251
|
Becker K, Beythien G, de Buhr N, Stanelle-Bertram S, Tuku B, Kouassi NM, Beck S, Zickler M, Allnoch L, Gabriel G, von Köckritz-Blickwede M, Baumgärtner W. Vasculitis and Neutrophil Extracellular Traps in Lungs of Golden Syrian Hamsters With SARS-CoV-2. Front Immunol 2021; 12:640842. [PMID: 33912167 PMCID: PMC8072219 DOI: 10.3389/fimmu.2021.640842] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/19/2021] [Indexed: 12/13/2022] Open
Abstract
Neutrophil extracellular traps (NETs) have been identified as one pathogenetic trigger in severe COVID-19 cases and therefore well-described animal models to understand the influence of NETs in COVID-19 pathogenesis are needed. SARS-CoV-2 infection causes infection and interstitial pneumonia of varying severity in humans and COVID-19 models. Pulmonary as well as peripheral vascular lesions represent a severe, sometimes fatal, disease complication of unknown pathogenesis in COVID-19 patients. Furthermore, neutrophil extracellular traps (NETs), which are known to contribute to vessel inflammation or endothelial damage, have also been shown as potential driver of COVID-19 in humans. Though most studies in animal models describe the pulmonary lesions characterized by interstitial inflammation, type II pneumocyte hyperplasia, edema, fibrin formation and infiltration of macrophages and neutrophils, detailed pathological description of vascular lesions or NETs in COVID-19 animal models are lacking so far. Here we report different types of pulmonary vascular lesions in the golden Syrian hamster model of COVID-19. Vascular lesions included endothelialitis and vasculitis at 3 and 6 days post infection (dpi), and were almost nearly resolved at 14 dpi. Importantly, virus antigen was present in pulmonary lesions, but lacking in vascular alterations. In good correlation to these data, NETs were detected in the lungs of infected animals at 3 and 6 dpi. Hence, the Syrian hamster seems to represent a useful model to further investigate the role of vascular lesions and NETs in COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Kathrin Becker
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Georg Beythien
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nicole de Buhr
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany.,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Stephanie Stanelle-Bertram
- Department for Viral Zoonoses-One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Berfin Tuku
- Department for Viral Zoonoses-One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Nancy Mounogou Kouassi
- Department for Viral Zoonoses-One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Sebastian Beck
- Department for Viral Zoonoses-One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Martin Zickler
- Department for Viral Zoonoses-One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Lisa Allnoch
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Gülsah Gabriel
- Department for Viral Zoonoses-One Health, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany.,Institute for Virology, University for Veterinary Medicine Hannover, Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany.,Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| |
Collapse
|
252
|
Uludag G, Onghanseng N, Tran ANT, Hassan M, Halim MS, Sepah YJ, Do DV, Nguyen QD. Current concepts in the diagnosis and management of antiphospholipid syndrome and ocular manifestations. J Ophthalmic Inflamm Infect 2021; 11:11. [PMID: 33834305 PMCID: PMC8032459 DOI: 10.1186/s12348-021-00240-8] [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: 05/24/2020] [Accepted: 03/02/2021] [Indexed: 12/18/2022] Open
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disorder associated with obstetrical complications, thrombotic complications involving both arteries and veins, and non-thrombotic manifestations affecting multiple other systems presenting in various clinical forms. Diagnosis requires the presence of antiphospholipid antibodies. The exact pathogenesis of APS is not fully known. However, it has recently been shown that activation of different types of cells by antiphospholipid antibodies plays an important role in thrombosis formation. Ocular involvement is one of the important clinical manifestations of APS and can vary in presentations. Therefore, as an ophthalmologist, it is crucial to be familiar with the ocular findings of APS to prevent further complications that can develop. Furthermore, the ongoing identification of new and specific factors contributing to the pathogenesis of APS may provide new therapeutic options in the management of the disease in the future.
Collapse
Affiliation(s)
- Gunay Uludag
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA
| | - Neil Onghanseng
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA
| | - Anh N T Tran
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA
| | - Muhammad Hassan
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA
| | - Muhammad Sohail Halim
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA.,Ocular Imaging Research and Reading Center, Sunnyvale, CA, USA
| | - Yasir J Sepah
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA.,Ocular Imaging Research and Reading Center, Sunnyvale, CA, USA
| | - Diana V Do
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA
| | - Quan Dong Nguyen
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA.
| |
Collapse
|
253
|
Kawano T, Sasaki T, Gon Y, Kitano T, Kanki H, Todo K, Shimamura M, Matsumura Y, Huang A, Hattori S, Mochizuki H. High neutrophil/lymphocyte ratio at cancer diagnosis predicts incidence of stroke in cancer patients. Brain Commun 2021; 3:fcab071. [PMID: 33928250 PMCID: PMC8062330 DOI: 10.1093/braincomms/fcab071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2021] [Indexed: 01/09/2023] Open
Abstract
Although cancer increases the incidence and severity of ischaemic stroke, there is no reliable method for predicting ischaemic stroke in cancer patients. To evaluate the prognostic capacity of the neutrophil-to-lymphocyte ratio at cancer diagnosis for predicting the incidence of ischaemic stroke, we used a hospital-based cancer registry that contained clinical data from all patients treated for cancer at Osaka University Hospital between 2007 and 2015. The neutrophil-to-lymphocyte ratio was calculated after dividing absolute neutrophil counts by absolute lymphocyte counts. These counts were obtained within 1 month after cancer diagnosis. The primary endpoint was new-onset ischaemic stroke within 2 years after cancer diagnosis. Of the 18 217 included cancer patients (median age: 65.2 years), 69 (0.38%) had ischaemic stroke. Unadjusted Cox regression analysis stratified by cancer site demonstrated that each 1-unit increase in the neutrophil-to-lymphocyte ratio was associated with a significant 7.2% increase in the risk of an ischaemic stroke event (95% confidence interval 1.041–1.103, P < 0.001). Survival tree analysis and the Kaplan–Meier method suggested that patients with and without atrial fibrillation who had increased neutrophil-to-lymphocyte ratios had a higher risk of ischaemic stroke. Multivariate Cox proportional hazard models, adjusted for cancer site and stage, revealed that patients with high neutrophil-to-lymphocyte ratios (>15) had higher ischaemic stroke risk than patients with low neutrophil-to-lymphocyte ratios (<5). This was true among cancer patients both with (hazard ratio 11.598; 95% confidence interval 0.953–141.181) and without (hazard ratio 7.877; 95% confidence interval 2.351–26.389) atrial fibrillation. The neutrophil-to-lymphocyte ratio at cancer diagnosis is associated with the incidence of ischaemic stroke among cancer patients and might thus be useful for identifying patients at high risk of ischaemic stroke, allowing us to guide future preventive interventions.
Collapse
Affiliation(s)
- Tomohiro Kawano
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Tsutomu Sasaki
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yasufumi Gon
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Takaya Kitano
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Hideaki Kanki
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Kenichi Todo
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Munehisa Shimamura
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.,Department of Health Development and Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yasushi Matsumura
- Department of Medical Informatics Science, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Ao Huang
- Department of Biomedical Statistics, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Satoshi Hattori
- Department of Biomedical Statistics, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.,Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
254
|
Mauracher LM, Krall M, Roiß J, Hell L, Koder S, Hofbauer TM, Gebhart J, Hayden H, Brostjan C, Ay C, Pabinger I. Neutrophil subpopulations and their activation potential in patients with antiphospholipid syndrome and healthy individuals. Rheumatology (Oxford) 2021; 60:1687-1699. [PMID: 33026085 PMCID: PMC8024003 DOI: 10.1093/rheumatology/keaa532] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/27/2020] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES Patients with APS are at increased risk of thromboembolism. Neutrophils have been shown to play a role in inducing thrombosis. We aimed to investigate differences in neutrophil subpopulations, their potential of activation and neutrophil extracellular trap (NET) formation comparing high and low-density neutrophils (HDNs/LDNs) as well as subpopulations in patients with APS and controls to gain deeper insight into their potential role in thrombotic manifestations in patients with APS. METHODS HDNs and LDNs of 20 patients with APS and 20 healthy donors were isolated by density gradient centrifugation and stimulated. Neutrophil subpopulations, their activation and NET release were assessed by flow cytometry. RESULTS LDNs of both groups showed higher baseline activation, lower response to stimulation (regulation of activation markers CD11b/CD66b), but higher NET formation compared with HDNs. In patients with APS, the absolute number of LDNs was higher compared with controls. HDNs of APS patients showed higher spontaneous activation [%CD11b high: median (interquartile range): 2.78% (0.58-10.24) vs 0.56% (0.19-1.37)] and response to stimulation with ionomycin compared with HDNs of healthy donors [%CD11b high: 98.20 (61.08-99.13) vs 35.50% (13.50-93.85)], whereas no difference was found in LDNs. NET formation was increased in patients' HDNs upon stimulation. CONCLUSION HDNs and LDNs act differently, unstimulated and upon various stimulations in both healthy controls and APS patients. Differences in HDNs and LDNs between patients with APS and healthy controls indicate that neutrophils may enhance the risk of thrombosis in these patients and could thus be a target for prevention of thrombosis in APS.
Collapse
Affiliation(s)
- Lisa-Marie Mauracher
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Vienna, Austria
| | - Moritz Krall
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Vienna, Austria
| | - Johanna Roiß
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Vienna, Austria
| | - Lena Hell
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Vienna, Austria
| | - Silvia Koder
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Vienna, Austria
| | - Thomas M Hofbauer
- Division of Cardiology, Department of Internal Medicine II, Vienna, Austria
| | - Johanna Gebhart
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Vienna, Austria
| | - Hubert Hayden
- Department of Surgery, Surgical Research Laboratories, Medical University of Vienna, Vienna, Austria
| | - Christine Brostjan
- Department of Surgery, Surgical Research Laboratories, Medical University of Vienna, Vienna, Austria
| | - Cihan Ay
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Vienna, Austria
| | - Ingrid Pabinger
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Vienna, Austria
| |
Collapse
|
255
|
Kanji R, Kubica J, Navarese EP, Gorog DA. Endogenous fibrinolysis-Relevance to clinical thrombosis risk assessment. Eur J Clin Invest 2021; 51:e13471. [PMID: 33296082 DOI: 10.1111/eci.13471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/05/2020] [Accepted: 12/06/2020] [Indexed: 12/11/2022]
Abstract
The development of an obstructive luminal thrombus is pathological and considered a failure of endogenous fibrinolysis. The consequences may be fatal, or result in lasting downstream organ damage. Therefore, assessment of endogenous fibrinolytic status in an individual may identify those at risk of occlusive thrombus formation and provide prognostic information. Arterial thrombi are more platelet rich and more resistant to fibrinolysis than venous thrombi. Several recent studies using global tests of fibrinolysis in patients with acute coronary syndromes (ACS) have shown that despite dual antiplatelet therapy, patients with impaired fibrinolytic status have an increased risk of adverse cardiovascular events, compared with those with effective fibrinolytic function. Such data add significantly to the predictive value of established cardiovascular risk factors and conventional biomarkers. Most data reported have been obtained with the Global Thrombosis Test and the turbidimetric plasma clot lysis assay. A few small studies in patients with ischaemic stroke suggest a similar predictive role of fibrinolytic status assessment in these patients. Studies reporting an association between impaired fibrinolysis and future venous thrombotic events are limited, and in the form of case-control studies. Viscoelastic assays may have a role in the prediction of venous thromboembolic risk. Assays of fibrinolytic function should be used to obtain a more accurate risk of future thrombotic events, particularly in the setting of ACS. The availability of point-of-care tests helps facilitate this and should encourage future studies to assess personalised antithrombotic treatment combinations to optimise fibrinolytic status and reduce thrombosis risk.
Collapse
Affiliation(s)
- Rahim Kanji
- Faculty of Medicine, National Heart and Lung Institute, Imperial College, London, UK.,Cardiology Department, East and North Hertfordshire NHS Trust, Stevenage, UK
| | - Jacek Kubica
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Eliano P Navarese
- Department of Cardiology and Internal Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland.,Faculty of Medicine, University of Alberta, Edmonton, Canada
| | - Diana A Gorog
- Faculty of Medicine, National Heart and Lung Institute, Imperial College, London, UK.,Cardiology Department, East and North Hertfordshire NHS Trust, Stevenage, UK.,School of Life and Medical Sciences, Postgraduate Medical School, University of Hertfordshire, Stevenage, UK
| |
Collapse
|
256
|
Indirli R, Ferrante E, Scalambrino E, Profka E, Clerici M, Lettera T, Serban AL, Vena W, Pizzocaro A, Bonomi M, Cangiano B, Carosi G, Mazziotti G, Persani L, Lania A, Arosio M, Peyvandi F, Mantovani G, Tripodi A. Procoagulant Imbalance in Klinefelter Syndrome Assessed by Thrombin Generation Assay and Whole-Blood Thromboelastometry. J Clin Endocrinol Metab 2021; 106:e1660-e1672. [PMID: 33382882 PMCID: PMC7993570 DOI: 10.1210/clinem/dgaa936] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 11/19/2022]
Abstract
CONTEXT Klinefelter syndrome (KS) is a condition at increased risk of thrombosis compared to 46,XY men. OBJECTIVE This work aimed to investigate the coagulation balance of KS patients by thrombin generation assay (TGA) and thromboelastometry. METHODS An observational, cross-sectional study was conducted at 3 tertiary endocrinological centers in Milan, Italy. Fifty-eight KS patients and 58 age-matched healthy controls were included. Anticoagulant or antiplatelet therapy and known coagulation disorders were exclusion criteria. TGA was performed in platelet-poor plasma (PPP) and platelet-rich plasma (PRP). Whole-blood thromboelastometry and activities of coagulation factors were assessed. Endogenous thrombin potential (ETP), the area under the thrombin generation curve, assessed with and without thrombomodulin (ETP-TM+ and ETP-TM-), and their ratio (ETP ratio), were considered as indexes of procoagulant imbalance. RESULTS Patients with KS displayed higher PPP-ETP-TM+ (mean 1528 vs 0.1315 nM × min; P < .001), PPP-ETP ratio (0.78 vs 0.0.70; P < .001), factor (F)VIII (135% vs 0.107%; P = .001), fibrinogen (283 vs 0.241 mg/dL; P < .001), and FVIII/protein C ratio (1.21 vs 0.1.06; P < .05) compared to controls. Protein C was comparable in the 2 groups. Similar results were observed in PRP. The ETP ratio was positively associated with FVIII (ρ = 0.538, P < .001) in KS. Thromboelastometry parameters confirmed evidence of hypercoagulability in KS. CONCLUSION Patients with KS display a procoagulant imbalance expressed by increased thrombin generation both in PPP and PRP, which is at least in part explained by increased FVIII levels. The procoagulant imbalance, which was confirmed by thromboelastometry, may be responsible for the thrombotic events observed in these patients. Further investigation on the benefit/risk ratio of antithrombotic prophylaxis is warranted.
Collapse
Affiliation(s)
- Rita Indirli
- Endocrinology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Emanuele Ferrante
- Endocrinology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Erica Scalambrino
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - Eriselda Profka
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Marigrazia Clerici
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - Tommaso Lettera
- Laboratorio analisi, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andreea Liliana Serban
- Endocrinology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Walter Vena
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Endocrinology, Diabetology and Andrology Unit, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Alessandro Pizzocaro
- Endocrinology, Diabetology and Andrology Unit, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
| | - Marco Bonomi
- IRCCS Istituto Auxologico Italiano, Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Diseases, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Biagio Cangiano
- IRCCS Istituto Auxologico Italiano, Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Diseases, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Giulia Carosi
- Endocrinology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Gherardo Mazziotti
- Endocrinology, Diabetology and Andrology Unit, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Milan, Italy
| | - Luca Persani
- IRCCS Istituto Auxologico Italiano, Department of Endocrine and Metabolic Diseases and Lab of Endocrine and Metabolic Diseases, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Andrea Lania
- Endocrinology, Diabetology and Andrology Unit, Humanitas Clinical and Research Center IRCCS, Rozzano, Milan, Italy
- Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, Milan, Italy
| | - Maura Arosio
- Endocrinology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Flora Peyvandi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| | - Giovanna Mantovani
- Endocrinology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Correspondence: Giovanna Mantovani, MD, PhD, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Department: Endocrinology Unit, Via Francesco Sforza, 35, Milan, 20122, Italy.
| | - Armando Tripodi
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore, Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Milan, Italy
| |
Collapse
|
257
|
Nielsen T, Kristensen SR, Gregersen H, Teodorescu EM, Pedersen S. Prothrombotic abnormalities in patients with multiple myeloma and monoclonal gammopathy of undetermined significance. Thromb Res 2021; 202:108-118. [PMID: 33819778 DOI: 10.1016/j.thromres.2021.03.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Multiple myeloma (MM) and its precursor condition, monoclonal gammopathy of undetermined significance (MGUS) have an increased risk of thrombotic events, especially during anti-myeloma treatment. Many different underlying causes for this hypercoagulability have been suggested, but current techniques to identify abnormalities in these patients are sparse and inefficient. The aim of this study was to assess the hypercoagulability in MGUS and MM patients through various coagulation analyses and identify changes in the MM patients throughout their treatment regimen. MATERIALS AND METHODS Platelet-free plasma from 38 MM patients, 19 MGUS patients and 34 healthy controls were tested for hypercoagulability using calibrated automated thrombogram, a procoagulant phospholipid assay, a microvesicle-associated (MV) tissue factor (TF) assay, and a cell-free deoxyribonucleic acid (cf-DNA) assay as a surrogate measurement for neutrophil extracellular traps (NETs). RESULTS MGUS and MM patients both had elevated thrombin generation and procoagulant phospholipid activity in comparison to the control subjects. MM, and partly MGUS, showed increased MV-TF activity, however, only MM had increased levels of the cf-DNA. CONCLUSIONS Here we demonstrated that hypercoagulability was present in patients with MGUS and MM through increased thrombin generation, possibly due to higher TF and procoagulant phospholipids (PPL) activity. This may be associated to MVs and, for MM patients, be attributed to procoagulant NETs activity; however, this remains to be determined.
Collapse
Affiliation(s)
- Thøger Nielsen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| | - Søren Risom Kristensen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| | | | | | - Shona Pedersen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
258
|
Ren J, He J, Zhang H, Xia Y, Hu Z, Loughran P, Billiar T, Huang H, Tsung A. Platelet TLR4-ERK5 Axis Facilitates NET-Mediated Capturing of Circulating Tumor Cells and Distant Metastasis after Surgical Stress. Cancer Res 2021; 81:2373-2385. [PMID: 33687949 DOI: 10.1158/0008-5472.can-20-3222] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/21/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022]
Abstract
Surgical removal of malignant tumors is a mainstay in controlling most solid cancers. However, surgical insult also increases the risk of tumor recurrence and metastasis. Tissue trauma activates the innate immune system locally and systemically, mounting an inflammatory response. Platelets and neutrophils are two crucial players in the early innate immune response that heals tissues, but their actions may also contribute to cancer cell dissemination and distant metastasis. Here we report that surgical stress-activated platelets enhance the formation of platelet-tumor cell aggregates, facilitating their entrapment by neutrophil extracellular traps (NET) and subsequent distant metastasis. A murine hepatic ischemia/reperfusion (I/R) injury model of localized surgical stress showed that I/R promotes capturing of aggregated circulating tumor cells (CTC) by NETs and eventual metastasis to the lungs, which are abrogated when platelets are depleted. Hepatic I/R also increased deposition of NETs within the lung microvasculature, but depletion of platelets had no effect. TLR4 was essential for platelet activation and platelet-tumor cell aggregate formation in an ERK5-GPIIb/IIIa integrin-dependent manner. Such aggregation facilitated NET-mediated capture of CTCs in vitro under static and dynamic conditions. Blocking platelet activation or knocking out TLR4 protected mice from hepatic I/R-induced metastasis with no CTC entrapment by NETs. These results uncover a novel mechanism where platelets and neutrophils contribute to metastasis in the setting of acute inflammation. Targeted disruption of the interaction between platelets and NETs holds therapeutic promise to prevent postoperative distant metastasis. SIGNIFICANCE: Targeting platelet activation via TLR4/ERK5/integrin GPIIb/IIIa signaling shows potential for preventing NET-driven distant metastasis in patients post-resection.
Collapse
Affiliation(s)
- Jinghua Ren
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio.,Cancer center, Union Hospital, Huazhong University of Science and Technology, Wuhan, P.R. China.,Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jiayi He
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio.,Department of Pediatrics, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hongji Zhang
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio.,Department of Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yujia Xia
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio.,Department of Gastroenterology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zhiwei Hu
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Center for Biologic Imaging, Department of Cell Biology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Timothy Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Hai Huang
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio.
| | - Allan Tsung
- Department of Surgery, The Ohio State University Medical Center, Columbus, Ohio.
| |
Collapse
|
259
|
Elastase inhibitor agaphelin protects from acute ischemic stroke in mice by reducing thrombosis, blood-brain barrier damage, and inflammation. Brain Behav Immun 2021; 93:288-298. [PMID: 33401017 PMCID: PMC7979502 DOI: 10.1016/j.bbi.2020.12.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/14/2020] [Accepted: 12/22/2020] [Indexed: 12/23/2022] Open
Abstract
Recently it was shown that the hematophagous salivary gland protein agaphelin exhibits multiple antithrombotic effects without promoting the risk of bleeding. Agaphelin inhibits neutrophil elastase and thereby reduces cathepsin G-induced platelet aggregation. However, it is still unclear, whether pharmacological treatment with agaphelin in brain ischemia is protective and, regarding its bleeding risk, safe. To elucidate this issue, male C57BL/6 mice were subjected to 60 min of transient middle cerebral artery occlusion (tMCAO) and treated with 0.25 mg/kg agaphelin intravenously immediately after tMCAO. On day 1 and 7, infarct volume and functional neurological outcome were assessed by behavioural tests, histochemistry and magnetic resonance imaging. Thrombus formation, intracerebral bleeding risk, blood-brain barrier damage and the local inflammatory response were determined on day 1. This study shows for the first time a protective effect of agaphelin characterized by smaller infarct volume, reduced neurological deficits and reduced animal mortality. This protective effect was associated with reduced local thrombus formation, increased blood-brain barrier integrity and reduced brain inflammatory response. It is essential to mention that the protective effect of agaphelin was not linked to an increased risk of intracerebral bleeding. The promotion of brain tissue survival and inhibition of thromboinflammation identifies agaphelin as a promising treatment option in ischemic stroke, which considering the lack of bleeding risk should potentially be safe.
Collapse
|
260
|
Abstract
Thrombi retrieved from patients with acute ischemic stroke are highly heterogeneous. Recent data suggest that thrombus composition may impact on mechanical thrombectomy, the number of recanalization manoeuvres, resistance to retrieval, and on thrombolytic potential. Our aim was to summarize evidence describing the impact of thrombus composition on efficacy of mechanical thrombectomy and thrombolysis in patients with acute ischemic stroke. The scoping review methodology guided by the Joanna Briggs Institute, an adaption of the Arksey and O'Malley, was followed. Comprehensive searches were conducted in MEDLINE, EMBASE, SCOPUS, and Web of Science. Articles were classified into 4 key themes: (1) composition of stroke thrombi, (2) thrombus composition and mechanical thrombectomy, (3) thrombus composition and thrombolytic therapy, and (4) novel imaging and endovascular approaches. Our search identified 698 articles published from 1987 to June 2020. Additional articles were extracted from reference lists of the selected articles. Overall, 95 topic-specific articles identified for inclusion published in 40 different journals were included. Reports showed that thrombus composition in stroke was highly heterogeneous, containing fibrin, platelets, red blood cells, VWF (von Willebrand Factor), and neutrophil extracellular traps. Thrombi could roughly be divided into fibrin- and red blood cell-rich clots. Fibrin-rich clots were associated with increased recanalization manoeuvres, longer procedure time, and less favorable clinical outcomes compared with red blood cell-rich clots. Advances in detection or treatment of thrombi that take into account clot heterogeneity may be able to improve future endovascular and thrombolytic treatment of stroke.
Collapse
Affiliation(s)
- Precious Jolugbo
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom
| | - Robert A S Ariëns
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom
| |
Collapse
|
261
|
Dhami SPS, Patmore S, O'Sullivan JM. Advances in the Management of Cancer-Associated Thrombosis. Semin Thromb Hemost 2021; 47:139-149. [PMID: 33636745 DOI: 10.1055/s-0041-1722863] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The association between cancer and venous thromboembolism (VTE) has been established for more than 150 years. Nevertheless, cancer-associated thrombosis still remains a major clinical challenge and is associated with significant morbidity and mortality for patients with cancer. The clinical presentation of cancer-associated thrombosis can be distinct from that of a patient without an underlying malignancy. Moreover, specific cancer types, including pancreatic cancer and hematological malignancies, as well as advanced stage disease can confer a significant thrombotic risk. This risk is further augmented by specific anticancer treatment modalities. The pathophysiology of cancer-associated thrombosis is complex and multifactorial. However, understanding the biological mechanisms underpinning VTE risk may provide insight into novel targeted prophylaxis in cancer patients. Over the last decade, low-molecular-weight heparin has been the preferred anticoagulant agent for patients with cancer-associated thrombosis due to improved efficacy compared with vitamin K antagonists. However, the advent of direct oral anticoagulants (DOACs) has added to the repertoire of ammunition now at the disposal of clinicians to aid in the management of cancer-associated thrombosis. Several randomized controlled trials have now been published, demonstrating DOAC as a noninferior alternative for both the treatment and prevention of cancer-associated thrombosis. Notwithstanding this, limitations for their widespread use remain, with the potential for increased bleeding risk, drug interactions, and poor DOAC metabolism. This review discusses the evidence base for the incidence and risk factors associated with VTE in cancer, development, and refinement of risk prediction models and novel advances in the therapeutic management of cancer-associated thrombosis.
Collapse
Affiliation(s)
- Sukhraj Pal Singh Dhami
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Sean Patmore
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Jamie M O'Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
262
|
Mata R, Yao Y, Cao W, Ding J, Zhou T, Zhai Z, Gao C. The Dynamic Inflammatory Tissue Microenvironment: Signality and Disease Therapy by Biomaterials. RESEARCH 2021; 2021:4189516. [PMID: 33623917 PMCID: PMC7879376 DOI: 10.34133/2021/4189516] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022]
Abstract
Tissue regeneration is an active multiplex process involving the dynamic inflammatory microenvironment. Under a normal physiological framework, inflammation is necessary for the systematic immunity including tissue repair and regeneration as well as returning to homeostasis. Inflammatory cellular response and metabolic mechanisms play key roles in the well-orchestrated tissue regeneration. If this response is dysregulated, it becomes chronic, which in turn causes progressive fibrosis, improper repair, and autoimmune disorders, ultimately leading to organ failure and death. Therefore, understanding of the complex inflammatory multiple player responses and their cellular metabolisms facilitates the latest insights and brings novel therapeutic methods for early diseases and modern health challenges. This review discusses the recent advances in molecular interactions of immune cells, controlled shift of pro- to anti-inflammation, reparative inflammatory metabolisms in tissue regeneration, controlling of an unfavorable microenvironment, dysregulated inflammatory diseases, and emerging therapeutic strategies including the use of biomaterials, which expand therapeutic views and briefly denote important gaps that are still prevailing.
Collapse
Affiliation(s)
- Rani Mata
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yuejun Yao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Wangbei Cao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jie Ding
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Tong Zhou
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zihe Zhai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.,Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
263
|
Pizzolo F, Castagna A, Olivieri O, Girelli D, Friso S, Stefanoni F, Udali S, Munerotto V, Baroni M, Cetera V, Luciani GB, Faggian G, Bernardi F, Martinelli N. Basophil Blood Cell Count Is Associated With Enhanced Factor II Plasma Coagulant Activity and Increased Risk of Mortality in Patients With Stable Coronary Artery Disease: Not Only Neutrophils as Prognostic Marker in Ischemic Heart Disease. J Am Heart Assoc 2021; 10:e018243. [PMID: 33624506 PMCID: PMC8174269 DOI: 10.1161/jaha.120.018243] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background White blood cell count, which is inexpensive and widely available in clinical practice, has been proposed to provide prognostic information in coronary artery disease (CAD). Elevated levels of white blood cell subtypes may play different roles in atherothrombosis and predict cardiovascular outcomes. Methods and Results The association between white blood cell counts and mortality was evaluated in 823 subjects with angiographically demonstrated and clinically stable CAD in an observational-longitudinal study. The correlation among white blood cell counts and factor II plasma coagulant activity was analyzed in 750 subjects (554 CAD and 196 CAD-free) not taking anticoagulant drugs. Subjects with overt leukocytosis or leukopenia were excluded. In the longitudinal study after a median follow-up of 61 months, 160 (19.4%) subjects died, 107 (13.0%) of whom from cardiovascular causes. High levels of neutrophils, monocytes, eosinophils, and basophils were associated with an increased mortality rate. In multiadjusted Cox regression models, only neutrophils and basophils remained predictors of total and cardiovascular mortality. The associations remained significant after adjustment for traditional cardiovascular risk factors and by including D-dimer and the chemokine CXCL12 in the regression models. Neutrophils and basophils were also significant predictors of factor II plasma coagulant activity variability after adjustment for blood cell counts, age, sex, inflammatory markers, CAD diagnosis, and prothrombin G20210A polymorphism. Factor II plasma coagulant activity was similarly increased in subjects with high neutrophil and basophil counts and in carriers of the prothrombin 20210A allele. Conclusions Both high neutrophil and basophil blood counts may predict mortality in patients with clinically stable CAD and are associated with enhanced factor II plasma coagulant activity, thereby suggesting underlying prothrombotic mechanisms.
Collapse
Affiliation(s)
- Francesca Pizzolo
- Department of Medicine Unit of Internal Medicine University of Verona Verona Italy
| | - Annalisa Castagna
- Department of Medicine Unit of Internal Medicine University of Verona Verona Italy
| | - Oliviero Olivieri
- Department of Medicine Unit of Internal Medicine University of Verona Verona Italy
| | - Domenico Girelli
- Department of Medicine Unit of Internal Medicine University of Verona Verona Italy
| | - Simonetta Friso
- Department of Medicine Unit of Internal Medicine University of Verona Verona Italy
| | - Filippo Stefanoni
- Department of Medicine Unit of Internal Medicine University of Verona Verona Italy
| | - Silvia Udali
- Department of Medicine Unit of Internal Medicine University of Verona Verona Italy
| | - Veronica Munerotto
- Department of Medicine Unit of Internal Medicine University of Verona Verona Italy
| | - Marcello Baroni
- Department of Life Sciences and Biotechnology University of Ferrara Ferrara Italy
| | - Vera Cetera
- Division of Cardiac Surgery Department of Surgery, Dentistry, Pediatrics and Gynecology University of Verona Verona Italy
| | - Giovanni Battista Luciani
- Division of Cardiac Surgery Department of Surgery, Dentistry, Pediatrics and Gynecology University of Verona Verona Italy
| | - Giuseppe Faggian
- Division of Cardiac Surgery Department of Surgery, Dentistry, Pediatrics and Gynecology University of Verona Verona Italy
| | - Francesco Bernardi
- Department of Life Sciences and Biotechnology University of Ferrara Ferrara Italy
| | - Nicola Martinelli
- Department of Medicine Unit of Internal Medicine University of Verona Verona Italy
| |
Collapse
|
264
|
Águila S, de los Reyes-García AM, Fernández-Pérez MP, Reguilón-Gallego L, Zapata-Martínez L, Ruiz-Lorente I, Vicente V, González-Conejero R, Martínez C. MicroRNAs as New Regulators of Neutrophil Extracellular Trap Formation. Int J Mol Sci 2021; 22:ijms22042116. [PMID: 33672737 PMCID: PMC7924615 DOI: 10.3390/ijms22042116] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/27/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are formed after neutrophils expelled their chromatin content in order to primarily capture and eliminate pathogens. However, given their characteristics due in part to DNA and different granular proteins, NETs may induce a procoagulant response linking inflammation and thrombosis. Unraveling NET formation molecular mechanisms as well as the intracellular elements that regulate them is relevant not only for basic knowledge but also to design diagnostic and therapeutic tools that may prevent their deleterious effects observed in several inflammatory pathologies (e.g., cardiovascular and autoimmune diseases, cancer). Among the potential elements involved in NET formation, several studies have investigated the role of microRNAs (miRNAs) as important regulators of this process. miRNAs are small non-coding RNAs that have been involved in the control of almost all physiological processes in animals and plants and that are associated with the development of several pathologies. In this review, we give an overview of the actual knowledge on NETs and their implication in pathology with a special focus in cardiovascular diseases. We also give a brief overview on miRNA biology to later focus on the different miRNAs implicated in NET formation and the perspectives opened by the presented data.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rocío González-Conejero
- Correspondence: (R.G.-C.); (C.M.); Tel.: +34-968341990 (R.G.-C. & C.M.); Fax: +34-968261914 (R.G.-C. & C.M.)
| | - Constantino Martínez
- Correspondence: (R.G.-C.); (C.M.); Tel.: +34-968341990 (R.G.-C. & C.M.); Fax: +34-968261914 (R.G.-C. & C.M.)
| |
Collapse
|
265
|
Stevens H, McFadyen J, Chan N. Advances in the Management of Acute Venous Thromboembolism and New Therapeutic Agents. Semin Respir Crit Care Med 2021; 42:218-232. [PMID: 33601429 DOI: 10.1055/s-0041-1723953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Important advances in the understanding and management of venous thromboembolism (VTE) have enhanced our ability to diagnose, prevent, and treat VTE. In this narrative review, we discuss how recent advances in the understanding and management of VTE are changing practice, highlight ongoing unmet needs in VTE management, and outline how novel therapeutic targets with little or no influence on hemostasis may help address these unmet needs.
Collapse
Affiliation(s)
- Hannah Stevens
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia.,Department of Clinical Haematology, Alfred Hospital, Melbourne, Victoria, Australia
| | - James McFadyen
- Atherothrombosis and Vascular Biology Program, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia.,Department of Clinical Haematology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Noel Chan
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
266
|
Vorobjeva NV. Neutrophil Extracellular Traps: New Aspects. ACTA ACUST UNITED AC 2021; 75:173-188. [PMID: 33583971 PMCID: PMC7869772 DOI: 10.3103/s0096392520040112] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/28/2020] [Accepted: 10/03/2020] [Indexed: 12/20/2022]
Abstract
Neutrophils are the "first line" of defense against pathogens in the locus of inflammation, where they use effector functions such as phagocytosis, degranulation, and formation of reactive oxygen species (ROS). In 2004, Artuto Zychlinsky characterized one more neutrophil effector function-the release of neutrophil extracellular traps (or NETs). NETs are a modified chromatin "decorated" by bactericidal proteins of granules, nucleus, and cytoplasm. The release of NETs can be activated by diverse physiological and pharmacological stimuli and depends on ROS, for which NADPH oxidase is the main source. In the process of NET formation, the release of bactericidal components of granules into the cytoplasm, modification of histones leading to chromatin decondensation, destruction of the nuclear envelope and cytoplasmic membrane with the involvement of gasdermin D protein, and, finally, the release of chromatin outside the cell occurs. At the same time, uncontrolled formation of NETs is a provoking factor in the development of many inflammatory and autoimmune diseases. NETs were found at autoimmune diseases such as systemic lupus erythematosus, rheumatoid arthritis, psoriasis, and vasculitis; NETs are involved in the pathogenesis of cardiovascular, pulmonary, and oncological diseases. In this review, the main ideas about the mechanisms of NET formation, as well as their role in physiological processes and pathogenesis of a number of diseases (including COVID-19), are discussed.
Collapse
Affiliation(s)
- N V Vorobjeva
- Department of Biology, Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
267
|
Tremblay D, Kosiorek HE, Dueck AC, Hoffman R. Evaluation of Therapeutic Strategies to Reduce the Number of Thrombotic Events in Patients With Polycythemia Vera and Essential Thrombocythemia. Front Oncol 2021; 10:636675. [PMID: 33665170 PMCID: PMC7921696 DOI: 10.3389/fonc.2020.636675] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Thrombosis is the largest contributor to morbidity and mortality in patients with polycythemia vera (PV) and essential thrombocythemia (ET). Our understanding of the risk factors and pathophysiology of thrombosis in PV and ET patients is developing, including recent insights into the role of aberrant platelet-neutrophil interactions, JAK2 mutated endothelial cells and the pro-thrombotic inflammatory milieu. To date, few available therapies have demonstrated the ability to reduce the thrombotic burden in patients with these diseases. Although numerous therapeutic agents have been investigated in both PV and ET patients, few studies are designed to assess their impact on thrombotic events. In this review, we first describe the burden of thrombosis in patients with these myeloproliferative neoplasms (MPNs) and briefly explore their pathophysiologic mechanisms. We then critically assess and summarize the evidence behind currently available therapies with attention toward thrombotic endpoints. Finally, we describe a path forward for clinical research in MPNs that involves surrogate endpoint validation, biomarker development, and clinical trial design strategies in order to accurately assess reduction of thrombotic events when evaluating novel therapies.
Collapse
Affiliation(s)
- Douglas Tremblay
- Hematology/Oncology Section, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Heidi E. Kosiorek
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ, United States
| | - Amylou C. Dueck
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ, United States
| | - Ronald Hoffman
- Hematology/Oncology Section, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
268
|
Ali RA, Gandhi AA, Dai L, Weiner J, Estes SK, Yalavarthi S, Gockman K, Sun D, Knight JS. Antineutrophil properties of natural gingerols in models of lupus. JCI Insight 2021; 6:138385. [PMID: 33373329 PMCID: PMC7934838 DOI: 10.1172/jci.insight.138385] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 12/16/2020] [Indexed: 12/23/2022] Open
Abstract
Ginger is known to have antiinflammatory and antioxidative effects and has traditionally been used as an herbal supplement in the treatment of various chronic diseases. Here, we report antineutrophil properties of 6-gingerol, the most abundant bioactive compound of ginger root, in models of lupus and antiphospholipid syndrome (APS). Specifically, we demonstrate that 6-gingerol attenuates neutrophil extracellular trap (NET) release in response to lupus- and APS-relevant stimuli through a mechanism that is at least partially dependent on inhibition of phosphodiesterases. At the same time, administration of 6-gingerol to mice reduces NET release in various models of lupus and APS, while also improving other disease-relevant endpoints, such as autoantibody formation and large-vein thrombosis. In summary, this study is the first to our knowledge to demonstrate a protective role for ginger-derived compounds in the context of lupus. Importantly, it provides a potential mechanism for these effects via phosphodiesterase inhibition and attenuation of neutrophil hyperactivity.
Collapse
Affiliation(s)
- Ramadan A Ali
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alex A Gandhi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Lipeng Dai
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| | - Julia Weiner
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Shanea K Estes
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Srilakshmi Yalavarthi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kelsey Gockman
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason S Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
269
|
Sui J, Lu R, Halkidis K, Kocher NK, Cao W, Marques MB, Zheng XL. Plasma levels of S100A8/A9, histone/DNA complexes, and cell-free DNA predict adverse outcomes of immune thrombotic thrombocytopenic purpura. J Thromb Haemost 2021; 19:370-379. [PMID: 33188723 PMCID: PMC8058879 DOI: 10.1111/jth.15176] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/28/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Immune thrombotic thrombocytopenic purpura (iTTP) is a life-threatening blood disorder, primarily resulting from autoantibodies against ADAMTS13. Infection or inflammation often precedes acute iTTP. However, the association of inflammation and inflammatory mediators with disease severity and outcome of acute iTTP is not fully assessed. OBJECTIVES Here, we determined plasma levels of S100A8/A9, histone/DNA complexes, citrullinated histone H3 (CitH3), and cell-free DNA (cfDNA) in a cohort of 108 acute episodes from 94 unique iTTP patients and healthy controls, and assessed the association of each of these biomarkers with the disease severity and mortality. RESULTS All acute iTTP patients had significantly increased plasma levels of S100A8/A9 (median 84.8, interquartile range [IQR] 31.2-157.4 µg/mL), histone/DNA complexes (median 55.7, IQR 35.8-130.8 U/mL), CitH3 (median 3.8, IQR 2.2-6.4 ng/mL), and cfDNA (median 937.7, IQR 781.3-1420.0 ng/mL) on the admission blood samples when compared with healthy controls. An increased plasma level of S100A8/A9, histone/DNA complex and cfDNA was associated with organ damage, coagulopathy, and mortality in iTTP. After being adjusted for age and history of hypertension, Cox proportional hazard regression analysis demonstrated that a hazard ratio (95% confidence interval) for an elevated plasma level of S100A8/A9, histone/DNA complexes, and cfDNA was 11.5 (1.4-90.9) (P = .021), 10.3 (2.7-38.5) (P = .001), and 12.8 (3.9-42.0) (P = .014), respectively. CONCLUSION These results indicate that inflammation or plasma inflammatory mediators such as S100A8/A9 or NETosis markers such as histone/DNA complexes and cfDNA may play a role in pathogenesis of iTTP, which may help stratify patients with a high risk of death during acute iTTP episodes.
Collapse
Affiliation(s)
- Jingrui Sui
- Division of Laboratory Medicine, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ruinan Lu
- Division of Laboratory Medicine, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Konstantine Halkidis
- Division of Laboratory Medicine, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
- Division of Hematology and Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Nicole K. Kocher
- Division of Laboratory Medicine, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wenjing Cao
- Division of Laboratory Medicine, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Marisa B. Marques
- Division of Laboratory Medicine, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - X. Long Zheng
- Division of Laboratory Medicine, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
270
|
Defects in ubiquitination and NETosis and their associations with human diseases. Pathology 2021; 53:439-445. [PMID: 33518384 DOI: 10.1016/j.pathol.2020.10.014] [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: 01/29/2019] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 11/24/2022]
Abstract
Various autoimmune diseases are associated with defects in protein degradation and NETosis. This review aims to examine defects in ubiquitination and NETosis and their associations with human disease. This study involved a systematic search of electronic databases, including PubMed, EBSCO, and LILACS, to locate articles on the relationship between human disease and defects in protein degradation and NETosis. Ubiquitination and NETosis can trigger a cascade of events that affect immune system function and impact the body's ability to fight disease. Ubiquitination is implicated in various disorders, such as Liddle's syndrome, Alzheimer's disease, and other neurodegenerative disorders, whereas NETosis has been linked to antineutrophil cytoplasmic antibody associated vasculitis, accelerated atherosclerosis, thrombosis, rheumatoid arthritis, antiphospholipid antibody syndrome, type 1 diabetes mellitus, and renal inflammatory complications. Researchers have attempted for years to identify the link between neurodegenerative disease and ubiquitination. Previous studies analysed the relationships between different autoimmune disorders and NETosis and identified various ubiquitin conjugates and NET remnants that trigger disease development and progression. Ubiquitination and NETosis play key roles in the emergence and progression of neurodegenerative and autoimmune disorders. Further investigation is needed to elucidate the mechanisms underlying the relationships between these disorders and biological processes.
Collapse
|
271
|
Emerging Role of Neutrophils in the Thrombosis of Chronic Myeloproliferative Neoplasms. Int J Mol Sci 2021; 22:ijms22031143. [PMID: 33498945 PMCID: PMC7866001 DOI: 10.3390/ijms22031143] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/10/2021] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
Thrombosis is a major cause of morbimortality in patients with chronic Philadelphia chromosome-negative myeloproliferative neoplasms (MPN). In the last decade, multiple lines of evidence support the role of leukocytes in thrombosis of MPN patients. Besides the increase in the number of cells, neutrophils and monocytes of MPN patients show a pro-coagulant activated phenotype. Once activated, neutrophils release structures composed of DNA, histones, and granular proteins, called extracellular neutrophil traps (NETs), which in addition to killing pathogens, provide an ideal matrix for platelet activation and coagulation mechanisms. Herein, we review the published literature related to the involvement of NETs in the pathogenesis of thrombosis in the setting of MPN; the effect that cytoreductive therapies and JAK inhibitors can have on markers of NETosis, and, finally, the novel therapeutic strategies targeting NETs to reduce the thrombotic complications in these patients.
Collapse
|
272
|
Shi Y, Gauer JS, Baker SR, Philippou H, Connell SD, Ariëns RAS. Neutrophils can promote clotting via FXI and impact clot structure via neutrophil extracellular traps in a distinctive manner in vitro. Sci Rep 2021; 11:1718. [PMID: 33462294 PMCID: PMC7814028 DOI: 10.1038/s41598-021-81268-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/04/2021] [Indexed: 12/23/2022] Open
Abstract
Neutrophils and neutrophil extracellular traps (NETs) have been shown to be involved in coagulation. However, the interactions between neutrophils or NETs and fibrin(ogen) in clots, and the mechanisms behind these interactions are not yet fully understood. In this in vitro study, the role of neutrophils or NETs on clot structure, formation and dissolution was studied with a combination of confocal microscopy, turbidity and permeation experiments. Factor (F)XII, FXI and FVII-deficient plasmas were used to investigate which factors may be involved in the procoagulant effects. We found both neutrophils and NETs promote clotting in plasma without the addition of other coagulation triggers, but not in purified fibrinogen, indicating that other factors mediate the interaction. The procoagulant effects of neutrophils and NETs were also observed in FXII- and FVII-deficient plasma. In FXI-deficient plasma, only the procoagulant effects of NETs were observed, but not of neutrophils. NETs increased the density of clots, particularly in the vicinity of the NETs, while neutrophils-induced clots were less stable and more porous. In conclusion, NETs accelerate clotting and contribute to the formation of a denser, more lysis resistant clot architecture. Neutrophils, or their released mediators, may induce clotting in a different manner to NETs, mediated by FXI.
Collapse
Affiliation(s)
- Y Shi
- LIGHT Laboratories, Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds, LS2 9LU, UK
| | - J S Gauer
- LIGHT Laboratories, Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds, LS2 9LU, UK
| | - S R Baker
- LIGHT Laboratories, Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds, LS2 9LU, UK
- Department of Physics, Wake Forest University, Winston Salem, NC, USA
| | - H Philippou
- LIGHT Laboratories, Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds, LS2 9LU, UK
| | - S D Connell
- The Astbury Centre for Structural Molecular Biology, Molecular & Nanoscale Physics, University of Leeds, Leeds, LS2 9JT, UK
| | - R A S Ariëns
- LIGHT Laboratories, Discovery and Translational Science Department, Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Clarendon Way, Leeds, LS2 9LU, UK.
| |
Collapse
|
273
|
Zhang S, Zhang J, Wang C, Chen X, Zhao X, Jing H, Liu H, Li Z, Wang L, Shi J. COVID‑19 and ischemic stroke: Mechanisms of hypercoagulability (Review). Int J Mol Med 2021; 47:21. [PMID: 33448315 PMCID: PMC7849983 DOI: 10.3892/ijmm.2021.4854] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022] Open
Abstract
During the coronavirus disease 2019 (COVID-19) pandemic, some patients with severe COVID-19 exhibited complications such as acute ischemic stroke (AIS), which was closely associated with a poor prognosis. These patients often had an abnormal coagulation, namely, elevated levels of D-dimer and fibrinogen, and a low platelet count. Certain studies have suggested that COVID-19 induces AIS by promoting hypercoagulability. Nevertheless, the exact mechanisms through which COVID-19 leads to a hypercoagulable state in infected patients remain unclear. Understanding the underlying mechanisms of hypercoagulability is of utmost importance for the effective treatment of these patients. The present review aims to summarize the current status of research on COVID-19, hypercoagulability and ischemic stroke. The present review also aimed to shed light into the underlying mechanisms through which COVID-19 induces hypercoagulability, and to provide therapies for different mechanisms for the more effective treatment of patients with COVID-19 with ischemic stroke and prevent AIS during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Shuoqi Zhang
- Department of Neurology, The Second Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jinming Zhang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Chunxu Wang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiaojing Chen
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xinyi Zhao
- Department of Cardiology, The Second Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Haijiao Jing
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Huan Liu
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Zhuxin Li
- Department of Acupuncture and Moxibustion, College of Acupuncture and Moxibustion, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Lihua Wang
- Department of Neurology, The Second Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jialan Shi
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
274
|
Janiuk K, Jabłońska E, Garley M. Significance of NETs Formation in COVID-19. Cells 2021; 10:cells10010151. [PMID: 33466589 PMCID: PMC7828704 DOI: 10.3390/cells10010151] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Severe contagious respiratory disease—COVID-19—caused by the SARS-CoV-2 coronavirus, can lead to fatal respiratory failure associated with an excessive inflammatory response. Infiltration and spread of SARS-CoV-2 are based on the interaction between the virus’ structural protein S and the cell’s receptor–angiotensin-converting enzyme 2 (ACE2), with the simultaneous involvement of human trans-membrane protease, serine 2 (TMPRSS2). Many scientific reports stress the importance of elevated recruitment and activity of neutrophils, which can form extracellular neutrophil traps (NETs) playing a significant role in the mechanism of combating pathogens, in the pathogenesis of COVID-19. Excessive generation of NETs during prolonged periods of inflammation predisposes for the occurrence of undesirable reactions including thromboembolic complications and damage to surrounding tissues and organs. Within the present manuscript, we draw attention to the impact of NET generation on the severe course of COVID-19 in patients with concurrent cardiovascular and metabolic diseases. Additionally, we indicate the necessity to explore not only the cellular but also the molecular bases of COVID-19 pathogenesis, which may aid the development of dedicated therapies meant to improve chances for the successful treatment of patients. We also present new directions of research into medications that display NETs formation regulatory properties as potential significant therapeutic strategies in the progress of COVID-19.
Collapse
|
275
|
Jooss NJ, Poulter NS. A large-scale histological investigation gives insight into the structure of ischemic stroke thrombi. Platelets 2021; 32:147-150. [PMID: 33427010 DOI: 10.1080/09537104.2020.1869713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Natalie J Jooss
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Natalie S Poulter
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK.,Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Midlands, UK
| |
Collapse
|
276
|
Donovan K, Shah A, Day J, McKechnie SR. Adjunctive treatments for the management of septic shock - a narrative review of the current evidence. Anaesthesia 2021; 76:1245-1258. [PMID: 33421029 DOI: 10.1111/anae.15369] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2020] [Indexed: 12/13/2022]
Abstract
Septic shock is a leading cause of death and morbidity worldwide. The cornerstones of management include prompt identification of sepsis, early initiation of antibiotic therapy, adequate fluid resuscitation and organ support. Over the past two decades, there have been considerable improvements in our understanding of the pathophysiology of sepsis and the host response, including regulation of inflammation, endothelial disruption and impaired immunity. This has offered opportunities for innovative adjunctive treatments such as vitamin C, corticosteroids and beta-blockers. Some of these approaches have shown promising results in early phase trials in humans, while others, such as corticosteroids, have been tested in large, international, multicentre randomised controlled trials. Contemporary guidelines make a weak recommendation for the use of corticosteroids to reduce mortality in sepsis and septic shock. Vitamin C, despite showing initial promise in observational studies, has so far not been shown to be clinically effective in randomised trials. Beta-blocker therapy may have beneficial cardiac and non-cardiac effects in septic shock, but there is currently insufficient evidence to recommend their use for this condition. The results of ongoing randomised trials are awaited. Crucial to reducing heterogeneity in the trials of new sepsis treatments will be the concept of enrichment, which refers to the purposive selection of patients with clinical and biological characteristics that are likely to be responsive to the intervention being tested.
Collapse
Affiliation(s)
- K Donovan
- Adult Intensive Care Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.,Adult Intensive Care Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - A Shah
- Adult Intensive Care Unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.,Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - J Day
- Adult Intensive Care Unit and Nuffield Department of Anaesthesia, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - S R McKechnie
- Adult Intensive Care Unit and Nuffield Department of Anaesthesia, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
277
|
Suzuki M, Ikari J, Anazawa R, Tanaka N, Katsumata Y, Shimada A, Suzuki E, Tatsumi K. PAD4 Deficiency Improves Bleomycin-induced Neutrophil Extracellular Traps and Fibrosis in Mouse Lung. Am J Respir Cell Mol Biol 2021; 63:806-818. [PMID: 32915635 DOI: 10.1165/rcmb.2019-0433oc] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Excessive release of neutrophil extracellular traps (NETs) has been implicated in several organ fibrosis, including pulmonary fibrosis. NETs constitute a phenomenon in which decorated nuclear chromatin with cytosolic proteins is released into the extracellular space. PAD4 (peptidylarginine deiminase 4) plays an important role in the formation of NETs. However, the role of NETs in the pathogenesis of pulmonary fibrosis remains undefined. Here, we identified NETs in the alveolar and interstitial lung space of mice undergoing bleomycin (BLM)-induced lung fibrosis, which was suppressed by a pan-PAD inhibitor, Cl-amidine. In vitro, BLM directly induced NETs in blood neutrophils, which was also inhibited by Cl-amidine. Furthermore, Padi4 gene knockout (PAD4-KO) in mice led to the alleviation of BLM-induced NETs and pulmonary fibrosis and to the expression of inflammatory and fibrotic genes. PAD4 deficiency prevented decreases in alveolar epithelial and pulmonary vascular endothelial cell numbers and increases in ACTA2-positive mesenchymal cells and S100A4-positive fibroblasts in the lung. Hematopoietic cell grafts from PAD4-KO mice, not wild-type mice, resolved BLM-induced lung fibrosis and fibrotic gene expression in wild-type and PAD4-KO mice, suggesting that expression of PAD4 in hematopoietic cells may be involved in the development of lung fibrosis. These data suggest that PAD4 deficiency could ameliorate BLM-induced formation of NETs and lung fibrosis, suggesting that this pathway could serve as a therapeutic target for pulmonary fibrosis treatment.
Collapse
Affiliation(s)
- Masaki Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Jun Ikari
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Rie Anazawa
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Nozomi Tanaka
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Yusuke Katsumata
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Ayako Shimada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Eiko Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba City, Chiba, Japan
| |
Collapse
|
278
|
Farouk AF, Shafqat A, Shafqat S, Kashir J, Alkattan K, Yaqinuddin A. COVID-19 associated cardiac disease: Is there a role of neutrophil extracellular traps in pathogenesis? AIMS MOLECULAR SCIENCE 2021. [DOI: 10.3934/molsci.2021021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
<abstract>
<p>The COVID-19 pandemic has driven an upheaval of new research, providing key insights into the pathogenesis of this disease. Lymphocytopenia, hyper-inflammation and cardiac involvement are prominent features of the disease and have prognostic value. However, the mechanistic links among these phenomena are not well understood. Likewise, some COVID-19 patients exhibit multi-organ failure with diseases affecting the cardiac system, appearing to be an emerging feature of the COVID-19 pandemic. Neutrophil extracellular traps (NETs) have been frequently correlated with larger infarct sizes and can predict major adverse cardiac events. However, the exact mechanism behind this remains unknown. Although the excessive NET formation can drive inflammation, particularly endothelial and promote thrombosis, it is essential to normal immunity. In this paper, we postulate the role of NETs in cardiac disease by providing an overview of the relationship between NET and inflammasome activities in lung and liver diseases, speculating a link between these entities in cardiac diseases as well. Future research is required to specify the role of NETs in COVID-19, since this carries potential therapeutic significance, as inhibition of NETosis could alleviate symptoms of this disease. Knowledge gained from this could serve to inform the assessment and therapeutics of other hyper inflammatory diseases affecting the heart and vasculature alike.</p>
</abstract>
Collapse
|
279
|
De Lucchi L, Nardin C, Sponchiado A, Raggi D, Faggin E, Martini E, Pagliara V, Callegari E, Caberlotto L, Plebani M, Pauletto P, Cinetto F, Agostini C, Villalta S, Rattazzi M. Serum uric acid levels and the risk of recurrent venous thromboembolism. J Thromb Haemost 2021; 19:194-201. [PMID: 33078502 DOI: 10.1111/jth.15139] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 08/31/2023]
Abstract
Essentials Increase in serum uric acid (SUA) levels has been widely associated with higher risk of cardiovascular disease. We investigated the link between SUA levels and the risk of venous thromboembolism (VTE) recurrence. Patients with SUA levels ≥ 4.38 mg/dL showed a three-fold increase in the risk of VTE recurrence. Elevated SUA levels are associated with increased risk of recurrent VTE independently from traditional risk factors. ABSTRACT: Background The link between serum uric acid (SUA) and the risk of cardiovascular disease is well established. However, the impact of SUA levels on the risk of venous thromboembolism (VTE) recurrence is unknown. Objectives To investigate the association between SUA and the risk of VTE recurrence. Patients and Methods We performed a monocenter, prospective study on 280 patients with a previous episode of VTE that completed the oral anticoagulant period. SUA levels at enrollment were correlated with the risk of VTE recurrence (mean follow-up 71.1 ± 29.2 months). Results Patients were stratified according to SUA tertiles distribution at baseline (tertiles cut-off: I ≤ 4.37 mg/dL, II 4.38--5.54 mg/dL, III ≥ 5.55 mg/dL). Fifty episodes of VTE recurrence occurred during the follow-up and Kaplan-Meier survival analysis showed that subjects in the lower tertile of SUA distribution had significantly lower risk of future VTE recurrence (P = .003). No differences were seen among patients belonging to the second and the third tertile of SUA distribution. A multivariate Cox regression analysis showed that higher tertiles of SUA distribution had about three-fold increase in the risk of VTE recurrence as compared to subjects with SUA ≤ 4.37, independently from potential confounders (hazard ratio [HR] 3.04, 95% confidence interval [CI] 1.15--8.05 P = .025). Moreover, we observed that the adjusted hazard of VTE recurrence increased by 30% for each additional unit of SUA (mg/dL; HR 1.30, 95% CI 1.01--1.22, P = .040). Conclusion Elevated SUA levels are associated with increased risk of future VTE recurrence independently from traditional risk factors.
Collapse
Affiliation(s)
- Lara De Lucchi
- Medicina Generale I^, Ca' Foncello University Hospital, Treviso, Italy
| | - Chiara Nardin
- Medicina Generale I^, Ca' Foncello University Hospital, Treviso, Italy
| | | | - Davide Raggi
- Department of Economics, University of Bologna, Bologna, Italy
| | | | - Elena Martini
- Medicina Generale I^, Ca' Foncello University Hospital, Treviso, Italy
| | - Valeria Pagliara
- Medicina Generale I^, Ca' Foncello University Hospital, Treviso, Italy
| | - Elena Callegari
- Medicina Generale I^, Ca' Foncello University Hospital, Treviso, Italy
| | - Livio Caberlotto
- Laboratory Medicine, Ca' Foncello University Hospital, Treviso, Italy
| | - Mario Plebani
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Paolo Pauletto
- ORAS Rehabilitation Hospital, Motta di Livenza, Treviso, Italy
| | - Francesco Cinetto
- Medicina Generale I^, Ca' Foncello University Hospital, Treviso, Italy
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Carlo Agostini
- Medicina Generale I^, Ca' Foncello University Hospital, Treviso, Italy
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Sabina Villalta
- Medicina Generale, Castelfranco Veneto Hospital, Castelfranco Veneto, Italy
| | - Marcello Rattazzi
- Medicina Generale I^, Ca' Foncello University Hospital, Treviso, Italy
- Department of Medicine - DIMED, University of Padova, Padova, Italy
| |
Collapse
|
280
|
Northup PG, Garcia-Pagan JC, Garcia-Tsao G, Intagliata NM, Superina RA, Roberts LN, Lisman T, Valla DC. Vascular Liver Disorders, Portal Vein Thrombosis, and Procedural Bleeding in Patients With Liver Disease: 2020 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology 2021; 73:366-413. [PMID: 33219529 DOI: 10.1002/hep.31646] [Citation(s) in RCA: 358] [Impact Index Per Article: 89.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Patrick G Northup
- Division of Gastroenterology and Hepatology, Center for the Study of Hemostasis in Liver Disease, University of Virginia, Charlottesville, VA
| | - Juan Carlos Garcia-Pagan
- Barcelona Hepatic Hemodynamic Laboratory, Liver Unit, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi I i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain.,Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE-Liver), Barcelona, Spain
| | - Guadalupe Garcia-Tsao
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT.,Veterans Administration Healthcare System, West Haven, CT
| | - Nicolas M Intagliata
- Division of Gastroenterology and Hepatology, Center for the Study of Hemostasis in Liver Disease, University of Virginia, Charlottesville, VA
| | - Riccardo A Superina
- Department of Transplant Surgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
| | - Lara N Roberts
- Department of Haematological Medicine, King's Thrombosis Centre, King's College Hospital National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Ton Lisman
- Section of Hepatobiliary Surgery and Liver Transplantation, Surgical Research Laboratory, Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Dominique C Valla
- Hepatology Service, Hospital Beaujon, Clichy, France.,Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE-Liver), Barcelona, Spain
| |
Collapse
|
281
|
Arepally GM, Padmanabhan A. Heparin-Induced Thrombocytopenia: A Focus on Thrombosis. Arterioscler Thromb Vasc Biol 2021; 41:141-152. [PMID: 33267665 PMCID: PMC7769912 DOI: 10.1161/atvbaha.120.315445] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 11/13/2020] [Indexed: 01/19/2023]
Abstract
Heparin-induced thrombocytopenia is an immune-mediated disorder caused by antibodies that recognize complexes of platelet factor 4 and heparin. Thrombosis is a central and unpredictable feature of this syndrome. Despite optimal management, disease morbidity and mortality from thrombosis remain high. The hypercoagulable state in heparin-induced thrombocytopenia is biologically distinct from other thrombophilic disorders in that clinical complications are directly attributable to circulating ultra-large immune complexes. In some individuals, ultra-large immune complexes elicit unchecked cellular procoagulant responses that culminate in thrombosis. To date, the clinical and biologic risk factors associated with thrombotic risk in heparin-induced thrombocytopenia remain elusive. This review will summarize our current understanding of thrombosis in heparin-induced thrombocytopenia with attention to its clinical features, cellular mechanisms, and its management.
Collapse
Affiliation(s)
| | - Anand Padmanabhan
- Divisions of Hematopathology, Transfusion Medicine, and Experimental Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN (A.P.)
| |
Collapse
|
282
|
Agrati C, Mazzotta V, Pinnetti C, Biava G, Bibas M. Venous thromboembolism in people living with HIV infection (PWH). Transl Res 2021; 227:89-99. [PMID: 32693031 DOI: 10.1016/j.trsl.2020.07.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 10/23/2022]
Abstract
The risk of venous thromboembolism (VTE) and of recurrent VTE remain elevated in people living with HIV compared to controls still with contemporary antiretroviral therapy (ART). The pathophysiology of VTE in HIV is multi factorial and includes an interplay among traditional risk factors, HIV-specific factors, behavioral factors, exposure to ART and other therapies, coinfections, and co-morbidities.
Collapse
Affiliation(s)
- Chiara Agrati
- Cellular Immunology and Pharmacology Laboratory, National Institute for Infectious Diseases 'Lazzaro Spallanzani'-IRCCS, Rome, Italy
| | - Valentina Mazzotta
- The Clinical and Research Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani'-IRCCS, Rome, Italy
| | - Carmela Pinnetti
- The Clinical and Research Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani'-IRCCS, Rome, Italy
| | - Gianluigi Biava
- The Clinical and Research Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani'-IRCCS, Rome, Italy
| | - Michele Bibas
- The Clinical and Research Department, National Institute for Infectious Diseases 'Lazzaro Spallanzani'-IRCCS, Rome, Italy.
| |
Collapse
|
283
|
Abstract
Neutrophils produce neutrophil extracellular traps (NETs) by expelling their extracellular chromatin embedded with citrullinated histone H3, myeloperoxidase, and other intracellular molecules. Since their discovery in 2004, numerous articles have demonstrated the mechanism of NET formation and their function in innate immunity and inflammation. NET components often play an antimicrobial role, but excessive NETs are deleterious and can cause inflammation and tissue damage. This review highlights recent advancements in the identification of novel pathways and mechanisms of NET formation. We also focus on the specific damaging impact of NETs in individual organs. We then discuss the progress and limitations of various NET detection assays. Collectively, these vital aspects of NETs significantly improve our understanding of the pathobiology of NETs and future diagnostics and therapeutic tools for examining and modulating NETs in inflammatory diseases.
Collapse
Affiliation(s)
- Chuyi Tan
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Departments of Surgery and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|
284
|
Leiva O, Newcomb R, Connors JM, Al-Samkari H. Cancer and thrombosis: new insights to an old problem. JOURNAL DE MÉDECINE VASCULAIRE 2020; 45:6S8-6S16. [PMID: 33276943 DOI: 10.1016/s2542-4513(20)30514-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Venous thromboembolism (VTE) is a common complication in patients with cancer and portends a poor prognosis. Our understanding of the underlying pathophysiology of VTE in cancer has advanced since Trousseau first described hypercoagulability in patients with malignancy and Virchow described his famous triad of thrombosis formation. Malignancy itself induces a thrombophilic state by increasing the risk of venous stasis, endothelial injury and an imbalance of pro and anti-thrombotic factors leading to a hypercoaguable state. Additional insults to this thrombotic balance are introduced by patient-specific, treatment related and tumor-specific factors. The importance of understanding the factors associated with increased thrombosis in cancer is paramount in order to adequately identify patients who will benefit from thromboprophylaxis.
Collapse
Affiliation(s)
- O Leiva
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - R Newcomb
- Division of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - J M Connors
- Hematology Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - H Al-Samkari
- Division of Hematology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
285
|
Rangaswamy C, Englert H, Deppermann C, Renné T. Polyanions in Coagulation and Thrombosis: Focus on Polyphosphate and Neutrophils Extracellular Traps. Thromb Haemost 2020; 121:1021-1030. [PMID: 33307564 DOI: 10.1055/a-1336-0526] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neutrophil extracellular traps (NETs) and polyphosphates (polyP) have been recognized as procoagulant polyanions. This review summarizes the activities and regulation of the two procoagulant mediators and compares their functions. NETs are composed of DNA which like polyP is built of phosphate units linked by high-energy phosphoanhydride bonds. Both NETs and polyP form insoluble particulate surfaces composed of a DNA/histone meshwork or Ca2+-rich nanoparticles, respectively. These polyanionic molecules modulate coagulation involving an array of mechanisms and trigger thrombosis via activation of the factor XII-driven procoagulant and proinflammatory contact pathway. Here, we outline the current knowledge on NETs and polyP with respect to their procoagulant and prothrombotic nature, strategies for interference of their activities in circulation, as well as the crosstalk between these two molecules. A better understanding of the underlying, cellular mechanisms will shed light on the therapeutic potential of targeting NETs and polyP in coagulation and thrombosis.
Collapse
Affiliation(s)
- Chandini Rangaswamy
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hanna Englert
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Deppermann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
286
|
Liu X, Fu Y, Wang J, Wu D, Li S, Wang C, Yang Z, Zhou E. β-Conglycinin induces the formation of neutrophil extracellular traps dependent on NADPH oxidase-derived ROS, PAD4, ERK1/2 and p38 signaling pathways in mice. Food Funct 2020; 12:154-161. [PMID: 33289753 DOI: 10.1039/d0fo02337j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
β-Conglycinin is one of the key thermostable anti-nutritional factors in soybean, which has strong immunogenicity that usually leads to weaning in some young animals such as piglets and calves and allergic reaction in rats. Neutrophils are involved in the pathogenesis of an allergy. However, the contribution of functional neutrophils to allergy needs to be clarified. The formation of neutrophil extracellular traps is a novel effector mechanism of neutrophils and has been extensively investigated in recent years. To the best of our knowledge, there is no information available on β-conglycinin-induced NETs. In this study, β-conglycinin-induced NET formation in mice was examined via immunofluorescence analysis and fluorescence microplate reader. The mechanism of β-conglycinin-induced NETs was investigated using inhibitors and fluorescent microplate methods. The results showed that β-conglycinin induced the classical characteristics of NETs, which mainly consist of DNA as the backbone and decorated with histones, myeloperoxidase (MPO) and neutrophil elastase (NE). Moreover, β-conglycinin significantly induced the formation of NETs in a dose-dependent way. NET degrading enzyme DNase I markedly reduced β-conglycinin-induced NETs, which suggests that β-conglycinin indeed triggered the release of NETs. Further investigation showed that the quantitation of NETs was markedly decreased by the inhibitors of reactive oxygen species (ROS)-derived-NADPH oxidase, ERK1/2, p38, Rac and PAD4 signaling pathways, indicating the crucial role of these signaling pathways in β-conglycinin-induced NETs. Furthermore, our findings revealed that β-conglycinin induced the formation of NETs, which is dependent on NADPH oxidase-derived ROS, ERK1/2, p38, Rac and PAD4 signaling pathways. This study is the first to demonstrate the underlying mechanisms of β-conglycinin-induced NET formation, and it could be helpful to understand diarrhea caused by β-conglycinin overexposure in young animals and provides the corresponding theoretical basis for clinical applications.
Collapse
Affiliation(s)
- Xiao Liu
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China and College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong 528231, People's Republic of China.
| | - Yunhe Fu
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Jingjing Wang
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China and College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong 528231, People's Republic of China.
| | - Di Wu
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Shuangqiu Li
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Chaoqun Wang
- College of Veterinary Medicine, Jilin University, Jilin, Changchun 130062, People's Republic of China
| | - Zhengtao Yang
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong 528231, People's Republic of China.
| | - Ershun Zhou
- College of Life Sciences and Engineering, Foshan University, Foshan, Guangdong 528231, People's Republic of China.
| |
Collapse
|
287
|
Vorobjeva NV, Chernyak BV. NETosis: Molecular Mechanisms, Role in Physiology and Pathology. BIOCHEMISTRY (MOSCOW) 2020; 85:1178-1190. [PMID: 33202203 PMCID: PMC7590568 DOI: 10.1134/s0006297920100065] [Citation(s) in RCA: 243] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
NETosis is a program for formation of neutrophil extracellular traps (NETs), which consist of modified chromatin decorated with bactericidal proteins from granules and cytoplasm. Various pathogens, antibodies and immune complexes, cytokines, microcrystals, and other physiological stimuli can cause NETosis. Induction of NETosis depends on reactive oxygen species (ROS), the main source of which is NADPH oxidase. Activation of NADPH oxidase depends on increase in the concentration of Ca2+ in the cytoplasm and in some cases on the generation of ROS in mitochondria. NETosis includes release of the granule components into the cytosol, modification of histones leading to chromatin decondensation, destruction of the nuclear envelope, as well as formation of pores in the plasma membrane. In this review, basic mechanisms of NETosis, as well as its role in the pathogenesis of some diseases including COVID-19 are discussed.
Collapse
Affiliation(s)
- N V Vorobjeva
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | - B V Chernyak
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.
| |
Collapse
|
288
|
Yang J, Wu Z, Long Q, Huang J, Hong T, Liu W, Lin J. Insights Into Immunothrombosis: The Interplay Among Neutrophil Extracellular Trap, von Willebrand Factor, and ADAMTS13. Front Immunol 2020; 11:610696. [PMID: 33343584 PMCID: PMC7738460 DOI: 10.3389/fimmu.2020.610696] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 11/02/2020] [Indexed: 12/24/2022] Open
Abstract
Both neutrophil extracellular traps (NETs) and von Willebrand factor (VWF) are essential for thrombosis and inflammation. During these processes, a complex series of events, including endothelial activation, NET formation, VWF secretion, and blood cell adhesion, aggregation and activation, occurs in an ordered manner in the vasculature. The adhesive activity of VWF multimers is regulated by a specific metalloprotease ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motifs, member 13). Increasing evidence indicates that the interaction between NETs and VWF contributes to arterial and venous thrombosis as well as inflammation. Furthermore, contents released from activated neutrophils or NETs induce the reduction of ADAMTS13 activity, which may occur in both thrombotic microangiopathies (TMAs) and acute ischemic stroke (AIS). Recently, NET is considered as a driver of endothelial damage and immunothrombosis in COVID-19. In addition, the levels of VWF and ADAMTS13 can predict the mortality of COVID-19. In this review, we summarize the biological characteristics and interactions of NETs, VWF, and ADAMTS13, and discuss their roles in TMAs, AIS, and COVID-19. Targeting the NET-VWF axis may be a novel therapeutic strategy for inflammation-associated TMAs, AIS, and COVID-19.
Collapse
Affiliation(s)
- Junxian Yang
- Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Zhiwei Wu
- Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Quan Long
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Jiaqi Huang
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Tiantian Hong
- Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Wang Liu
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Jiangguo Lin
- Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
289
|
Current knowledge into the role of the peptidylarginine deiminase (PAD) enzyme family in cardiovascular disease. Eur J Pharmacol 2020; 891:173765. [PMID: 33249073 DOI: 10.1016/j.ejphar.2020.173765] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/12/2020] [Accepted: 11/23/2020] [Indexed: 11/20/2022]
Abstract
Peptidylarginine deiminase (PAD) family members have a vital role in maintaining the stability of the extracellular matrix (ECM) during remodelling in several heart diseases. PAD-mediated deamination, or citrullination, has been studied in different physiological and pathological conditions in the body. However, the role of PAD isoforms has not been fully studied in cardiovascular system. Citrullination is a post-translational modification that involves conversion of peptidyl-based arginine to peptidyl-based citrulline by PAD family members in a calcium-dependent manner. Upregulation of PADs have been observed in various cardiovascular diseases, including venous thrombosis, cardiac fibrosis, heart failure, atherosclerosis, coronary heart disease and acute inflammation. In this review, experimental aspects of in vivo and in vitro studies related to the roles PAD isoforms in cardiovascular diseases including mechanisms, pathophysiological and therapeutic properties are discussed. Pharmacological strategies for targeting PAD family proteins in cardiac diseases have not yet been studied. Furthermore, the role played by PAD family members in the remodelling process during the progression of cardiovascular diseases is not fully understood.
Collapse
|
290
|
Anderson R, Theron AJ, Steel HC, Nel JG, Tintinger GR. ADP-Mediated Upregulation of Expression of CD62P on Human Platelets Is Critically Dependent on Co-Activation of P2Y1 and P2Y12 Receptors. Pharmaceuticals (Basel) 2020; 13:ph13120420. [PMID: 33255391 PMCID: PMC7760858 DOI: 10.3390/ph13120420] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
This study probed the differential utilization of P2Y1 and P2Y12 receptors in mobilizing CD62P (P-selectin) from intracellular granules following activation of human platelets with adenosine 5′-diphosphate (ADP, 100 µmol·L−1) Platelet-rich plasma (PRP) was prepared from the blood of adult humans. CD62P was measured by flow cytometry following activation of PRP with ADP in the absence and presence of the selective antagonists of P2Y1 and P2Y12 receptors, MRS2500 and PSB0739 (both 0.155–10 µmol·L−1), respectively. Effects of the test agents on ADP-activated, CD62P-dependent formation of neutrophil:platelet (NP) aggregates were also measured by flow cytometry, while phosphatidylinositol 3-kinase (PI3K) activity was measured according to Akt1 phosphorylation in platelet lysates. Treatment with MRS2500 or PSB0739 at 10 µmol·L−1 almost completely attenuated (94.6% and 86% inhibition, respectively) ADP-activated expression of CD62P and also inhibited NP aggregate formation. To probe the mechanisms involved in P2Y1/P2Y12 receptor-mediated expression of CD62P, PRP was pre-treated with U73122 (phospholipase C (PLC) inhibitor), 2-aminoethoxy-diphenyl borate (2-APB, inositol triphosphate receptor antagonist), calmidazolium chloride (calmodulin inhibitor), or wortmannin (PI3K inhibitor). U73122, 2-APB, and wortmannin caused almost complete inhibition of ADP-activated expression of CD62P, while calmidazolium chloride caused statistically significant, partial inhibition. PSB0739, but not MRS2500, caused potent inhibition of PI3K-mediated phosphorylation of Akt1. Optimal mobilization of CD62P by ADP-stimulated platelets is critically dependent on the co-activation of platelet P2Y1 and P2Y12 receptors. P2Y12 receptor activation is the key event in activation of PI3K, while activation of the P2Y1 receptor appears to create a high cytosolic Ca2+ environment conducive to optimum PI3K activity.
Collapse
Affiliation(s)
- Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (A.J.T.); (H.C.S.)
- Correspondence: ; Tel.: +27-12-318-2425; Fax: +27-12-323-0732
| | - Annette J. Theron
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (A.J.T.); (H.C.S.)
| | - Helen C. Steel
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (A.J.T.); (H.C.S.)
| | - Jan G. Nel
- Department of Haematology, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
- Tshwane Academic Division of the National Laboratory Health Service of South Africa, Pretoria 0001, South Africa
| | - Gregory R. Tintinger
- Department of Internal Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa;
| |
Collapse
|
291
|
The role of anticoagulation in pylephlebitis: a retrospective examination of characteristics and outcomes. J Thromb Thrombolysis 2020; 49:325-331. [PMID: 31493290 DOI: 10.1007/s11239-019-01949-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pylephlebitis, or suppurative thrombophlebitis of the portal vein, typically occurs in the context of an intraabdominal infection or abdominal sepsis. Antibiotics are the mainstay of treatment. The role of anticoagulation in the management of pylephlebitis is controversial, and data regarding its impact on outcomes is limited. The records of 67 consecutive patients with pylephlebitis treated at our institution over a 19 year period were retrospectively reviewed. Data was gathered regarding their baseline characteristics, presentations, management, and outcomes. Patients who did and did not receive anticoagulation were compared. Outcomes of interest included survival, portal vein thrombosis (PVT) resolution, development of chronic symptomatic portal hypertension, and major bleeding. Forty-seven patients received anticoagulation and 20 did not. The anticoagulated and non-anticoagulated groups did not differ significantly with respect to potential covariates or confounders. Anticoagulated patients had significantly higher rates of PVT resolution than non-anticoagulated patients (58% vs. 21%, p = 0.0201). This translated to lower rates of future chronic portal hypertensive symptoms among anticoagulated patients (11% vs. 47%, p = 0.0034). Anticoagulated patients had a trend toward improved survival however this improvement was not significant on multivariable analysis. There was no significant difference in rates of major bleeding between groups. Thrombophilia testing was common in this cohort however the occurrence of meaningful positive results was exceedingly low. Anticoagulation significantly improves the rate of PVT resolution, and significantly reduces the rate of chronic symptomatic portal hypertension, among patients with pylephlebitis. Treatment of pylephlebitis should incorporate the use of systemic anticoagulation whenever possible.
Collapse
|
292
|
Jiang L, Tang K, Levin M, Irfan O, Morris SK, Wilson K, Klein JD, Bhutta ZA. COVID-19 and multisystem inflammatory syndrome in children and adolescents. THE LANCET. INFECTIOUS DISEASES 2020; 20:e276-e288. [PMID: 32818434 PMCID: PMC7431129 DOI: 10.1016/s1473-3099(20)30651-4] [Citation(s) in RCA: 515] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/17/2020] [Accepted: 07/20/2020] [Indexed: 02/07/2023]
Abstract
As severe acute respiratory syndrome coronavirus 2 continues to spread worldwide, there have been increasing reports from Europe, North America, Asia, and Latin America describing children and adolescents with COVID-19-associated multisystem inflammatory conditions. However, the association between multisystem inflammatory syndrome in children and COVID-19 is still unknown. We review the epidemiology, causes, clinical features, and current treatment protocols for multisystem inflammatory syndrome in children and adolescents associated with COVID-19. We also discuss the possible underlying pathophysiological mechanisms for COVID-19-induced inflammatory processes, which can lead to organ damage in paediatric patients who are severely ill. These insights provide evidence for the need to develop a clear case definition and treatment protocol for this new condition and also shed light on future therapeutic interventions and the potential for vaccine development. TRANSLATIONS: For the French, Chinese, Arabic, Spanish and Russian translations of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Li Jiang
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Kun Tang
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, ON, Canada; Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Mike Levin
- Department of Infectious Disease, Imperial College London, London, UK
| | - Omar Irfan
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Shaun K Morris
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, ON, Canada; Division of Infectious Diseases, The Hospital for Sick Children, Toronto, ON, Canada
| | - Karen Wilson
- Mount Sinai Kravis Children's Hospital, New York, NY, USA
| | - Jonathan D Klein
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, USA
| | - Zulfiqar A Bhutta
- Centre for Global Child Health, The Hospital for Sick Children, Toronto, ON, Canada; Institute for Global Health and Development, Aga Khan University, Karachi, Pakistan.
| |
Collapse
|
293
|
McBane RD, Torres Roldan VD, Niven AS, Pruthi RK, Franco PM, Linderbaum JA, Casanegra AI, Oyen LJ, Houghton DE, Marshall AL, Ou NN, Siegel JL, Wysokinski WE, Padrnos LJ, Rivera CE, Flo GL, Shamoun FE, Silvers SM, Nayfeh T, Urtecho M, Shah S, Benkhadra R, Saadi SM, Firwana M, Jawaid T, Amin M, Prokop LJ, Murad MH. Anticoagulation in COVID-19: A Systematic Review, Meta-analysis, and Rapid Guidance From Mayo Clinic. Mayo Clin Proc 2020; 95:2467-2486. [PMID: 33153635 PMCID: PMC7458092 DOI: 10.1016/j.mayocp.2020.08.030] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/04/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022]
Abstract
A higher risk of thrombosis has been described as a prominent feature of coronavirus disease 2019 (COVID-19). This systematic review synthesizes current data on thrombosis risk, prognostic implications, and anticoagulation effects in COVID-19. We included 37 studies from 4070 unique citations. Meta-analysis was performed when feasible. Coagulopathy and thrombotic events were frequent among patients with COVID-19 and further increased in those with more severe forms of the disease. We also present guidance on the prevention and management of thrombosis from a multidisciplinary panel of specialists from Mayo Clinic. The current certainty of evidence is generally very low and continues to evolve.
Collapse
Key Words
- aptt, activated thromboplastin time
- covid-19, coronavirus disease 2019
- dic, disseminated intravascular coagulation
- doac, direct oral anticoagulant
- dvt, deep venous thrombosis
- icu, intensive care unit
- lmwh, low-molecular-weight heparin
- or, odds ratio
- pe, pulmonary embolism
- pt, prothrombin time
- sars-cov, severe acute respiratory syndrome coronavirus
- sc, subcutaneously
- vte, venous thromboembolism
Collapse
Affiliation(s)
- Robert D McBane
- Gonda Vascular Center, Mayo Clinic, Rochester, MN; Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Victor D Torres Roldan
- Evidence-based Practice Center and Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Alexander S Niven
- Division of Pulmonary and Critical Care, Center for Sleep Medicine, Mayo Clinic, Rochester, MN
| | - Rajiv K Pruthi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Division of Hematology, Mayo Clinic, Rochester, MN
| | | | | | - Ana I Casanegra
- Gonda Vascular Center, Mayo Clinic, Rochester, MN; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Lance J Oyen
- Department of Pharmacy, Mayo Clinic, Rochester, MN
| | - Damon E Houghton
- Gonda Vascular Center, Mayo Clinic, Rochester, MN; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Ariela L Marshall
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Division of Hematology, Mayo Clinic, Rochester, MN
| | - Narith N Ou
- Department of Pharmacy, Mayo Clinic, Rochester, MN
| | | | - Waldemar E Wysokinski
- Gonda Vascular Center, Mayo Clinic, Rochester, MN; Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | | | - Candido E Rivera
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL
| | - Gayle L Flo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Fadi E Shamoun
- Department of Cardiovascular Medicine, Mayo Clinic, Scottsdale, AZ
| | - Scott M Silvers
- Department of Emergency Medicine, Mayo Clinic, Jacksonville, FL
| | - Tarek Nayfeh
- Evidence-based Practice Center and Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Meritxell Urtecho
- Evidence-based Practice Center and Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Sahrish Shah
- Evidence-based Practice Center and Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Raed Benkhadra
- Evidence-based Practice Center and Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Samer Mohir Saadi
- Evidence-based Practice Center and Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Mohammed Firwana
- Evidence-based Practice Center and Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Tabinda Jawaid
- Evidence-based Practice Center and Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Mustapha Amin
- Evidence-based Practice Center and Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | | | - M Hassan Murad
- Evidence-based Practice Center and Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN.
| |
Collapse
|
294
|
Li JH, Tong DX, Wang Y, Gao L, Liu Y, Zhang XH, Chen WJ, Chi JY, Liu N, Yang K, Wang SP, Xu Y, Li Y, Yin XH, Liu WX. Neutrophil extracellular traps exacerbate coagulation and endothelial damage in patients with essential hypertension and hyperhomocysteinemia. Thromb Res 2020; 197:36-43. [PMID: 33166900 DOI: 10.1016/j.thromres.2020.10.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 10/23/2022]
Abstract
Patients with essential hypertension (EH) and hyperhomocysteinemia (HHCY) suffer from more increased thrombotic events than those in EH alone. However, the underlying mechanisms for this effect are not well understood. This study hypothesized that neutrophil extracellular trap (NET) releasing may be triggered by HHCY in patients in EH, thereby predisposing them to a more hypercoagulable state. Using a modified-capture enzyme-linked immunosorbent assay (ELISA) method, we observed that cell-free DNA (CF-DNA) and myeloperoxidase DNA (MPO-DNA) in patients With EH and HHCY were significantly higher. The NET formation was also positively correlated with homocysteine levels, neutrophil-lymphocyte ratio (NLR), and hypercoagulable markers (thrombin-antithrombin complex, D-dimers). Furthermore, neutrophils from patients in EH with HHCY were found to be predisposed to amplified NET release when compared to patients in EH without HHCY or CTR. Coagulation function assays showed that NETs in patients With EH and HHCY resulted in a significantly increased ability to generate thrombin and fibrin than in those in EH without HHCY or CTR. These procoagulant effects of NETs in patients With EH and HHCY were markedly inhibited (approximately 70%) by the cleavage of NETs with DNase I. Isolated NETs from patients With EH and HHCY neutrophils also exerted a strong cytotoxic effect on endothelial cells (ECs), converted them to apoptosis. This study revealed a previously unrecognized association between the hypercoagulable state and neutrophils in patients With EH and HHCY. Therefore, blocking NETs may represent a new therapeutic objective for preventing thrombosis in these patients.
Collapse
Affiliation(s)
- Ji-He Li
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Dong-Xia Tong
- Department of Oncology, Qingdao Municipal Hospital Group, Qingdao 266000, Shandong Province, China
| | - Ying Wang
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Lei Gao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Yue Liu
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Xiao-Hui Zhang
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Wen-Jia Chen
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Jin-Yv Chi
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Na Liu
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Kelaier Yang
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Shi-Peng Wang
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Yang Xu
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Yang Li
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Xin-Hua Yin
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
| | - Wen-Xiu Liu
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|
295
|
DeRoo E, Zhou T, Liu B. The Role of RIPK1 and RIPK3 in Cardiovascular Disease. Int J Mol Sci 2020; 21:E8174. [PMID: 33142926 PMCID: PMC7663726 DOI: 10.3390/ijms21218174] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/29/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases, including peripheral arterial and venous disease, myocardial infarction, and stroke, are the number one cause of death worldwide annually. In the last 20 years, the role of necroptosis, a newly identified form of regulated necrotic cell death, in cardiovascular disease has come to light. Specifically, the damaging role of two kinase proteins pivotal in the necroptosis pathway, Receptor Interacting Protein Kinase 1 (RIPK1) and Receptor Interacting Protein Kinase 3 (RIPK3), in cardiovascular disease has become a subject of great interest and importance. In this review, we provide an overview of the current evidence supporting a pathologic role of RIPK1 and RIPK3 in cardiovascular disease. Moreover, we highlight the evidence behind the efficacy of targeted RIPK1 and RIPK3 inhibitors in the prevention and treatment of cardiovascular disease.
Collapse
Affiliation(s)
| | | | - Bo Liu
- Department of Surgery, Division of Vascular Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA; (E.D.); (T.Z.)
| |
Collapse
|
296
|
Sriram K, Insel PA. Inflammation and thrombosis in COVID-19 pathophysiology: proteinase-activated and purinergic receptors as drivers and candidate therapeutic targets. Physiol Rev 2020; 101:545-567. [PMID: 33124941 PMCID: PMC8238137 DOI: 10.1152/physrev.00035.2020] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Evolving information has identified disease mechanisms and dysregulation of host biology that might be targeted therapeutically in coronavirus disease 2019 (COVID-19). Thrombosis and coagulopathy, associated with pulmonary injury and inflammation, are emerging clinical features of COVID-19. We present a framework for mechanisms of thrombosis in COVID-19 that initially derive from interaction of SARS-CoV-2 with ACE2, resulting in dysregulation of angiotensin signaling and subsequent inflammation and tissue injury. These responses result in increased signaling by thrombin (proteinase-activated) and purinergic receptors, which promote platelet activation and exert pathological effects on other cell types (e.g., endothelial cells, epithelial cells, and fibroblasts), further enhancing inflammation and injury. Inhibitors of thrombin and purinergic receptors may, thus, have therapeutic effects by blunting platelet-mediated thromboinflammation and dysfunction in other cell types. Such inhibitors include agents (e.g., anti-platelet drugs) approved for other indications, and that could be repurposed to treat, and potentially improve the outcome of, COVID-19 patients. COVID-19, caused by the SARS-CoV-2 virus, drives dysregulation of angiotensin signaling, which, in turn, increases thrombin-mediated and purinergic-mediated activation of platelets and increase in inflammation. This thromboinflammation impacts the lungs and can also have systemic effects. Inhibitors of receptors that drive platelet activation or inhibitors of the coagulation cascade provide opportunities to treat COVID-19 thromboinflammation.
Collapse
Affiliation(s)
- Krishna Sriram
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Paul A Insel
- Department of Pharmacology and Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
297
|
Vergallo R, Jang IK, Crea F. New prediction tools and treatment for ACS patients with plaque erosion. Atherosclerosis 2020; 318:45-51. [PMID: 33127074 DOI: 10.1016/j.atherosclerosis.2020.10.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/15/2020] [Accepted: 10/08/2020] [Indexed: 11/26/2022]
Abstract
For decades, we have known from autopsy observations that the proximate cause of the majority of acute coronary syndromes ( ACS) is occlusive thrombosis generated by plaque rupture or, less frequently, superficial erosion. Patients with ACS caused by plaque erosion seem to have a better long-term prognosis compared to those with plaque rupture, and may be stabilized by dual antiplatelet therapy without the need for stenting in a non-trivial proportion of cases, limiting the expenses and potential complications of invasive procedures. The accurate prediction of plaque erosion and the identification of specific biomarkers that could be used at the point-of-care without the need of invasive imaging would take us a step closer to the holy grail of precision medicine in patients with ACS.
Collapse
Affiliation(s)
- Rocco Vergallo
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Filippo Crea
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Cardiovascular and Pulmonary Sciences, Università Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
298
|
Abstract
CONTEXT.— The coronavirus disease 2019 (COVID-19) is a highly contagious respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Coagulation dysfunction is a hallmark in patients with COVID-19. Fulminant thrombotic complications emerge as critical issues in patients with severe COVID-19. OBJECTIVE.— To present a review of the literature and discuss the mechanisms of COVID-19 underlying coagulation activation and the implications for anticoagulant and thrombolytic treatment in the management of COVID-19. DATA SOURCES.— We performed a systemic review of scientific papers on the topic of COVID-19, available online via the PubMed NCBI, medRxiv, and Preprints as of May 15, 2020. We also shared our experience on the management of thrombotic events in patients with COVID-19. CONCLUSIONS.— COVID-19-associated coagulopathy ranges from mild laboratory alterations to disseminated intravascular coagulation (DIC) with a predominant phenotype of thrombotic/multiple organ failure. Characteristically, high D-dimer levels on admission and/or continuously increasing concentrations of D-dimer are associated with disease progression and poor overall survival. SARS-CoV-2 infection triggers the immune-hemostatic response. Drastic inflammatory responses including, but not limited to, cytokine storm, vasculopathy, and NETosis may contribute to an overwhelming activation of coagulation. Hypercoagulability and systemic thrombotic complications necessitate anticoagulant and thrombolytic interventions, which provide opportunities to prevent or reduce "excessive" thrombin generation while preserving "adaptive" hemostasis and bring additional benefit via their anti-inflammatory effect in the setting of COVID-19.
Collapse
Affiliation(s)
- Yang Fei
- From the Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Fei, Tang)
| | - Ning Tang
- From the Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (Fei, Tang)
| | - Hefei Liu
- the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City (Liu, Cao)
| | - Wenjing Cao
- the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City (Liu, Cao)
| |
Collapse
|
299
|
Essig F, Kollikowski AM, Pham M, Solymosi L, Stoll G, Haeusler KG, Kraft P, Schuhmann MK. Immunohistological Analysis of Neutrophils and Neutrophil Extracellular Traps in Human Thrombemboli Causing Acute Ischemic Stroke. Int J Mol Sci 2020; 21:ijms21197387. [PMID: 33036337 PMCID: PMC7582743 DOI: 10.3390/ijms21197387] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 12/28/2022] Open
Abstract
Ischemic stroke caused by thromboembolic occlusion of large cerebral arteries, such as the internal carotid (ICA) and/or the middle cerebral artery (MCA), is treated by mechanical thrombectomy (MT). MT allows salvage of the vessel-occluding thrombemboli, which most frequently originate from the left atrium or the left ventricle of the heart or from sites of plaque rupture within large arteries above the heart. Clot composition may influence the efficacy of (intravenous) thrombolysis and MT, respectively. We analyzed 37 human thrombemboli obtained from acute ischemic stroke patients during MT with special emphasis on histological staining of neutrophils and neutrophil extracellular traps (NETs). We found neutrophils as the main cellular component of cerebral thrombemboli but encountered considerable morphological heterogeneity. Neutrophils accumulated in the border region of fibrin-rich structures indicating possible interaction of neutrophils with distinct structural thrombembolus components. Web-like NETs were found in 35 of 37 thrombemboli in varying amounts. NETs were almost exclusively found within fibrin-rich areas. Importantly, stroke etiology, age and present oral anticoagulation was associated with morphological patterns and the amount of neutrophils. Correlation of histological data and imaging data revealed that relative Hounsfield units of cerebral thrombemboli positively correlated with the amount of red blood cells. In summary, our results demonstrate that neutrophils and NETs are substantial constituents of cerebral thrombemboli and contribute to their structural complexity.
Collapse
Affiliation(s)
- Fabian Essig
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany; (F.E.); (G.S.); (K.G.H.); (P.K.)
| | - Alexander M. Kollikowski
- Department of Neuroradiology, University Hospital Würzburg, 97080 Würzburg, Germany; (A.M.K.); (M.P.); (L.S.)
| | - Mirko Pham
- Department of Neuroradiology, University Hospital Würzburg, 97080 Würzburg, Germany; (A.M.K.); (M.P.); (L.S.)
| | - László Solymosi
- Department of Neuroradiology, University Hospital Würzburg, 97080 Würzburg, Germany; (A.M.K.); (M.P.); (L.S.)
| | - Guido Stoll
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany; (F.E.); (G.S.); (K.G.H.); (P.K.)
| | - Karl Georg Haeusler
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany; (F.E.); (G.S.); (K.G.H.); (P.K.)
| | - Peter Kraft
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany; (F.E.); (G.S.); (K.G.H.); (P.K.)
- Department of Neurology, Klinikum Main-Spessart, 97816 Lohr, Germany
| | - Michael K. Schuhmann
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany; (F.E.); (G.S.); (K.G.H.); (P.K.)
- Correspondence: ; Tel.: +49-931-201-23653
| |
Collapse
|
300
|
Shi C, Yang L, Braun A, Anders HJ. Extracellular DNA-A Danger Signal Triggering Immunothrombosis. Front Immunol 2020; 11:568513. [PMID: 33117353 PMCID: PMC7575749 DOI: 10.3389/fimmu.2020.568513] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022] Open
Abstract
Clotting and inflammation are effective danger response patterns positively selected by evolution to limit fatal bleeding and pathogen invasion upon traumatic injuries. As a trade-off, thrombotic, and thromboembolic events complicate severe forms of infectious and non-infectious states of acute and chronic inflammation, i.e., immunothrombosis. Factors linked to thrombosis and inflammation include mediators released by platelet granules, complement, and lipid mediators and certain integrins. Extracellular deoxyribonucleic acid (DNA) was a previously unrecognized cellular component in the blood, which elicits profound proinflammatory and prothrombotic effects. Pathogens trigger the release of extracellular DNA together with other pathogen-associated molecular patterns. Dying cells in the inflamed or infected tissue release extracellular DNA together with other danger associated molecular pattern (DAMPs). Neutrophils release DNA by forming neutrophil extracellular traps (NETs) during infection, trauma or other forms of vascular injury. Fluorescence tissue imaging localized extracellular DNA to sites of injury and to intravascular thrombi. Functional studies using deoxyribonuclease (DNase)-deficient mouse strains or recombinant DNase show that extracellular DNA contributes to the process of immunothrombosis. Here, we review rodent models of immunothrombosis and the evolving evidence for extracellular DNA as a driver of immunothrombosis and discuss challenges and prospects for extracellular DNA as a potential therapeutic target.
Collapse
Affiliation(s)
- Chongxu Shi
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Luying Yang
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| | - Attila Braun
- German Center for Lung Research, Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Hans-Joachim Anders
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians University Munich, Munich, Germany
| |
Collapse
|