs play a role in this process. Our findings demonstrate that PLY activates neutrophils and platelets to release EVs and support an important role for neutrophil EVs in modulating platelet functions in pneumococcal infections.
Collapse
|
180
|
Knight JS, Caricchio R, Casanova JL, Combes AJ, Diamond B, Fox SE, Hanauer DA, James JA, Kanthi Y, Ladd V, Mehta P, Ring AM, Sanz I, Selmi C, Tracy RP, Utz PJ, Wagner CA, Wang JY, McCune WJ. The intersection of COVID-19 and autoimmunity. J Clin Invest 2021; 131:e154886. [PMID: 34710063 PMCID: PMC8670833 DOI: 10.1172/jci154886] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Acute COVID-19, caused by SARS-CoV-2, is characterized by diverse clinical presentations, ranging from asymptomatic infection to fatal respiratory failure, and often associated with varied longer-term sequelae. Over the past 18 months, it has become apparent that inappropriate immune responses contribute to the pathogenesis of severe COVID-19. Researchers working at the intersection of COVID-19 and autoimmunity recently gathered at an American Autoimmune Related Diseases Association Noel R. Rose Colloquium to address the current state of knowledge regarding two important questions: Does established autoimmunity predispose to severe COVID-19? And, at the same time, can SARS-CoV-2 infection trigger de novo autoimmunity? Indeed, work to date has demonstrated that 10% to 15% of patients with critical COVID-19 pneumonia exhibit autoantibodies against type I interferons, suggesting that preexisting autoimmunity underlies severe disease in some patients. Other studies have identified functional autoantibodies following infection with SARS-CoV-2, such as those that promote thrombosis or antagonize cytokine signaling. These autoantibodies may arise from a predominantly extrafollicular B cell response that is more prone to generating autoantibody-secreting B cells. This Review highlights the current understanding, evolving concepts, and unanswered questions provided by this unique opportunity to determine mechanisms by which a viral infection can be exacerbated by, and even trigger, autoimmunity. The potential role of autoimmunity in post-acute sequelae of COVID-19 is also discussed.
Collapse
Affiliation(s)
- Jason S. Knight
- Division of Rheumatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Roberto Caricchio
- Section of Rheumatology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, The Rockefeller University, New York, New York, USA
- Howard Hughes Medical Institute, New York, New York, USA
- Laboratory of Human Genetics of Infectious Diseases, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | - Alexis J. Combes
- Department of Pathology, ImmunoX Initiative, UCSF Immunoprofiler Initiative, UCSF CoLabs, UCSF, San Francisco, California, USA
| | - Betty Diamond
- Center for Autoimmune and Musculoskeletal Diseases, Northwell Health’s Feinstein Institute for Medical Research, New York, New York, USA
| | - Sharon E. Fox
- Pathology and Laboratory Medicine Service, Southeast Louisiana Veterans Healthcare System, New Orleans, Louisiana, USA
- Department of Pathology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - David A. Hanauer
- Department of Pediatrics and School of Information, University of Michigan, Ann Arbor, Michigan, USA
| | - Judith A. James
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Yogendra Kanthi
- National Heart, Lung, and Blood Institute Division of Intramural Research, Bethesda, Maryland, USA
| | - Virginia Ladd
- American Autoimmune Related Diseases Association Inc., Eastpointe, Michigan, USA
| | - Puja Mehta
- Centre for Inflammation and Tissue Repair, University College London, London, United Kingdom
| | - Aaron M. Ring
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ignacio Sanz
- Division of Rheumatology, Emory University, Atlanta, Georgia, USA
| | - Carlo Selmi
- Rheumatology and Clinical Immunology, Humanitas Research Hospital–Scientific Institute for Research, Hospitalization and Healthcare, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine and Department of Biochemistry, University of Vermont Larner College of Medicine, Burlington, Vermont, USA
| | - Paul J. Utz
- Division of Immunology, Department of Medicine, Stanford University, Stanford, California, USA
| | - Catriona A. Wagner
- American Autoimmune Related Diseases Association Inc., Eastpointe, Michigan, USA
| | | | - William J. McCune
- Division of Rheumatology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
181
|
Moon MJ, McFadyen JD, Peter K. Caught at the Scene of the Crime: Platelets and Neutrophils Are Conspirators in Thrombosis. Arterioscler Thromb Vasc Biol 2021; 42:63-66. [PMID: 34852641 DOI: 10.1161/atvbaha.121.317187] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Mitchell J Moon
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.J.M., J.D.M., K.P.).,Baker Department of Cardiometabolic Health, University of Melbourne, Victoria, Australia (M.J.M., J.D.M., K.P.)
| | - James D McFadyen
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.J.M., J.D.M., K.P.).,Baker Department of Cardiometabolic Health, University of Melbourne, Victoria, Australia (M.J.M., J.D.M., K.P.).,Department of Clinical Hematology (J.D.M.), The Alfred Hospital, Melbourne, Victoria, Australia.,Departments of Medicine (J.D.M., K.P.), Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (M.J.M., J.D.M., K.P.).,Baker Department of Cardiometabolic Health, University of Melbourne, Victoria, Australia (M.J.M., J.D.M., K.P.).,Department of Cardiology (K.P.), The Alfred Hospital, Melbourne, Victoria, Australia.,Departments of Medicine (J.D.M., K.P.), Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Immunology (K.P.), Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
182
|
Kalita B, Saviola AJ, Samuel SP, Mukherjee AK. State-of-the-art review - A review on snake venom-derived antithrombotics: Potential therapeutics for COVID-19-associated thrombosis? Int J Biol Macromol 2021; 192:1040-1057. [PMID: 34656540 PMCID: PMC8514616 DOI: 10.1016/j.ijbiomac.2021.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/03/2021] [Accepted: 10/04/2021] [Indexed: 12/30/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent responsible for the Coronavirus Disease-2019 (COVID-19) pandemic, has infected over 185 million individuals across 200 countries since December 2019 resulting in 4.0 million deaths. While COVID-19 is primarily associated with respiratory illnesses, an increasing number of clinical reports indicate that severely ill patients often develop thrombotic complications that are associated with increased mortality. As a consequence, treatment strategies that target COVID-associated thrombosis are of utmost clinical importance. An array of pharmacologically active compounds from natural products exhibit effects on blood coagulation pathways, and have generated interest for their potential therapeutic applications towards thrombotic diseases. In particular, a number of snake venom compounds exhibit high specificity on different blood coagulation factors and represent excellent tools that could be utilized to treat thrombosis. The aim of this review is to provide a brief summary of the current understanding of COVID-19 associated thrombosis, and highlight several snake venom compounds that could be utilized as antithrombotic agents to target this disease.
Collapse
Affiliation(s)
- Bhargab Kalita
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; National Centre for Cell Science, Pune 411007, Maharashtra, India
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Stephen P Samuel
- Queen Elizabeth Hospital King's Lynn NHS Foundation Trust, King's Lynn, Norfolk PE30 4ET, UK
| | - Ashis K Mukherjee
- Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur 784028, Assam, India; Institute of Advanced Study in Science and Technology, Guwahati 781035, Assam, India.
| |
Collapse
|
183
|
Sugimoto T, Yamada H, Wada N, Motoyama S, Saburi M, Kubota H, Miyawaki D, Wakana N, Kami D, Ogata T, Ibi M, Matoba S. Repeated Social Defeat Exaggerates Fibrin-Rich Clot Formation by Enhancing Neutrophil Extracellular Trap Formation via Platelet-Neutrophil Interactions. Cells 2021; 10:3344. [PMID: 34943852 PMCID: PMC8699805 DOI: 10.3390/cells10123344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 12/15/2022] Open
Abstract
Depression is an independent risk factor for cardiovascular disease (CVD). We have previously shown that repeated social defeat (RSD) exaggerates atherosclerosis development by enhancing neutrophil extracellular trap (NET) formation. In this study, we investigated the impact of RSD on arterial thrombosis. Eight-week-old male wild-type mice (C57BL/6J) were exposed to RSD by housing with larger CD-1 mice in a shared home cage. They were subjected to vigorous physical contact daily for 10 consecutive days. After confirming depression-like behaviors, mice underwent FeCl3-induced carotid arterial injury and were analyzed after 3 h. Although the volume of thrombi was comparable between the two groups, fibrin(ogen)-positive areas were significantly increased in defeated mice, in which Ly-6G-positive cells were appreciably co-localized with Cit-H3-positive staining. Treatment with DNase I completely diminished exaggerated fibrin-rich clot formation in defeated mice. Flow cytometric analysis showed that neutrophil CD11b expression before FeCl3 application was significantly higher in defeated mice than in control mice. In vitro NET formation induced by activated platelets was significantly augmented in defeated mice, which was substantially inhibited by anti-CD11b antibody treatment. Our findings demonstrate that RSD enhances fibrin-rich clot formation after arterial injury by enhancing NET formation, suggesting that NET can be a new therapeutic target in depression-related CVD.
Collapse
Affiliation(s)
- Takeshi Sugimoto
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.S.); (N.W.); (S.M.); (M.S.); (H.K.); (D.M.); (N.W.); (S.M.)
| | - Hiroyuki Yamada
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.S.); (N.W.); (S.M.); (M.S.); (H.K.); (D.M.); (N.W.); (S.M.)
| | - Naotoshi Wada
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.S.); (N.W.); (S.M.); (M.S.); (H.K.); (D.M.); (N.W.); (S.M.)
| | - Shinichiro Motoyama
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.S.); (N.W.); (S.M.); (M.S.); (H.K.); (D.M.); (N.W.); (S.M.)
| | - Makoto Saburi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.S.); (N.W.); (S.M.); (M.S.); (H.K.); (D.M.); (N.W.); (S.M.)
| | - Hiroshi Kubota
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.S.); (N.W.); (S.M.); (M.S.); (H.K.); (D.M.); (N.W.); (S.M.)
| | - Daisuke Miyawaki
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.S.); (N.W.); (S.M.); (M.S.); (H.K.); (D.M.); (N.W.); (S.M.)
| | - Noriyuki Wakana
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.S.); (N.W.); (S.M.); (M.S.); (H.K.); (D.M.); (N.W.); (S.M.)
| | - Daisuke Kami
- Department of Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan;
| | - Takehiro Ogata
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan;
| | - Masakazu Ibi
- Department of Pharmacy, Kinjo Gakuin University, Nagoya 463-8521, Japan;
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.S.); (N.W.); (S.M.); (M.S.); (H.K.); (D.M.); (N.W.); (S.M.)
| |
Collapse
|
184
|
Kaltenmeier C, Yazdani HO, Morder K, Geller DA, Simmons RL, Tohme S. Neutrophil Extracellular Traps Promote T Cell Exhaustion in the Tumor Microenvironment. Front Immunol 2021; 12:785222. [PMID: 34899751 PMCID: PMC8652262 DOI: 10.3389/fimmu.2021.785222] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/09/2021] [Indexed: 12/17/2022] Open
Abstract
While neutrophil extracellular traps (NETs) are important for directly promoting cancer growth, little is known about their impact on immune cells within the tumor microenvironment (TME). We hypothesize that NETs can directly interact with infiltrating T cells to promote an immunosuppressive TME. Herein, to induce a NET-rich TME, we performed liver Ischemia/Reperfusion (I/R) in an established cancer metastasis model or directly injected NETs in subcutaneous tumors. In this NET-rich TME, the majority of CD4+ and CD8+ tumor infiltrating lymphocytes expressed multiple inhibitory receptors, in addition these cells showed a functional and metabolic exhausted phenotype. Targeting of NETs in vivo by treating mice with DNAse lead to decreased tumor growth, decreased NET formation and higher levels of functioning T cells. In vitro, NETs contained the immunosuppressive ligand PD-L1 responsible for T cell exhaustion and dysfunction; an effect abrogated by using PD-L1 KO NETs or culturing NETs with PD-1 KO T cells. Furthermore, we found elevated levels of sPDL-1 and MPO-DNA, a NET marker, in the serum of patients undergoing surgery for colorectal liver metastases resection. Neutrophils isolated from patients after surgery were primed to form NETs and induced exhaustion and dysfunction of human CD4+ and CD8+ T cells. We next targeted PD-L1 in vivo by injecting a blocking antibody during liver I/R. A single dose of anti-PD-L1 during surgery lead to diminished tumors at 3 weeks and functional T cells in the TME. Our data thus reveal that NETs have the capability of suppressing T cell responses through metabolic and functional exhaustion and thereby promote tumor growth. Furthermore, targeting of PD-L1 containing NETs at time of surgery with DNAse or anti-PD-L1 lead to diminished tumor growth, which represents a novel and viable strategy for sustaining immune competence within the TME.
Collapse
Affiliation(s)
| | | | | | | | | | - Samer Tohme
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
185
|
Tang X, Wang P, Zhang R, Watanabe I, Chang E, Vinayachandran V, Nayak L, Lapping S, Liao S, Madera A, Sweet DR, Luo J, Fei J, Jeong HW, Adams RH, Zhang T, Liao X, Jain MK. KLF2 regulates neutrophil activation and thrombosis in cardiac hypertrophy and heart failure progression. J Clin Invest 2021; 132:147191. [PMID: 34793333 PMCID: PMC8803339 DOI: 10.1172/jci147191] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 11/17/2021] [Indexed: 11/22/2022] Open
Abstract
It is widely recognized that inflammation plays a critical role in cardiac hypertrophy and heart failure. However, clinical trials targeting cytokines have shown equivocal effects, indicating the need for a deeper understanding of the precise role of inflammation and inflammatory cells in heart failure. Leukocytes from human subjects and a rodent model of heart failure were characterized by a marked reduction in expression of Klf2 mRNA. Using a mouse model of angiotensin II–induced nonischemic cardiac dysfunction, we showed that neutrophils played an essential role in the pathogenesis and progression of heart failure. Mechanistically, chronic angiotensin II infusion activated a neutrophil KLF2/NETosis pathway that triggered sporadic thrombosis in small myocardial vessels, leading to myocardial hypoxia, cell death, and hypertrophy. Conversely, targeting neutrophils, neutrophil extracellular traps (NETs), or thrombosis ameliorated these pathological changes and preserved cardiac dysfunction. KLF2 regulated neutrophil activation in response to angiotensin II at the molecular level, partly through crosstalk with HIF1 signaling. Taken together, our data implicate neutrophil-mediated immunothrombotic dysregulation as a critical pathogenic mechanism leading to cardiac hypertrophy and heart failure. This neutrophil KLF2-NETosis-thrombosis mechanism underlying chronic heart failure can be exploited for therapeutic gain by therapies targeting neutrophils, NETosis, or thrombosis.
Collapse
Affiliation(s)
- Xinmiao Tang
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, United States of America
| | - Peiwei Wang
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rongli Zhang
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, United States of America
| | - Ippei Watanabe
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, United States of America
| | - Eugene Chang
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, United States of America
| | - Vinesh Vinayachandran
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, United States of America
| | - Lalitha Nayak
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, United States of America
| | - Stephanie Lapping
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, United States of America
| | - Sarah Liao
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, United States of America
| | - Annmarie Madera
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, United States of America
| | - David R Sweet
- Case Western Reserve University, Cleveland, United States of America
| | - Jiemeng Luo
- Cardiology, Minhang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Jinsong Fei
- Cardiology, Minhang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Hyun-Woo Jeong
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine, Münster, Germany
| | - Teng Zhang
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xudong Liao
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, United States of America
| | - Mukesh K Jain
- Case Cardiovascular Research Institute, Case Western Reserve University, Cleveland, United States of America
| |
Collapse
|
186
|
Foret T, Dufrost V, Salomon du Mont L, Costa P, Lakomy C, Lagrange J, Lacolley P, Regnault V, Zuily S, Wahl D. A new pro-thrombotic mechanism of neutrophil extracellular traps in antiphospholipid syndrome: impact on activated protein C resistance. Rheumatology (Oxford) 2021; 61:2993-2998. [PMID: 34791113 DOI: 10.1093/rheumatology/keab853] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/05/2021] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES In antiphospholipid syndrome (APS), precise evaluation of thrombotic risk is a major challenge. Different players, such as activated protein C (APC) resistance or neutrophil extracellular traps (NETs), contribute to the risk of thrombosis. Nevertheless, no study has investigated the interaction between these actors. The main objective of this study was to investigate the relation between NETs and APC resistance. METHODS We designed a cross-sectional study including APS/antiphospholipid antibodies (aPL) patients and patients with autoimmune diseases (AID). We performed thrombin generation tests without and with APC to determine APC resistance. To evaluate circulating NETs, we measured plasma levels of myeloperoxidase-DNA complexes and cell-free DNA with ELISA. RESULTS We recruited 117 patients with definite APS/aPL or AID. We found a positive correlation between NETs and APC resistance, in APS patients and specifically in patients with high thrombotic risk, displaying lupus anticoagulant (LA) or positivity of all 3 aPL tests (triple+), or anti-domain I IgG (aDI+). All these patient subgroups had increased NETs concentrations and APC resistance. As the risk profile for thrombosis increased, the relationship between NETs and APC resistance was stronger. CONCLUSION We have shown that NETs participate in the hypercoagulable state of APS patients by contributing to APC resistance, in particular in high-risk patients. In these most at-risk patients, a targeted action on NETs could reduce APC resistance and constitute a new therapeutic approach in the treatment of APS patients in addition to antithrombotic therapy.
Collapse
Affiliation(s)
- Thomas Foret
- Université de Lorraine, INSERM, DCAC, F-54000 Nancy, France
| | - Virginie Dufrost
- Université de Lorraine, INSERM, DCAC, F-54000 Nancy, France.,CHRU-Nancy, Vascular Medicine Division and Regional Competence Center for Rare Vascular and Systemic Autoimmune Diseases, F-54000 Nancy, France
| | - Lucie Salomon du Mont
- CHRU-Besancon, Vascular and Endovascular Surgery Department, F-25000, Besancon, France.,Université de Bourgogne Franche-Comté, EA3920, F-25000, Besancon, France
| | - Patricia Costa
- CHRU-Besancon, Vascular Medicine Unit, Vascular and Endovascular Surgery Department, F-25000 Besancon, France
| | - Cecile Lakomy
- Université de Lorraine, INSERM, DCAC, F-54000 Nancy, France
| | | | | | | | - Stephane Zuily
- Université de Lorraine, INSERM, DCAC, F-54000 Nancy, France.,CHRU-Nancy, Vascular Medicine Division and Regional Competence Center for Rare Vascular and Systemic Autoimmune Diseases, F-54000 Nancy, France
| | - Denis Wahl
- Université de Lorraine, INSERM, DCAC, F-54000 Nancy, France.,CHRU-Nancy, Vascular Medicine Division and Regional Competence Center for Rare Vascular and Systemic Autoimmune Diseases, F-54000 Nancy, France
| |
Collapse
|
187
|
Ciepiela O, Małecka-Giełdowska M, Czyżewska E. Neutrophil Extracellular Traps (NETs) and Hypercoagulability in Plasma Cell Dyscrasias-Is This Phenomenon Worthy of Exploration? J Clin Med 2021; 10:jcm10225243. [PMID: 34830525 PMCID: PMC8624472 DOI: 10.3390/jcm10225243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022] Open
Abstract
Plasma cell dyscrasias (PCDs) are neoplastic diseases derived from plasma cells. Patients suffering from PCDs are at high risk of hypercoagulability and thrombosis. These conditions are associated with disease-related factors, patient-related factors, or the use of immunomodulatory drugs. As PCDs belong to neoplastic diseases, some other factors related to the cancer-associated hypercoagulability state in the course of PCDs are also considered. One of the weakest issues studied in PCDs is the procoagulant activity of neutrophil extracellular traps (NETs). NETs are web-like structures released from neutrophils in response to different stimuli. These structures are made of deoxyribonucleic acid (DNA) and bactericidal proteins, such as histones, myeloperoxidase, neutrophil elastase, and over 300 other proteins, which are primarily stored in neutrophil granules. NETs immobilize, inactivate the pathogens, and expose them to specialized cells of immune response. Despite their pivotal role in innate immunity, they contribute to the development and exacerbation of autoimmune diseases, trigger inflammatory response, or even facilitate the formation of cancer metastases. NETs were also found to induce activity of coagulation and are considered one of the most important factors inducing thrombosis. Here, we summarize how PCDs influence the release of NETs, and hypothesize whether NETs contribute to hypercoagulability in PCDs patients.
Collapse
|
188
|
Neutrophil Extracellular Traps Exacerbate Ischemic Brain Damage. Mol Neurobiol 2021; 59:643-656. [PMID: 34748205 DOI: 10.1007/s12035-021-02635-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 11/02/2021] [Indexed: 12/11/2022]
Abstract
Most acute strokes are ischemic, and subsequent neuroinflammation promotes further damage leading to cell death but also plays a beneficial role by promoting cellular repair. Neutrophils are forerunners to brain lesions after ischemic stroke and exert elaborate functions. While neutrophil extracellular traps (NETs) possess a fundamental antimicrobial function within the innate immune system under physiological circumstances, increasing evidence indicates that NETosis, the release process of NETs, occurs in the pathogenic process of stroke. In this review, we focus on the processes of NET formation and clearance, the temporal and spatial alterations of neutrophils and NETs after ischemic damage, and how NETs are involved in several stroke-related phenomena. Generally, NET formation and release processes depend on the generation of reactive oxygen species (ROS) and the activation of nuclear peptidylarginine deiminase-4 (PAD4). The acid-base environment, oxygen concentration, and iron ions around the infarct may also impact NET formation. DNase 1 has been identified as the primary degrader of NETs in serum, while reactive microglia are expected to inhibit the formation of NETs around ischemic lesions by phagocytosis of neutrophils. The neutrophils and NETs are present in the perivascular space ipsilateral to the infarct arising after ischemic damage, peaking between 1 and 3 days postischemia, but their location in the brain parenchyma remains controversial. After the ischemic injury, NETs are involved in the destruction of neurological function primarily by disrupting the blood-brain barrier and promoting thrombosis. The potential effects of NETs on various ischemic nerve cells need to be further investigated, especially in the chronic ischemic phase.
Collapse
|
189
|
Jha V, Kumari T, Manickam V, Assar Z, Olson KL, Min JK, Cho J. ERO1-PDI Redox Signaling in Health and Disease. Antioxid Redox Signal 2021; 35:1093-1115. [PMID: 34074138 PMCID: PMC8817699 DOI: 10.1089/ars.2021.0018] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Significance: Protein disulfide isomerase (PDI) and endoplasmic reticulum oxidoreductase 1 (ERO1) are crucial for oxidative protein folding in the endoplasmic reticulum (ER). These enzymes are frequently overexpressed and secreted, and they contribute to the pathology of neurodegenerative, cardiovascular, and metabolic diseases. Recent Advances: Tissue-specific knockout mouse models and pharmacologic inhibitors have been developed to advance our understanding of the cell-specific functions of PDI and ERO1. In addition to their roles in protecting cells from the unfolded protein response and oxidative stress, recent studies have revealed that PDI and ERO1 also function outside of the cells. Critical Issues: Despite the well-known contributions of PDI and ERO1 to specific disease pathology, the detailed molecular and cellular mechanisms underlying these activities remain to be elucidated. Further, although PDI and ERO1 inhibitors have been identified, the results from previous studies require careful evaluation, as many of these agents are not selective and may have significant cytotoxicity. Future Directions: The functions of PDI and ERO1 in the ER have been extensively studied. Additional studies will be required to define their functions outside the ER.
Collapse
Affiliation(s)
- Vishwanath Jha
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tripti Kumari
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Vijayprakash Manickam
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zahra Assar
- Cayman Chemical Company, Inc., Ann Arbor, Michigan, USA
| | - Kirk L Olson
- Cayman Chemical Company, Inc., Ann Arbor, Michigan, USA
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
190
|
Netting Gut Disease: Neutrophil Extracellular Trap in Intestinal Pathology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5541222. [PMID: 34712384 PMCID: PMC8548149 DOI: 10.1155/2021/5541222] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/04/2021] [Accepted: 09/29/2021] [Indexed: 12/26/2022]
Abstract
Many gut disease etiologies are attributed to the presence of robust inflammatory cell recruitment. The recruitment of neutrophils plays a vital role in inflammatory infiltration. Neutrophils have various antimicrobial effector mechanisms, including phagocytosis, oxidative burst, and degranulation. It is suggested that neutrophils could release neutrophil extracellular traps (NETs) to kill pathogens. However, recent evidence indicates that neutrophil infiltration within the gut is associated with disrupted local immunological microenvironment and impaired epithelial barrier. Growing evidence implies that NETs are involved in the progression of many diseases, including cancer, diabetes, thrombosis, and autoimmune disease. Increased NET formation was found in acute or chronic conditions, including infection, sterile inflammation, cancer, and ischemia/reperfusion injury (IRI). Here, we present a comprehensive review of recent advances in the understanding of NETs, focusing on their effects in gut disease. We also discuss NETs as a potential therapeutic target in gut disease.
Collapse
|
191
|
Jin K, Bardes EE, Mitelpunkt A, Wang JY, Bhatnagar S, Sengupta S, Krummel DP, Rothenberg ME, Aronow BJ. An interactive single cell web portal identifies gene and cell networks in COVID-19 host responses. iScience 2021; 24:103115. [PMID: 34522848 PMCID: PMC8428985 DOI: 10.1016/j.isci.2021.103115] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/17/2021] [Accepted: 09/08/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous studies have provided single-cell transcriptome profiles of host responses to SARS-CoV-2 infection. Critically lacking however is a data mine that allows users to compare and explore cell profiles to gain insights and develop new hypotheses. To accomplish this, we harmonized datasets from COVID-19 and other control condition blood, bronchoalveolar lavage, and tissue samples, and derived a compendium of gene signature modules per cell type, subtype, clinical condition, and compartment. We demonstrate approaches to interacting with, exploring, and functional evaluating these modules via a new interactive web portal ToppCell (http://toppcell.cchmc.org/). As examples, we develop three hypotheses: (1) alternatively-differentiated monocyte-derived macrophages form a multicelllar signaling cascade that drives T cell recruitment and activation; (2) COVID-19-generated platelet subtypes exhibit dramatically altered potential to adhere, coagulate, and thrombose; and (3) extrafollicular B maturation is driven by a multilineage cell activation network that expresses an ensemble of genes strongly associated with risk for developing post-viral autoimmunity.
Collapse
Affiliation(s)
- Kang Jin
- Division of Biomedical Informatics, Cincinnati Children’s Hospital
Medical Center, Cincinnati, OH 45229, USA
- Department of Biomedical Informatics, University of Cincinnati,
Cincinnati, OH 45229, USA
| | - Eric E. Bardes
- Division of Biomedical Informatics, Cincinnati Children’s Hospital
Medical Center, Cincinnati, OH 45229, USA
| | - Alexis Mitelpunkt
- Division of Biomedical Informatics, Cincinnati Children’s Hospital
Medical Center, Cincinnati, OH 45229, USA
- Pediatric Rehabilitation, Dana-Dwek Children's Hospital, Tel Aviv Medical
Center, Tel Aviv, 6423906, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 6997801,
Israel
| | - Jake Y. Wang
- Division of Biomedical Informatics, Cincinnati Children’s Hospital
Medical Center, Cincinnati, OH 45229, USA
| | - Surbhi Bhatnagar
- Division of Biomedical Informatics, Cincinnati Children’s Hospital
Medical Center, Cincinnati, OH 45229, USA
- Department of Electrical Engineering and Computer Science, University of
Cincinnati, Cincinnati, OH 45221, USA
| | - Soma Sengupta
- Department of Neurology and Rehabilitation Medicine, University of
Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Daniel Pomeranz Krummel
- Department of Neurology and Rehabilitation Medicine, University of
Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Marc E. Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati
Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH
45229, USA
| | - Bruce J. Aronow
- Division of Biomedical Informatics, Cincinnati Children’s Hospital
Medical Center, Cincinnati, OH 45229, USA
- Department of Electrical Engineering and Computer Science, University of
Cincinnati, Cincinnati, OH 45221, USA
- Department of Pediatrics, University of Cincinnati School of Medicine,
Cincinnati, OH 45256, USA
| |
Collapse
|
192
|
Korkmaz B, Lamort AS, Domain R, Beauvillain C, Gieldon A, Yildirim AÖ, Stathopoulos GT, Rhimi M, Jenne DE, Kettritz R. Cathepsin C inhibition as a potential treatment strategy in cancer. Biochem Pharmacol 2021; 194:114803. [PMID: 34678221 DOI: 10.1016/j.bcp.2021.114803] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 02/08/2023]
Abstract
Epidemiological studies established an association between chronic inflammation and higher risk of cancer. Inhibition of proteolytic enzymes represents a potential treatment strategy for cancer and prevention of cancer metastasis. Cathepsin C (CatC) is a highly conserved lysosomal cysteine dipeptidyl aminopeptidase required for the activation of pro-inflammatory neutrophil serine proteases (NSPs, elastase, proteinase 3, cathepsin G and NSP-4). NSPs are locally released by activated neutrophils in response to pathogens and non-infectious danger signals. Activated neutrophils also release neutrophil extracellular traps (NETs) that are decorated with several neutrophil proteins, including NSPs. NSPs are not only NETs constituents but also play a role in NET formation and release. Although immune cells harbor large amounts of CatC, additional cell sources for this protease exists. Upregulation of CatC expression was observed in different tissues during carcinogenesis and correlated with metastasis and poor patient survival. Recent mechanistic studies indicated an important interaction of tumor-associated CatC, NSPs, and NETs in cancer development and metastasis and suggested CatC as a therapeutic target in a several cancer types. Cancer cell-derived CatC promotes neutrophil recruitment in the inflammatory tumor microenvironment. Because the clinical consequences of genetic CatC deficiency in humans resulting in the elimination of NSPs are mild, small molecule inhibitors of CatC are assumed as safe drugs to reduce the NSP burden. Brensocatib, a nitrile CatC inhibitor is currently tested in a phase 3 clinical trial as a novel anti-inflammatory therapy for patients with bronchiectasis. However, recently developed CatC inhibitors possibly have protective effects beyond inflammation. In this review, we describe the pathophysiological function of CatC and discuss molecular mechanisms substantiating pharmacological CatC inhibition as a potential strategy for cancer treatment.
Collapse
Affiliation(s)
- Brice Korkmaz
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and University of Tours, 37032 Tours, France.
| | - Anne-Sophie Lamort
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), Helmholtz Center Munich-German Research Center for Environmental Health (HMGU) and Ludwig-Maximilian-University (LMU), Munich, Bavaria 81377, Germany(2)
| | - Roxane Domain
- INSERM UMR-1100, "Research Center for Respiratory Diseases" and University of Tours, 37032 Tours, France
| | - Céline Beauvillain
- University of Angers, University of Nantes, Angers University Hospital, INSERM UMR-1232, CRCINA, Innate Immunity and Immunotherapy, SFR ICAT, 49000 Angers, France
| | - Artur Gieldon
- Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), Helmholtz Center Munich-German Research Center for Environmental Health (HMGU) and Ludwig-Maximilian-University (LMU), Munich, Bavaria 81377, Germany(2)
| | - Georgios T Stathopoulos
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), Helmholtz Center Munich-German Research Center for Environmental Health (HMGU) and Ludwig-Maximilian-University (LMU), Munich, Bavaria 81377, Germany(2)
| | - Moez Rhimi
- Microbiota Interaction with Human and Animal Team (MIHA), Micalis Institute, AgroParisTech, Université Paris-Saclay, INRAE, Jouy-en-Josas, France
| | - Dieter E Jenne
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD), Helmholtz Center Munich-German Research Center for Environmental Health (HMGU) and Ludwig-Maximilian-University (LMU), Munich, Bavaria 81377, Germany(2); Max Planck Institute of Neurobiology, 82152 Planegg-Martinsried, Germany
| | - Ralph Kettritz
- Experimental and Clinical Research Center, Charité und Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtz-Gemeinschaft (MDC), Berlin, Germany; Nephrology and Intensive Care Medicine, Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
193
|
Yaqinuddin A. Neutrophil extracellular traps and thrombogenesis in COVID-19 patients. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2021; 26:96. [PMID: 34899934 PMCID: PMC8607181 DOI: 10.4103/jrms.jrms_750_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 12/30/2020] [Accepted: 02/16/2021] [Indexed: 06/14/2023]
Abstract
COVID-19 has caused significant morbidity and mortality around the world. Recent reports point toward the "cytokine storm" as core of pathogenesis in SAR-CoV-2-induced acute lung injury, acute respiratory distress syndrome (ARDS), coagulopathy, and multiorgan failure. We have presented clinical data here wherein cytokine levels in COVID-19 patients do not match typical cytokine storm seen in ARDS. Interestingly, COVID-19 patients in early disease present with hypoxemia with no significant respiratory dysfunction. In addition, it is reported that hospitalized COVID-19 patients have a high incidence of thrombotic complications, especially involving the pulmonary vasculature. We hypothesized that core to pathogenesis of COVID-19 is the dysregulation of neutrophils, which culminates in excessive release of neutrophil extracellular traps (NETs). Recently, an increasing amount of NETs have been seen in sera of severe COVID-19 patients. We have discussed here mechanisms involved which lead to thrombogenesis and vasculitis because of excessive release of NETs.
Collapse
Affiliation(s)
- Ahmed Yaqinuddin
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
194
|
Mutua V, Gershwin LJ. A Review of Neutrophil Extracellular Traps (NETs) in Disease: Potential Anti-NETs Therapeutics. Clin Rev Allergy Immunol 2021; 61:194-211. [PMID: 32740860 PMCID: PMC7395212 DOI: 10.1007/s12016-020-08804-7] [Citation(s) in RCA: 358] [Impact Index Per Article: 89.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Activated neutrophils release neutrophil extracellular traps (NETs) in response to a variety of stimuli. NETosis is driven by protein-arginine deiminase type 4, with the release of intracellular granule components that function by capturing and destroying microbes, including viral, fungal, bacterial, and protozoal pathogens. The positive effects of pathogen control are countered by pro-inflammatory effects as demonstrated in a variety of diseases. Components of NETS are non-specific, and other than controlling microbes, they cause injury to surrounding tissue by themselves or by increasing the pro-inflammatory response. NETs can play a role in enhancement of the inflammation seen in autoimmune diseases including psoriasis, rheumatoid arthritis, and systemic lupus erythematosis. In addition, autoinflammatory diseases such as gout have been associated with NETosis. Inhibition of NETs may decrease the severity of many diseases improving survival. Herein, we describe NETosis in different diseases focusing on the detrimental effect of NETs and outline possible therapeutics that can be used to mitigate netosis. There is a need for more studies and clinical trials on these and other compounds that could prevent or destroy NETs, thereby decreasing damage to patients.
Collapse
Affiliation(s)
- Victoria Mutua
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, 1 Shields Ave, Davis, CA, USA.
| | - Laurel J Gershwin
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, 1 Shields Ave, Davis, CA, USA
| |
Collapse
|
195
|
Montinari MR, Minelli S, De Caterina R. Eighty years of oral anticoagulation: Learning from history. Vascul Pharmacol 2021; 141:106918. [PMID: 34537376 DOI: 10.1016/j.vph.2021.106918] [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: 08/10/2021] [Revised: 09/11/2021] [Accepted: 09/14/2021] [Indexed: 02/05/2023]
Abstract
In the year 2021 we celebrate the 80th anniversary of the first clinical use of vitamin K antagonists (VKAs), the mainstay of prevention and long-term treatment of thromboembolic disease. The discovery and development of oral anticoagulants is one of the most important chapters in the history of medicine, a goal pursued by physicians trying to combat the clinical manifestations of thrombosis since ancient times. Until the last decade, VKAs were the only oral anticoagulants available and used in clinical practice. Today, their clinical use has progressively shrunk, as the non-vitamin K antagonist oral anticoagulants (NOACs) are increasingly replacing VKAs in various conditions after the successful completion of several large randomized controlled trials. Currently, new research is tackling upstream components of the intrinsic pathway - particularly factor XI and factor XII - for the development of new, even safer anticoagulants promising to reduce bleeding without compromising efficacy. This review highlights the evolution of oral anticoagulant therapy tracing the key stages of a long and fascinating history that has unfolded from the first part of the twentieth century until today, indeed an intriguing journey where serendipity is intertwined with the tenacious work of many researchers.
Collapse
Affiliation(s)
- Maria Rosa Montinari
- Chair of History of Medicine, Department of Biological and Environmental Science and Technology, University of Salento, Lecce, Italy
| | | | - Raffaele De Caterina
- Chair of Cardiology, University of Pisa, University Cardiology Division, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Fondazione VillaSerena, Città Sant'Angelo, Pescara, Italy.
| |
Collapse
|
196
|
Chen X, Liu F, Xue Q, Weng X, Xu F. Metastatic pancreatic cancer: Mechanisms and detection (Review). Oncol Rep 2021; 46:231. [PMID: 34498718 PMCID: PMC8444192 DOI: 10.3892/or.2021.8182] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer (PC) is a lethal malignancy. Its prevalence rate remains low but continues to grow each year. Among all stages of PC, metastatic PC is defined as late-stage (stage IV) PC and has an even higher fatality rate. Patients with PC do not have any specific clinical manifestations. Most cases are inoperable at the time-point of diagnosis. Prognosis is also poor even with curative-intent surgery. Complications during surgery, postoperative pancreatic fistula and recurrence with metastatic foci make the management of metastatic PC difficult. While extensive efforts were made to improve survival outcomes, further elucidation of the molecular mechanisms of metastasis poses a formidable challenge. The present review provided an overview of the mechanisms of metastatic PC, summarizing currently known signaling pathways (e.g. epithelial-mesenchymal transition, NF-κB and KRAS), imaging that may be utilized for early detection and biomarkers (e.g. carbohydrate antigen 19-9, prostate cancer-associated transcript-1, F-box/LRR-repeat protein 7 and tumor stroma), giving insight into promising therapeutic targets.
Collapse
Affiliation(s)
- Xiangling Chen
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Fangfang Liu
- Department of Art, Art College, Southwest Minzu University, Chengdu, Sichuan 610041, P.R. China
| | - Qingping Xue
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Xiechuan Weng
- Department of Neuroscience, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Fan Xu
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| |
Collapse
|
197
|
Zenlander R, Havervall S, Magnusson M, Engstrand J, Ågren A, Thålin C, Stål P. Neutrophil extracellular traps in patients with liver cirrhosis and hepatocellular carcinoma. Sci Rep 2021; 11:18025. [PMID: 34504150 PMCID: PMC8429678 DOI: 10.1038/s41598-021-97233-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 08/20/2021] [Indexed: 12/20/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are web-like structures consisting of DNA, histones and granule proteins, released from neutrophils in thrombus formation, inflammation, and cancer. We asked if plasma levels of the NET markers myeloperoxidase (MPO)-DNA and citrullinated histone H3 (H3Cit)-DNA, are elevated in liver cirrhosis and hepatocellular carcinoma (HCC) and if the levels correlate with clinical parameters. MPO-DNA, H3Cit-DNA, and thrombin–antithrombin (TAT) complex, as a marker of coagulation activity, were measured using ELISA in plasma from 82 patients with HCC, 95 patients with cirrhosis and 50 healthy controls. Correlations were made to clinical parameters and laboratory data and patients were followed for a median of 22.5 months regarding thrombosis development. H3Cit-DNA was significantly (p < 0.01) elevated in plasma from cirrhosis (66.4 ng/mL) and HCC (63.8 ng/mL) patients compared to healthy controls (31.8 ng/mL). TAT levels showed similar pattern (3.1, 3.7, and 0.0 µg/mL respectively, p < 0.01). MPO-DNA was significantly (p < 0.01) elevated in cirrhosis patients (0.53 O.D.) as compared to controls (0.33 O.D.). Levels of MPO-DNA and H3Cit-DNA correlated positively with Child–Pugh and MELD score. TAT was increased in all Child–Pugh and MELD groups. In multivariable logistic regression, Child B and C liver cirrhosis were independent predictors of elevated H3Cit-DNA in plasma. Levels of MPO-DNA and H3Cit-DNA were similar in patients with or without history of thrombosis, or thrombus formation during follow-up. In conclusion, plasma markers of NET formation are elevated in liver cirrhosis and correlate to the degree of liver dysfunction in patients with liver cirrhosis and/or HCC. The presence of HCC did not further increase the plasma levels of NET markers as compared to patients with cirrhosis only.
Collapse
Affiliation(s)
- Robin Zenlander
- Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden. .,Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden. .,Department of Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden.
| | - Sebastian Havervall
- Division of Gastroenterology, Department of Specialized Medicine, Danderyd Hospital, Stockholm, Sweden.,Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Maria Magnusson
- Division of Pediatrics, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Jennie Engstrand
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Ågren
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Charlotte Thålin
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden.,Department of Internal Medicine and Infectious Diseases, Danderyd Hospital, Stockholm, Sweden
| | - Per Stål
- Department of Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden.,Division of Hepatology, Department of Upper GI Diseases, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
198
|
Makatsariya AD, Slukhanchuk EV, Bitsadze VO, Khizroeva JK, Tretyakova MV, Makatsariya NA, Akinshina SV, Shkoda AS, Pankratyeva LL, Di Renzo GC, Rizzo G, Grigorieva KN, Tsibizova VI, Gris JC, Elalamy I. Neutrophil extracellular traps: a role in inflammation and dysregulated hemostasis as well as in patients with COVID-19 and severe obstetric pathology. OBSTETRICS, GYNECOLOGY AND REPRODUCTION 2021; 15:335-350. [DOI: 10.17749/2313-7347/ob.gyn.rep.2021.238] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Numerous studies have proven a close relationship between inflammatory diseases and the state of hypercoagulability. In fact, thromboembolic complications represent one of the main causes of disability and mortality in acute and chronic inflammatory diseases, cancer and obstetric complications. Despite this, the processes of hemostasis and immune responses have long been considered separately; currently, work is underway to identify the molecular basis for a relationship between such systems. It has been identified that various pro-inflammatory stimuli are capable of triggering a coagulation cascade, which in turn modulates inflammatory responses. Neutrophil extracellular traps (NETs) are the networks of histones of extracellular DNA generated by neutrophils in response to inflammatory stimuli. The hemostasis is activated against infection in order to minimize the spread of infection and, if possible, inactivate the infectious agent. Another molecular network is based on fibrin. Over the last 10 years, there has been accumulated a whole body of evidence that NETs and fibrin are able to form a united network within a thrombus, stabilizing each other. Similarities and molecular cross-reactions are also present in the processes of fibrinolysis and lysis of NETs. Both NETs and von Willebrand factor (vWF) are involved in thrombosis as well as inflammation. During the development of these conditions, a series of events occurs in the microvascular network, including endothelial activation, NETs formation, vWF secretion, adhesion, aggregation, and activation of blood cells. The activity of vWF multimers is regulated by the specific metalloproteinase ADAMTS-13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13). Studies have shown that interactions between NETs and vWF can lead to arterial and venous thrombosis and inflammation. In addition, the contents released from activated neutrophils or NETs result in decreased ADAMTS-13 activity, which can occur in both thrombotic microangiopathies and acute ischemic stroke. Recently, NETs have been envisioned as a cause of endothelial damage and immunothrombosis in COVID-19. In addition, vWF and ADAMTS-13 levels predict COVID-19 mortality. In this review, we summarize the biological characteristics and interactions of NETs, vWF, and ADAMTS-13, the effect of NETs on hemostasis regulation and discuss their role in thrombotic conditions, sepsis, COVID-19, and obstetric complications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - A. S. Shkoda
- Vorokhobov City Clinical Hospital № 67, Moscow Healthcare Department
| | - L. L. Pankratyeva
- Vorokhobov City Clinical Hospital № 67, Moscow Healthcare Department; Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Health Ministry of Russian Federation
| | - G. C. Di Renzo
- Sechenov University; Center for Prenatal and Reproductive Medicine, University of Perugia
| | - G. Rizzo
- Sechenov University; University of Rome Tor Vergata
| | | | - V. I. Tsibizova
- Almazov National Medical Research Centre, Health Ministry of Russian Federation
| | - J.-C. Gris
- Sechenov University; University of Montpellier
| | - I. Elalamy
- Sechenov University; Medicine Sorbonne University; Hospital Tenon
| |
Collapse
|
199
|
Nader E, Garnier Y, Connes P, Romana M. Extracellular Vesicles in Sickle Cell Disease: Plasma Concentration, Blood Cell Types Origin Distribution and Biological Properties. Front Med (Lausanne) 2021; 8:728693. [PMID: 34490315 PMCID: PMC8417591 DOI: 10.3389/fmed.2021.728693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/30/2021] [Indexed: 01/08/2023] Open
Abstract
Prototype of monogenic disorder, sickle cell disease (SCD) is caused by a unique single mutation in the β-globin gene, leading to the production of the abnormal hemoglobin S (HbS). HbS polymerization in deoxygenated condition induces the sickling of red blood cells (RBCs), which become less deformable and more fragile, and thus prone to lysis. In addition to anemia, SCD patients may exhibit a plethora of clinical manifestations ranging from acute complications such as the frequent and debilitating painful vaso-occlusive crisis to chronic end organ damages. Several interrelated pathophysiological processes have been described, including impaired blood rheology, increased blood cell adhesion, coagulation, inflammation and enhanced oxidative stress among others. During the last two decades, it has been shown that extracellular vesicles (EVs), defined as cell-derived anucleated particles delimited by a lipid bilayer, and comprising small EVs (sEVs) and medium/large EVs (m/lEVs); are not only biomarkers but also subcellular actors in SCD pathophysiology. Plasma concentration of m/lEVs, originated mainly from RBCs and platelets (PLTs) but also from the other blood cell types, is higher in SCD patients than in healthy controls. The concentration and the density of externalized phosphatidylserine of those released from RBCs may vary according to clinical status (crisis vs. steady state) and treatment (hydroxyurea). Besides their procoagulant properties initially described, RBC-m/lEVs may promote inflammation through their effects on monocytes/macrophages and endothelial cells. Although less intensely studied, sEVs plasma concentration is increased in SCD and these EVs may cause endothelial damages. In addition, sEVs released from activated PLTs trigger PLT-neutrophil aggregation involved in lung vaso-occlusion in sickle mice. Altogether, these data clearly indicate that EVs are both biomarkers and bio-effectors in SCD, which deserve further studies.
Collapse
Affiliation(s)
- Elie Nader
- Laboratoire Inter-Universitaire de Biologie de la Motricité EA7424, Team "Vascular Biology and Red Blood Cell", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge, PRES Sorbonne, Paris, France
| | - Yohann Garnier
- Laboratoire d'Excellence du Globule Rouge, PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France
| | - Philippe Connes
- Laboratoire Inter-Universitaire de Biologie de la Motricité EA7424, Team "Vascular Biology and Red Blood Cell", Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France.,Laboratoire d'Excellence du Globule Rouge, PRES Sorbonne, Paris, France
| | - Marc Romana
- Laboratoire d'Excellence du Globule Rouge, PRES Sorbonne, Paris, France.,Université des Antilles, UMR_S1134, BIGR, Pointe-à-Pitre, France.,Université de Paris, UMR_S1134, BIGR, INSERM, Paris, France
| |
Collapse
|
200
|
Neutrophil and Eosinophil Extracellular Traps in Hodgkin Lymphoma. Hemasphere 2021; 5:e633. [PMID: 34485830 PMCID: PMC8410234 DOI: 10.1097/hs9.0000000000000633] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/12/2021] [Indexed: 12/15/2022] Open
Abstract
Classic Hodgkin lymphoma (cHL), nodular sclerosis (NS) subtype, is characterized by the presence of Hodgkin/Reed-Sternberg (HRS) cells in an inflammatory background containing neutrophils and/or eosinophils. Both types of granulocytes release extracellular traps (ETs), web-like DNA structures decorated with histones, enzymes, and coagulation factors that promote inflammation, thrombosis, and tumor growth. We investigated whether ETs from neutrophils (NETs) or eosinophils (EETs) are detected in cHL, and evaluated their association with fibrosis. We also studied expression of protease-activated receptor-2 (PAR-2) and phospho-extracellular signal-related kinase (p-ERK), potential targets/effectors of ETs-associated elastase, in HRS cells. Expression of tissue factor (TF) was evaluated, given the procoagulant properties of ETs. We analyzed 32 HL cases, subclassified as 12 NS, 5 mixed-cellularity, 5 lymphocyte-rich, 1 lymphocyte-depleted, 4 nodular lymphocyte-predominant HL (NLPHL), and 5 reactive nodes. Notably, a majority of NS cHL cases exhibited NET formation by immunohistochemistry for citrullinated histones, with 1 case revealing abundant EETs. All other cHL subtypes as well as NLPHL were negative. Immunofluorescence microscopy confirmed NETs with filamentous/delobulated morphology. Moreover, ETs formation correlates with concurrent fibrosis (r = 0.7999; 95% CI, 0.6192-0.9002; P ≤ 0.0001). Results also showed that HRS cells in NS cHL expressed PAR-2 with nuclear p-ERK staining, indicating a neoplastic or inflammatory phenotype. Remarkably, TF was consistently detected in the endothelium of NS cHL cases compared with other subtypes, in keeping with a procoagulant status. A picture emerges whereby the release of ETs and resultant immunothrombosis contribute to the inflammatory tumor microenvironment of NS cHL. This is the first description of NETs in cHL.
Collapse
|