251
|
Silva TF, Tomiotto-Pellissier F, Sanfelice RA, Gonçalves MD, da Silva Bortoleti BT, Detoni MB, Rodrigues ACJ, Carloto ACM, Concato VM, Siqueira EDS, Costa IN, Pavanelli WR, Conchon-Costa I, Miranda-Sapla MM. A 21st Century Evil: Immunopathology and New Therapies of COVID-19. Front Immunol 2020; 11:562264. [PMID: 33193331 PMCID: PMC7652766 DOI: 10.3389/fimmu.2020.562264] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus Disease 2019 (COVID-19) has been classified as a global threat, affecting millions of people and killing thousands. It is caused by the SARS-CoV-2 virus, which emerged at the end of 2019 in Wuhan, China, quickly spreading worldwide. COVID-19 is a disease with symptoms that range from fever and breathing difficulty to acute respiratory distress and death, critically affecting older patients and people with previous comorbidities. SARS-CoV-2 uses the angiotensin-converting enzyme 2 (ACE2) receptor and mainly spreads through the respiratory tract, which it then uses to reach several organs. The immune system of infected patients has been demonstrated to suffer important alterations, such as lymphopenia, exhausted lymphocytes, excessive amounts of inflammatory monocytes and macrophages, especially in the lungs, and cytokine storms, which may contribute to its severity and difficulty of establishing an effective treatment. Even though no specific treatment is currently available, several studies have been investigating potential therapeutic strategies, including the use of previously approved drugs and immunotherapy. In this context, this review addresses the interaction between SARS-CoV-2 and the patient's host immune system during infection, in addition to discussing the main immunopathological mechanisms involved in the development of the disease and potential new therapeutic approaches.
Collapse
Affiliation(s)
- Taylon Felipe Silva
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer—LIDNC, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | | | - Raquel Arruda Sanfelice
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer—LIDNC, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Manoela Daiele Gonçalves
- Laboratory of Biotransformation and Phytochemistry, Department of Chemistry, Center of Exact Sciences, State University of Londrina, Londrina, Brazil
| | | | - Mariana Barbosa Detoni
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer—LIDNC, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Ana Carolina Jacob Rodrigues
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer—LIDNC, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Amanda Cristina Machado Carloto
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer—LIDNC, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Virgínia Márcia Concato
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer—LIDNC, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Elaine da Silva Siqueira
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer—LIDNC, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Idessania Nazareth Costa
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer—LIDNC, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Wander Rogério Pavanelli
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer—LIDNC, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Ivete Conchon-Costa
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer—LIDNC, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| | - Milena Menegazzo Miranda-Sapla
- Laboratory of Immunoparasitology of Neglected Diseases and Cancer—LIDNC, Department of Pathological Sciences, Center of Biological Sciences, State University of Londrina, Londrina, Brazil
| |
Collapse
|
252
|
Sanguedolce F, Zanelli M, Ascani S, Zizzo M, Tortorella S, Soriano A, Cavazza A, Sollitto F, Loizzi D. SARS-CoV-2-related lung pathology: macroscopic and histologic features and their clinical implications. Panminerva Med 2020; 64:80-95. [PMID: 33073555 DOI: 10.23736/s0031-0808.20.04157-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The ongoing global coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been posing challenges to proper patients' management. Lungs are the first, and often the most affected organ by SARS-CoV-2; viral infection involves and damages both epithelial and vascular compartments, sometimes leading to severe and even fatal acute respiratory distress syndrome. Histopathological findings, mainly from postmortem examination of COVID-19 deceased patients, have been increasingly published in the last few months, helping to elucidate the sequence of events resulting in organ injury, and the complex multifactorial pathogenesis of this novel disease. A multidisciplinary approach to autopsy, including light microscopy examination along with the detection of viral proteins and/or RNA in tissue samples through ancillary techniques, provided crucial information on the mechanisms underlying the often-heterogeneous clinical picture of COVID-19.
Collapse
Affiliation(s)
- Francesca Sanguedolce
- Pathology Unit, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Foggia, Foggia, Italy -
| | - Magda Zanelli
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Stefano Ascani
- Pathology Unit, Azienda Ospedaliera S. Maria di Terni, University of Perugia, Terni, Italy
| | - Maurizio Zizzo
- Surgical Oncology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Simona Tortorella
- Pathology Unit, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Foggia, Foggia, Italy
| | - Alessandra Soriano
- Gastroenterology, Division and Inflammatory Bowel Disease Center Department of Internal Medicine, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alberto Cavazza
- Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | | | - Domenico Loizzi
- Institute of Thoracic Surgery, University of Foggia, Foggia, Italy
| |
Collapse
|
253
|
Ng J, Guo F, Marneth AE, Ghanta S, Kwon MY, Keegan J, Liu X, Wright KT, Kamaz B, Cahill LA, Mullally A, Perrella MA, Lederer JA. Augmenting emergency granulopoiesis with CpG conditioned mesenchymal stromal cells in murine neutropenic sepsis. Blood Adv 2020; 4:4965-4979. [PMID: 33049055 PMCID: PMC7556132 DOI: 10.1182/bloodadvances.2020002556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/02/2020] [Indexed: 12/17/2022] Open
Abstract
Patients with immune deficiencies from cancers and associated treatments represent a growing population within the intensive care unit with increased risk of morbidity and mortality from sepsis. Mesenchymal stromal cells (MSCs) are an integral part of the hematopoietic niche and express toll-like receptors, making them candidate cells to sense and translate pathogenic signals into an innate immune response. In this study, we demonstrate that MSCs administered therapeutically in a murine model of radiation-associated neutropenia have dual actions to confer a survival benefit in Pseudomonas aeruginosa pneumo-sepsis that is not from improved bacterial clearance. First, MSCs augment the neutrophil response to infection, an effect that is enhanced when MSCs are preconditioned with CpG oligodeoxynucleotide, a toll-like receptor 9 agonist. Using cytometry by time of flight, we identified proliferating neutrophils (Ly6GlowKi-67+) as the main expanded cell population within the bone marrow. Further analysis revealed that CpG-MSCs expand a lineage restricted progenitor population (Lin-Sca1+C-kit+CD150-CD48+) in the bone marrow, which corresponded to a doubling in the myeloid proliferation and differentiation potential in response to infection compared with control. Despite increased neutrophils, no reduction in organ bacterial count was observed between experimental groups. However, the second effect exerted by CpG-MSCs is to attenuate organ damage, particularly in the lungs. Neutrophils obtained from irradiated mice and cocultured with CpG-MSCs had decreased neutrophil extracellular trap formation, which was associated with decreased citrullinated H3 staining in the lungs of mice given CpG-MSCs in vivo. Thus, this preclinical study provides evidence for the therapeutic potential of MSCs in neutropenic sepsis.
Collapse
Affiliation(s)
- Julie Ng
- Division of Pulmonary and Critical Care, Department of Medicine
| | | | | | | | - Min-Young Kwon
- Division of Pulmonary and Critical Care, Department of Medicine
| | | | - Xiaoli Liu
- Division of Pulmonary and Critical Care, Department of Medicine
- Department of Pediatric Newborn Medicine, and
| | - Kyle T Wright
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | | | | | - Mark A Perrella
- Division of Pulmonary and Critical Care, Department of Medicine
- Department of Pediatric Newborn Medicine, and
| | | |
Collapse
|
254
|
Chiang CC, Korinek M, Cheng WJ, Hwang TL. Targeting Neutrophils to Treat Acute Respiratory Distress Syndrome in Coronavirus Disease. Front Pharmacol 2020; 11:572009. [PMID: 33162887 PMCID: PMC7583590 DOI: 10.3389/fphar.2020.572009] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/11/2020] [Indexed: 01/08/2023] Open
Abstract
This review describes targeting neutrophils as a potential therapeutic strategy for acute respiratory distress syndrome (ARDS) associated with coronavirus disease 2019 (COVID-19), a pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Neutrophil counts are significantly elevated in patients with COVID-19 and significantly correlated with disease severity. The neutrophil-to-lymphocyte ratio can serve as a clinical marker for predicting fatal complications related to ARDS in patients with COVID-19. Neutrophil-associated inflammation plays a critical pathogenic role in ARDS. The effector functions of neutrophils, acting as respiratory burst oxidants, granule proteases, and neutrophil extracellular traps, are linked to the pathogenesis of ARDS. Hence, neutrophils can not only be used as pathogenic markers but also as candidate drug targets for COVID-19 associated ARDS.
Collapse
Affiliation(s)
- Chih-Chao Chiang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Puxin Fengze Chinese Medicine Clinic, Taoyuan, Taiwan
| | - Michal Korinek
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wei-Jen Cheng
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| |
Collapse
|
255
|
Hazeldine J, Lord JM. Immunesenescence: A Predisposing Risk Factor for the Development of COVID-19? Front Immunol 2020; 11:573662. [PMID: 33123152 PMCID: PMC7573102 DOI: 10.3389/fimmu.2020.573662] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/28/2020] [Indexed: 01/08/2023] Open
Abstract
Bearing a strong resemblance to the phenotypic and functional remodeling of the immune system that occurs during aging (termed immunesenescence), the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of Coronavirus disease 2019 (COVID-19), is characterized by an expansion of inflammatory monocytes, functional exhaustion of lymphocytes, dysregulated myeloid responses and the presence of highly activated senescent T cells. Alongside advanced age, male gender and pre-existing co-morbidities [e.g., obesity and type 2 diabetes (T2D)] are emerging as significant risk factors for COVID-19. Interestingly, immunesenescence is more profound in males when compared to females, whilst accelerated aging of the immune system, termed premature immunesenescence, has been described in obese subjects and T2D patients. Thus, as three distinct demographic groups with an increased susceptibility to COVID-19 share a common immune profile, could immunesenescence be a generic contributory factor in the development of severe COVID-19? Here, by focussing on three key aspects of an immune response, namely pathogen recognition, elimination and resolution, we address this question by discussing how immunesenescence may weaken or exacerbate the immune response to SARS-CoV-2. We also highlight how aspects of immunesenescence could render potential COVID-19 treatments less effective in older adults and draw attention to certain therapeutic options, which by reversing or circumventing certain features of immunesenescence may prove to be beneficial for the treatment of groups at high risk of severe COVID-19.
Collapse
Affiliation(s)
- Jon Hazeldine
- Medical Research Council-Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Janet M. Lord
- Medical Research Council-Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
- National Institute for Health Research Birmingham Biomedical Research Centre, University Hospital Birmingham National Health Service Foundation Trust and University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
256
|
Abstract
An ongoing global pandemic of viral pneumonia (coronavirus disease [COVID-19]), due to the virus SARS-CoV-2, has infected millions of people and remains a threat to many more. Most critically ill patients have respiratory failure and there is an international effort to understand mechanisms and predictors of disease severity. Coagulopathy, characterized by elevations in D-dimer and fibrin(ogen) degradation products (FDPs), is associated with critical illness and mortality in patients with COVID-19. Furthermore, increasing reports of microvascular and macrovascular thrombi suggest that hemostatic imbalances may contribute to the pathophysiology of SARS-CoV-2 infection. We review the laboratory and clinical findings of patients with COVID-19-associated coagulopathy, and prior studies of hemostasis in other viral infections and acute respiratory distress syndrome. We hypothesize that an imbalance between coagulation and inflammation may result in a hypercoagulable state. Although thrombosis initiated by the innate immune system is hypothesized to limit SARS-CoV-2 dissemination, aberrant activation of this system can cause endothelial injury resulting in loss of thromboprotective mechanisms, excess thrombin generation, and dysregulation of fibrinolysis and thrombosis. The role various components including neutrophils, neutrophil extracellular traps, activated platelets, microparticles, clotting factors, inflammatory cytokines, and complement play in this process remains an area of active investigation and ongoing clinical trials target these different pathways in COVID-19.
Collapse
Affiliation(s)
- Meaghan E Colling
- Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yogendra Kanthi
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, and Ann Arbor Veterans Administration Healthcare System, Ann Arbor, MI, USA
| |
Collapse
|
257
|
Jarrahi A, Ahluwalia M, Khodadadi H, da Silva Lopes Salles E, Kolhe R, Hess DC, Vale F, Kumar M, Baban B, Vaibhav K, Dhandapani KM. Neurological consequences of COVID-19: what have we learned and where do we go from here? J Neuroinflammation 2020; 17:286. [PMID: 32998763 PMCID: PMC7525232 DOI: 10.1186/s12974-020-01957-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
The coronavirus disease-19 (COVID-19) pandemic is an unprecedented worldwide health crisis. COVID-19 is caused by SARS-CoV-2, a highly infectious pathogen that is genetically similar to SARS-CoV. Similar to other recent coronavirus outbreaks, including SARS and MERS, SARS-CoV-2 infected patients typically present with fever, dry cough, fatigue, and lower respiratory system dysfunction, including high rates of pneumonia and acute respiratory distress syndrome (ARDS); however, a rapidly accumulating set of clinical studies revealed atypical symptoms of COVID-19 that involve neurological signs, including headaches, anosmia, nausea, dysgeusia, damage to respiratory centers, and cerebral infarction. These unexpected findings may provide important clues regarding the pathological sequela of SARS-CoV-2 infection. Moreover, no efficacious therapies or vaccines are currently available, complicating the clinical management of COVID-19 patients and emphasizing the public health need for controlled, hypothesis-driven experimental studies to provide a framework for therapeutic development. In this mini-review, we summarize the current body of literature regarding the central nervous system (CNS) effects of SARS-CoV-2 and discuss several potential targets for therapeutic development to reduce neurological consequences in COVID-19 patients.
Collapse
Affiliation(s)
- Abbas Jarrahi
- Department of Neurosurgery, Medical College of Georgia, Augusta University, 1120 15th Street, 30912, Augusta, Georgia
| | - Meenakshi Ahluwalia
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Hesam Khodadadi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia
| | - Evila da Silva Lopes Salles
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Fernando Vale
- Department of Neurosurgery, Medical College of Georgia, Augusta University, 1120 15th Street, 30912, Augusta, Georgia
| | - Manish Kumar
- Department of Allied Health Science, Shri B. M. Patil Medical College, Hospital and Research Centre, BLDE (Deemed to be University), Vijayapura, Karnataka, India
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, 1120 15th Street, 30912, Augusta, Georgia
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, 1120 15th Street, 30912, Augusta, Georgia.
| |
Collapse
|
258
|
Radermecker C, Detrembleur N, Guiot J, Cavalier E, Henket M, d'Emal C, Vanwinge C, Cataldo D, Oury C, Delvenne P, Marichal T. Neutrophil extracellular traps infiltrate the lung airway, interstitial, and vascular compartments in severe COVID-19. J Exp Med 2020; 217:152084. [PMID: 32926097 PMCID: PMC7488867 DOI: 10.1084/jem.20201012] [Citation(s) in RCA: 262] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/10/2020] [Accepted: 08/10/2020] [Indexed: 01/08/2023] Open
Abstract
Infection with SARS-CoV-2 is causing a deadly and pandemic disease called coronavirus disease-19 (COVID-19). While SARS-CoV-2-triggered hyperinflammatory tissue-damaging and immunothrombotic responses are thought to be major causes of respiratory failure and death, how they relate to lung immunopathological changes remains unclear. Neutrophil extracellular traps (NETs) can contribute to inflammation-associated lung damage, thrombosis, and fibrosis. However, whether NETs infiltrate particular compartments in severe COVID-19 lungs remains to be clarified. Here we analyzed postmortem lung specimens from four patients who succumbed to COVID-19 and four patients who died from a COVID-19-unrelated cause. We report the presence of NETs in the lungs of each COVID-19 patient. NETs were found in the airway compartment and neutrophil-rich inflammatory areas of the interstitium, while NET-prone primed neutrophils were present in arteriolar microthrombi. Our results support the hypothesis that NETs may represent drivers of severe pulmonary complications of COVID-19 and suggest that NET-targeting approaches could be considered for the treatment of uncontrolled tissue-damaging and thrombotic responses in COVID-19.
Collapse
Affiliation(s)
- Coraline Radermecker
- Laboratory of Immunophysiology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Institute, Liege University, Liege, Belgium.,Faculty of Veterinary Medicine, Liege University, Liege, Belgium
| | - Nancy Detrembleur
- Department of Pathology, Clinique Hospitalo-Universitaire (CHU) University Hospital, Liege University, Liege, Belgium.,Laboratory of Experimental Pathology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Institute, Liege University, Liege, Belgium
| | - Julien Guiot
- Pneumology department, Clinique Hospitalo-Universitaire (CHU) Liège, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Institute, Liege University, Liege, Belgium.,Laboratory of Pneumology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Institute, Liege University, Liege, Belgium
| | - Etienne Cavalier
- Medical Chemistry, Center for Interdisciplinary Research on Medicines Institute, Liege University, Liege, Belgium
| | - Monique Henket
- Pneumology department, Clinique Hospitalo-Universitaire (CHU) Liège, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Institute, Liege University, Liege, Belgium.,Laboratory of Pneumology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Institute, Liege University, Liege, Belgium
| | - Céline d'Emal
- Laboratory of Cardiology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Institute, Liege University, Liege, Belgium
| | - Céline Vanwinge
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Institute, Liege University, Liege, Belgium
| | - Didier Cataldo
- Laboratory of Tumor and Development Biology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Institute, Liege University, Liege, Belgium
| | - Cécile Oury
- Laboratory of Cardiology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Institute, Liege University, Liege, Belgium
| | - Philippe Delvenne
- Department of Pathology, Clinique Hospitalo-Universitaire (CHU) University Hospital, Liege University, Liege, Belgium.,Laboratory of Experimental Pathology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Institute, Liege University, Liege, Belgium
| | - Thomas Marichal
- Laboratory of Immunophysiology, Grappe Interdisciplinaire de Génoprotéomique Appliquée (GIGA) Institute, Liege University, Liege, Belgium.,Faculty of Veterinary Medicine, Liege University, Liege, Belgium.,Walloon Excellence in Life Sciences and Biotechnology, Wallonia, Belgium
| |
Collapse
|
259
|
Beltrán-García J, Osca-Verdegal R, Pallardó FV, Ferreres J, Rodríguez M, Mulet S, Sanchis-Gomar F, Carbonell N, García-Giménez JL. Oxidative Stress and Inflammation in COVID-19-Associated Sepsis: The Potential Role of Anti-Oxidant Therapy in Avoiding Disease Progression. Antioxidants (Basel) 2020; 9:E936. [PMID: 33003552 PMCID: PMC7599810 DOI: 10.3390/antiox9100936] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/20/2020] [Accepted: 09/27/2020] [Indexed: 02/07/2023] Open
Abstract
Since the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) outbreak emerged, countless efforts are being made worldwide to understand the molecular mechanisms underlying the coronavirus disease 2019 (COVID-19) in an attempt to identify the specific clinical characteristics of critically ill COVID-19 patients involved in its pathogenesis and provide therapeutic alternatives to minimize COVID-19 severity. Recently, COVID-19 has been closely related to sepsis, which suggests that most deceases in intensive care units (ICU) may be a direct consequence of SARS-CoV-2 infection-induced sepsis. Understanding oxidative stress and the molecular inflammation mechanisms contributing to COVID-19 progression to severe phenotypes such as sepsis is a current clinical need in the effort to improve therapies in SARS-CoV-2 infected patients. This article aims to review the molecular pathogenesis of SARS-CoV-2 and its relationship with oxidative stress and inflammation, which can contribute to sepsis progression. We also provide an overview of potential antioxidant therapies and active clinical trials that might prevent disease progression or reduce its severity.
Collapse
Affiliation(s)
- Jesús Beltrán-García
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, 46010 Valencia, Spain; (J.B.-G.); (F.V.P.)
- Department of Physiology, Faculty of Medicine & Dentistry, University of Valencia, 46010 Valencia, Spain; (R.O.-V.); (F.S.-G.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (J.F.); (M.R.); (S.M.)
- EpiDisease S.L. (Spin-Off CIBER-ISCIII), Parc Científic de la Universitat de València, 46980 Paterna, Valencia, Spain
| | - Rebeca Osca-Verdegal
- Department of Physiology, Faculty of Medicine & Dentistry, University of Valencia, 46010 Valencia, Spain; (R.O.-V.); (F.S.-G.)
| | - Federico V. Pallardó
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, 46010 Valencia, Spain; (J.B.-G.); (F.V.P.)
- Department of Physiology, Faculty of Medicine & Dentistry, University of Valencia, 46010 Valencia, Spain; (R.O.-V.); (F.S.-G.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (J.F.); (M.R.); (S.M.)
- EpiDisease S.L. (Spin-Off CIBER-ISCIII), Parc Científic de la Universitat de València, 46980 Paterna, Valencia, Spain
| | - José Ferreres
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (J.F.); (M.R.); (S.M.)
- Intensive Care Unit, Clinical University Hospital of Valencia, 46010 Valencia, Spain
| | - María Rodríguez
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (J.F.); (M.R.); (S.M.)
- Intensive Care Unit, Clinical University Hospital of Valencia, 46010 Valencia, Spain
| | - Sandra Mulet
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (J.F.); (M.R.); (S.M.)
- Intensive Care Unit, Clinical University Hospital of Valencia, 46010 Valencia, Spain
| | - Fabian Sanchis-Gomar
- Department of Physiology, Faculty of Medicine & Dentistry, University of Valencia, 46010 Valencia, Spain; (R.O.-V.); (F.S.-G.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (J.F.); (M.R.); (S.M.)
| | - Nieves Carbonell
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (J.F.); (M.R.); (S.M.)
- Intensive Care Unit, Clinical University Hospital of Valencia, 46010 Valencia, Spain
| | - José Luis García-Giménez
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, 46010 Valencia, Spain; (J.B.-G.); (F.V.P.)
- Department of Physiology, Faculty of Medicine & Dentistry, University of Valencia, 46010 Valencia, Spain; (R.O.-V.); (F.S.-G.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain; (J.F.); (M.R.); (S.M.)
- EpiDisease S.L. (Spin-Off CIBER-ISCIII), Parc Científic de la Universitat de València, 46980 Paterna, Valencia, Spain
| |
Collapse
|
260
|
Nebulized in-line endotracheal dornase alfa and albuterol administered to mechanically ventilated COVID-19 patients: a case series. Mol Med 2020; 26:91. [PMID: 32993479 PMCID: PMC7522910 DOI: 10.1186/s10020-020-00215-w] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022] Open
Abstract
Background Mechanically ventilated patients with COVID-19 have a mortality of 24–53%, in part due to distal mucopurulent secretions interfering with ventilation. DNA from neutrophil extracellular traps (NETs) contribute to the viscosity of mucopurulent secretions and NETs are found in the serum of COVID-19 patients. Dornase alfa is recombinant human DNase 1 and is used to digest DNA in mucoid sputum. Here, we report a single-center case series where dornase alfa was co-administered with albuterol through an in-line nebulizer system. Methods Demographic and clinical data were collected from the electronic medical records of five mechanically ventilated patients with COVID-19—including three requiring veno-venous extracorporeal membrane oxygenation—treated with nebulized in-line endotracheal dornase alfa and albuterol, between March 31 and April 24, 2020. Data on tolerability and response were analyzed. Results The fraction of inspired oxygen requirements was reduced for all five patients after initiating dornase alfa administration. All patients were successfully extubated, discharged from hospital and remain alive. No drug-associated toxicities were identified. Conclusions Results suggest that dornase alfa will be well-tolerated by patients with severe COVID-19. Clinical trials are required to formally test the dosing, safety, and efficacy of dornase alfa in COVID-19, and several have been recently registered.
Collapse
|
261
|
Yang SC, Tsai YF, Pan YL, Hwang TL. Understanding the role of neutrophils in acute respiratory distress syndrome. Biomed J 2020; 44:439-446. [PMID: 33087299 PMCID: PMC7481802 DOI: 10.1016/j.bj.2020.09.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is difficult to treat and is associated with a high mortality rate. The most severe form of coronavirus disease 2019 (COVID-19) also leads to life-threatening ARDS. Neutrophil counts are positively correlated with disease severity in ARDS. Neutrophil activation not only plays a significant role in immune defense against infections, but also causes tissue damage and leads to inflammatory diseases. Activated neutrophils rapidly migrate to inflamed lung tissue, releasing toxic granular contents and generating neutrophil extracellular traps. In the last few decades, it has become apparent that neutrophils occupy a central role in ARDS pathology. In this review, we summarize the neutrophil inflammatory responses and their relationships to ARDS. According to the current literature, understanding the function of neutrophils may be helpful in the treatment of ARDS.
Collapse
Affiliation(s)
- Shun-Chin Yang
- Department of Anesthesiology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan; Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yung-Fong Tsai
- Department of Anesthesiology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yen-Lin Pan
- Department of Pharmacy, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan.
| |
Collapse
|
262
|
Meijenfeldt FAV, Jenne CN. Netting Liver Disease: Neutrophil Extracellular Traps in the Initiation and Exacerbation of Liver Pathology. Semin Thromb Hemost 2020; 46:724-734. [PMID: 32906176 DOI: 10.1055/s-0040-1715474] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The liver plays a vital role in the immune system. Its unique position in the portal circulation and the architecture of the hepatic sinusoids, in combination with the wide-ranged population of immunocompetent cells, make the liver function as an immune filter. To aid in pathogen clearance, once challenged, the liver initiates the rapid recruitment of a wide variety of inflammatory cells, including neutrophils. These neutrophils, in conjunction with platelets, facilitate the release of neutrophil extracellular traps (NETs), which are web-like structures of decondensed nuclear DNA, histones, and neutrophil proteins. NETs function as both a physical and a chemical barrier, binding and killing pathogens circulating in the blood stream. In addition to their antimicrobial role, NETs also bind platelets, activate coagulation, and exacerbate host inflammatory response. This interplay between inflammation and coagulation drives microvascular occlusion, ischemia, and (sterile) damage in liver disease. Although direct clinical evidence of this interplay is scarce, preliminary studies indicate that NETs contribute to progression of liver disease and (thrombotic) complications. Here, we provide an overview of the pathological mechanisms of NETs in liver disease. In addition, we summarize clinical evidence for NETs in different disease etiologies and complications of liver disease and discuss the possible implications for the use of NETs as a diagnostic marker and a therapeutic target in liver disease.
Collapse
Affiliation(s)
- Fien A von Meijenfeldt
- Surgical Research Laboratory and Section of Hepatobiliary Surgery and Liver Transplantation, Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Craig N Jenne
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, The University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
263
|
Henry B, Cheruiyot I, Vikse J, Mutua V, Kipkorir V, Benoit J, Plebani M, Bragazzi N, Lippi G. Lymphopenia and neutrophilia at admission predicts severity and mortality in patients with COVID-19: a meta-analysis. ACTA BIO-MEDICA : ATENEI PARMENSIS 2020; 91:e2020008. [PMID: 32921706 PMCID: PMC7716963 DOI: 10.23750/abm.v91i3.10217] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND There is a compelling need to identify clinical and laboratory predictors of unfavorable clinical course and death in patients with coronavirus disease (COVID-19). A trend towards low lymphocyte count and high neutrophil counts in patients with poor outcomes has been reported by earlier studies. We aim to synthesize existing data evaluating the relationship between clinical outcomes and abnormal neutrophil and lymphocyte counts at admission in COVID-19 patients. METHODS An electronic search was carried out in PubMed, China National Knowledge Infrastructure (CNKI) and Cochrane Central Register of Controlled Trials (CENTRAL) to identify eligible studies reporting frequency data on neutrophilia and lymphopenia at admission in hospitalization in COVID-19 patients. Pooled odds ratios of clinical outcomes for each parameter were calculated using Comprehensive Meta-Analysis. RESULTS A total of 22 studies (4,969 patients) were included in this meta-analysis. Lymphopenia at admission was found to be significantly associated with increased odd of progression to severe disease (odds ratio [OR], 4.20; 95% confidence interval [95CI%], 3.46-5.09) and death (OR, 3.71; 95%CI, 1.63-8.44). Neutrophilia at admission was also found to be significantly associated with increased odd of progression to severe disease (OR, 7.99; 95%CI, 1.77-36.14) and death (OR, 7.87; 95%CI, 1.75-35.35). Subgroup analysis revealed that COVID-19 patients with severe lymphopenia (<0.5 x10×9/L) had 12-fold increased odds of in-hospital mortality. CONCLUSION Admission lymphopenia and neutrophilia are associated with poor outcomes in patients with COVID-19. Regular monitoring and early and even more aggressive intervention shall hence be advisable in patients with low lymphocyte and high neutrophil counts. These variables may be useful in risk stratification models.
Collapse
Affiliation(s)
- Brandon Henry
- 1. Cardiac Intensive Care Unit, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | | | - Jens Vikse
- Clinical Immunology Unit, Stavanger University Hospital, Norway.
| | | | | | - Justin Benoit
- 4. Department of Emergency Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Mario Plebani
- Department of Laboratory Medicine, University Hospital of Padova, Padova, Italy.
| | - Nicola Bragazzi
- 6. Laboratory for Industrial and Applied Mathematics (LIAM), Department of Mathematics and Statistics, York University, Toronto, Canada.
| | - Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, Verona, Italy.
| |
Collapse
|
264
|
Middleton EA, He XY, Denorme F, Campbell RA, Ng D, Salvatore SP, Mostyka M, Baxter-Stoltzfus A, Borczuk AC, Loda M, Cody MJ, Manne BK, Portier I, Harris ES, Petrey AC, Beswick EJ, Caulin AF, Iovino A, Abegglen LM, Weyrich AS, Rondina MT, Egeblad M, Schiffman JD, Yost CC. Neutrophil extracellular traps contribute to immunothrombosis in COVID-19 acute respiratory distress syndrome. Blood 2020; 136:1169-1179. [PMID: 32597954 PMCID: PMC7472714 DOI: 10.1182/blood.2020007008] [Citation(s) in RCA: 1075] [Impact Index Per Article: 215.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/25/2020] [Indexed: 01/08/2023] Open
Abstract
COVID-19 affects millions of patients worldwide, with clinical presentation ranging from isolated thrombosis to acute respiratory distress syndrome (ARDS) requiring ventilator support. Neutrophil extracellular traps (NETs) originate from decondensed chromatin released to immobilize pathogens, and they can trigger immunothrombosis. We studied the connection between NETs and COVID-19 severity and progression. We conducted a prospective cohort study of COVID-19 patients (n = 33) and age- and sex-matched controls (n = 17). We measured plasma myeloperoxidase (MPO)-DNA complexes (NETs), platelet factor 4, RANTES, and selected cytokines. Three COVID-19 lung autopsies were examined for NETs and platelet involvement. We assessed NET formation ex vivo in COVID-19 neutrophils and in healthy neutrophils incubated with COVID-19 plasma. We also tested the ability of neonatal NET-inhibitory factor (nNIF) to block NET formation induced by COVID-19 plasma. Plasma MPO-DNA complexes increased in COVID-19, with intubation (P < .0001) and death (P < .0005) as outcome. Illness severity correlated directly with plasma MPO-DNA complexes (P = .0360), whereas Pao2/fraction of inspired oxygen correlated inversely (P = .0340). Soluble and cellular factors triggering NETs were significantly increased in COVID-19, and pulmonary autopsies confirmed NET-containing microthrombi with neutrophil-platelet infiltration. Finally, COVID-19 neutrophils ex vivo displayed excessive NETs at baseline, and COVID-19 plasma triggered NET formation, which was blocked by nNIF. Thus, NETs triggering immunothrombosis may, in part, explain the prothrombotic clinical presentations in COVID-19, and NETs may represent targets for therapeutic intervention.
Collapse
Affiliation(s)
- Elizabeth A Middleton
- Molecular Medicine Program and
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Xue-Yan He
- Cold Spring Harbor Laboratory, Cancer Center, Cold Spring Harbor, NY
| | | | - Robert A Campbell
- Molecular Medicine Program and
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - David Ng
- Cold Spring Harbor Laboratory, Cancer Center, Cold Spring Harbor, NY
| | - Steven P Salvatore
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital, New York, NY
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Maria Mostyka
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital, New York, NY
| | - Amelia Baxter-Stoltzfus
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital, New York, NY
| | - Alain C Borczuk
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital, New York, NY
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, New York Presbyterian Hospital, New York, NY
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Mark J Cody
- Molecular Medicine Program and
- Department of Pediatrics and
| | | | | | - Estelle S Harris
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Aaron C Petrey
- Molecular Medicine Program and
- Department of Pathology, University of Utah, Salt Lake City, UT
| | - Ellen J Beswick
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | | | - Anthony Iovino
- Department of Pediatrics and
- PEEL Therapeutics, Inc, Salt Lake City, UT; and
| | - Lisa M Abegglen
- Department of Pediatrics and
- PEEL Therapeutics, Inc, Salt Lake City, UT; and
| | - Andrew S Weyrich
- Molecular Medicine Program and
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
| | - Matthew T Rondina
- Molecular Medicine Program and
- Department of Internal Medicine, University of Utah, Salt Lake City, UT
- Department of Internal Medicine and
- Geriatric Research, Education, and Clinical Center (GRECC), George E. Wahlen Veterans Affairs (VA) Medical Center, Salt Lake City, UT
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cancer Center, Cold Spring Harbor, NY
| | - Joshua D Schiffman
- Molecular Medicine Program and
- Department of Pediatrics and
- PEEL Therapeutics, Inc, Salt Lake City, UT; and
| | | |
Collapse
|
265
|
Allegra A, Innao V, Allegra AG, Musolino C. Coagulopathy and thromboembolic events in patients with SARS-CoV-2 infection: pathogenesis and management strategies. Ann Hematol 2020; 99:1953-1965. [PMID: 32671455 PMCID: PMC7363407 DOI: 10.1007/s00277-020-04182-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023]
Abstract
In October 2019, a viral infectious disease appeared in the city of Wuhan in China. A new betacoronavirus, SARS-CoV-2, has been recognized as the responsible pathogen in this infection. Although coronavirus disease is principally expressed as a pulmonary infection, critical SARS-CoV-2 infection is frequently complicated with coagulopathy, and thromboembolic events are recognizable in several patients. Dehydration, acute inflammatory condition, protracted immobilization during disease, existence of multiple cardiovascular risk factors such as diabetes, obesity or hypertension, previous coronary artery disease, ischemic stroke, peripheral artery disease are frequent comorbidities in SARS-CoV-2 hospitalized subjects, which possibly augment thrombo-embolic risk. However, other causal factors can still be identified such as unrestricted angiotensin II action, the use of immunoglobulins, an increased production of adhesion molecules able to induce vascular inflammation and endothelial activation, complement stimulation, excessive production of neutrophil extracellular traps (NETs), and increased platelet count. Low-molecular-weight heparin should be chosen as early treatment because of its anti-inflammatory action and its ability to antagonize histones and so defend the endothelium. However, several therapeutic possibilities have also been proposed such as fibrinolytic treatment, drugs that target NETs, and complement inhibition. Nevertheless, although the violence of the pandemic may suggest the use of heroic treatments to reduce the frightening mortality that accompanies SARS-CoV-2 infection, we believe that experimental treatments should only be used within approved and controlled protocols, the only ones that can provide useful and specify information on the validity of the treatments.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125, Messina, Italy.
- COVID Centre AOU Policlinic G. Martino, Messina, Italy.
| | - Vanessa Innao
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125, Messina, Italy
| | - Andrea Gaetano Allegra
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125, Messina, Italy
| | - Caterina Musolino
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, 98125, Messina, Italy
| |
Collapse
|
266
|
Semler MW, Bernard GR, Aaron SD, Angus DC, Biros MH, Brower RG, Calfee CS, Colantuoni EA, Ferguson ND, Gong MN, Hopkins RO, Hough CL, Iwashyna TJ, Levy BD, Martin TR, Matthay MA, Mizgerd JP, Moss M, Needham DM, Self WH, Seymour CW, Stapleton RD, Thompson BT, Wunderink RG, Aggarwal NR, Reineck LA. Identifying Clinical Research Priorities in Adult Pulmonary and Critical Care. NHLBI Working Group Report. Am J Respir Crit Care Med 2020; 202:511-523. [PMID: 32150460 PMCID: PMC7427373 DOI: 10.1164/rccm.201908-1595ws] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 03/06/2020] [Indexed: 12/14/2022] Open
Abstract
Preventing, treating, and promoting recovery from critical illness due to pulmonary disease are foundational goals of the critical care community and the NHLBI. Decades of clinical research in acute respiratory distress syndrome, acute respiratory failure, pneumonia, and sepsis have yielded improvements in supportive care, which have translated into improved patient outcomes. Novel therapeutics have largely failed to translate from promising preclinical findings into improved patient outcomes in late-phase clinical trials. Recent advances in personalized medicine, "big data," causal inference using observational data, novel clinical trial designs, preclinical disease modeling, and understanding of recovery from acute illness promise to transform the methods of pulmonary and critical care clinical research. To assess the current state of, research priorities for, and future directions in adult pulmonary and critical care research, the NHLBI assembled a multidisciplinary working group of investigators. This working group identified recommendations for future research, including 1) focusing on understanding the clinical, physiological, and biological underpinnings of heterogeneity in syndromes, diseases, and treatment response with the goal of developing targeted, personalized interventions; 2) optimizing preclinical models by incorporating comorbidities, cointerventions, and organ support; 3) developing and applying novel clinical trial designs; and 4) advancing mechanistic understanding of injury and recovery to develop and test interventions targeted at achieving long-term improvements in the lives of patients and families. Specific areas of research are highlighted as especially promising for making advances in pneumonia, acute hypoxemic respiratory failure, and acute respiratory distress syndrome.
Collapse
Affiliation(s)
| | | | - Shawn D. Aaron
- Division of Respirology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Michelle H. Biros
- Department of Emergency Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Roy G. Brower
- Division of Pulmonary and Critical Care Medicine and
| | - Carolyn S. Calfee
- Department of Medicine and
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | | | - Niall D. Ferguson
- Interdepartmental Division of Critical Care Medicine
- Department of Medicine
- Department of Physiology, and
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Michelle N. Gong
- Department of Epidemiology
- Department of Population Health, and
- Department of Medicine, Montefiore Medical Center, Bronx, New York
| | - Ramona O. Hopkins
- Department of Psychology, Brigham Young University, Provo, Utah
- Pulmonary and Critical Care Division, Intermountain Medical Center, Murray, Utah
| | - Catherine L. Hough
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington
| | - Theodore J. Iwashyna
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, Michigan
| | - Bruce D. Levy
- Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thomas R. Martin
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington
| | - Michael A. Matthay
- Department of Medicine and
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Joseph P. Mizgerd
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Marc Moss
- Division of Pulmonary Sciences & Critical Care, University of Colorado, Denver, Colorado
| | | | - Wesley H. Self
- Department of Emergency Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christopher W. Seymour
- Department of Critical Care Medicine and
- Department of Emergency Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Renee D. Stapleton
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Vermont, Burlington, Vermont
| | - B. Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Harvard University, Boston, Massachusetts
| | - Richard G. Wunderink
- Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, Illinois; and
| | | | | |
Collapse
|
267
|
Gautam S, Stahl Y, Young GM, Howell R, Cohen AJ, Tsang DA, Martin T, Sharma L, Dela Cruz CS. Quantification of bronchoalveolar neutrophil extracellular traps and phagocytosis in murine pneumonia. Am J Physiol Lung Cell Mol Physiol 2020; 319:L661-L669. [PMID: 32783617 DOI: 10.1152/ajplung.00316.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The past two decades have witnessed a resurgence in neutrophil research, inspired in part by the discovery of neutrophil extracellular traps (NETs) and their myriad roles in health and disease. Within the lung, dysregulation of neutrophils and NETosis have been linked to an array of diseases including pneumonia, cystic fibrosis, acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), and severe asthma. However, our understanding of pathologic neutrophil responses in the lung remains incomplete. Two methodologic issues have contributed to this gap: first, an emphasis on studying neutrophils from blood rather than the lung and second, the technical difficulties of interrogating neutrophil responses in mice, which has largely restricted basic murine research to specialized laboratories. To address these limitations, we have developed a suite of techniques for studying neutrophil effector functions specifically in the mouse lung. These include ex vivo assays for phagocytosis and NETosis using bronchoalveolar neutrophils and in situ evaluation of NETosis in a murine model of pneumonia. Throughout, we have prioritized technical ease and robust, quantitative readouts. We hope these assays will help to standardize research on lung neutrophils and improve accessibility to this burgeoning field.
Collapse
Affiliation(s)
- Samir Gautam
- Yale School of Medicine, Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University, New Haven, Connecticut
| | - Yannick Stahl
- Yale School of Medicine, Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University, New Haven, Connecticut
| | - Grant M Young
- Yale School of Medicine, Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University, New Haven, Connecticut
| | - Rebecca Howell
- Department of Chemistry, Yale University, New Haven, Connecticut
| | - Avi J Cohen
- Yale School of Medicine, Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University, New Haven, Connecticut
| | - Derek A Tsang
- Yale School of Medicine, Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University, New Haven, Connecticut
| | - Tommy Martin
- Yale School of Medicine, Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University, New Haven, Connecticut
| | - Lokesh Sharma
- Yale School of Medicine, Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University, New Haven, Connecticut
| | - Charles S Dela Cruz
- Yale School of Medicine, Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale University, New Haven, Connecticut.,Yale School of Medicine, Department of Microbial Pathogenesis, Yale University, New Haven, Connecticut
| |
Collapse
|
268
|
Neutrophil Extracellular Traps: Signaling Properties and Disease Relevance. Mediators Inflamm 2020; 2020:9254087. [PMID: 32774152 PMCID: PMC7407020 DOI: 10.1155/2020/9254087] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
Neutrophil extracellular traps (NETs) are characterized as extracellular DNA fibers comprised of histone and cytoplasmic granule proteins. NETs were first described as a form of innate response against pathogen invasion, which can capture pathogens, degrade bacterial toxic factors, and kill bacteria. Additionally, NETs also provide a scaffold for protein and cell binding. Protein binding to NETs further activate the coagulation system which participates in thrombosis. In addition, NETs also can damage the tissues due to the proteins they carry. Many studies have suggested that the excessive formation of NETs may contribute to a range of diseases, including thrombosis, atherosclerosis, autoimmune diseases, and sepsis. In this review, we describe the structure and components of NETs, models of NET formation, and detection methods. We also discuss the molecular mechanism of NET formation and their disease relevance. Modulation of NET formation may provide a new route for the prevention and treatment of releated human diseases.
Collapse
|
269
|
Finnerty DT, Buggy DJ. A novel role for lidocaine in COVID-19 patients? Br J Anaesth 2020; 125:e391-e394. [PMID: 32792138 PMCID: PMC7377768 DOI: 10.1016/j.bja.2020.07.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 12/22/2022] Open
Affiliation(s)
- Dylan T Finnerty
- Division of Anaesthesiology, Mater Misericordiae University Hospital, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland; EU COST Action 15204, Euro-Periscope, Dublin, Ireland.
| | - Donal J Buggy
- Division of Anaesthesiology, Mater Misericordiae University Hospital, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland; EU COST Action 15204, Euro-Periscope, Dublin, Ireland; Outcomes Research, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
270
|
Ramirez DC, Gomez Mejiba SE. Pulmonary Neutrophilic Inflammation and Noncommunicable Diseases: Pathophysiology, Redox Mechanisms, Biomarkers, and Therapeutics. Antioxid Redox Signal 2020; 33:211-227. [PMID: 32319787 DOI: 10.1089/ars.2020.8098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significance: Pulmonary neurophilic inflammation (PNI) is the homing and activation of neutrophil with damage to the microvasculature. This process is involved in pulmonary damage in patients exposed to airborne pollutants (exogenous stressors) and also to systemic inflammation/oxidative stress (endogenous stressors) associated with noncommunicable diseases (NCDs). Recent Advances: PNI is an important trigger of the early onset and progression of NCD in susceptible patients exposed to airborne pollutants. Irritation of the lung microvasculature by exogenous and endogenous stressors causes PNI. Circulating endogenous stressors in NCD can cause PNI. Critical Issues: Air pollution-triggered PNI causes increased circulating endogenous stressors that can trigger NCD in susceptible patients. Systemic inflammation/oxidative stress associated with NCD can cause PNI. Inflammation/end-oxidation products of macromolecules are also potential biomarkers and therapeutic targets for NCD-triggered PNI- and PNI-triggered NCD. Future Directions: Understanding the molecular mechanism of PNI triggered by exogenous or endogenous stressors will help explain the early onset of NCD in susceptible patients exposed to air pollution. It can also help undercover biomarkers and mechanism-based therapeutic targets in air pollutant-triggered PNI, PNI-triggered NCD, and NCD-triggered PNI.
Collapse
Affiliation(s)
- Dario C Ramirez
- Laboratory of Experimental and Translational Medicine, IMIBIO-SL, CCT-San Luis, CONICET, School of Chemistry, Biochemistry and Pharmacy, National University of San Luis, San Luis, Argentina
| | - Sandra E Gomez Mejiba
- Laboratory of Experimental Therapeutics and Nutrition, IMIBIO-SL, CCT-San Luis, CONICET, School of Health Sciences, National University of San Luis, San Luis, Argentina
| |
Collapse
|
271
|
Overmyer KA, Shishkova E, Miller IJ, Balnis J, Bernstein MN, Peters-Clarke TM, Meyer JG, Quan Q, Muehlbauer LK, Trujillo EA, He Y, Chopra A, Chieng HC, Tiwari A, Judson MA, Paulson B, Brademan DR, Zhu Y, Serrano LR, Linke V, Drake LA, Adam AP, Schwartz BS, Singer HA, Swanson S, Mosher DF, Stewart R, Coon JJ, Jaitovich A. Large-scale Multi-omic Analysis of COVID-19 Severity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.07.17.20156513. [PMID: 32743614 PMCID: PMC7388490 DOI: 10.1101/2020.07.17.20156513] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We performed RNA-Seq and high-resolution mass spectrometry on 128 blood samples from COVID-19 positive and negative patients with diverse disease severities. Over 17,000 transcripts, proteins, metabolites, and lipids were quantified and associated with clinical outcomes in a curated relational database, uniquely enabling systems analysis and cross-ome correlations to molecules and patient prognoses. We mapped 219 molecular features with high significance to COVID-19 status and severity, many involved in complement activation, dysregulated lipid transport, and neutrophil activation. We identified sets of covarying molecules, e.g., protein gelsolin and metabolite citrate or plasmalogens and apolipoproteins, offering pathophysiological insights and therapeutic suggestions. The observed dysregulation of platelet function, blood coagulation, acute phase response, and endotheliopathy further illuminated the unique COVID-19 phenotype. We present a web-based tool (covid-omics.app) enabling interactive exploration of our compendium and illustrate its utility through a comparative analysis with published data and a machine learning approach for prediction of COVID-19 severity.
Collapse
Affiliation(s)
- Katherine A. Overmyer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Evgenia Shishkova
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ian J. Miller
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Joseph Balnis
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | | | - Trenton M. Peters-Clarke
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Jesse G. Meyer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Qiuwen Quan
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Laura K. Muehlbauer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Edna A. Trujillo
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Yuchen He
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Amit Chopra
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| | - Hau C. Chieng
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| | - Anupama Tiwari
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
- Division of Sleep Medicine, Albany Medical Center, Albany, NY, USA
| | - Marc A. Judson
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| | - Brett Paulson
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Dain R. Brademan
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Yunyun Zhu
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Lia R. Serrano
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Vanessa Linke
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Lisa A. Drake
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Alejandro P. Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
- Department of Ophthalmology, Albany Medical College, Albany, NY, USA
| | | | - Harold A. Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Scott Swanson
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Deane F. Mosher
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Joshua J. Coon
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Morgridge Institute for Research, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
272
|
Filep JG, Ariel A. Neutrophil heterogeneity and fate in inflamed tissues: implications for the resolution of inflammation. Am J Physiol Cell Physiol 2020; 319:C510-C532. [PMID: 32667864 DOI: 10.1152/ajpcell.00181.2020] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neutrophils are polymorphonuclear leukocytes that play a central role in host defense against infection and tissue injury. They are rapidly recruited to the inflamed site and execute a variety of functions to clear invading pathogens and damaged cells. However, many of their defense mechanisms are capable of inflicting collateral tissue damage. Neutrophil-driven inflammation is a unifying mechanism underlying many common diseases. Efficient removal of neutrophils from inflammatory loci is critical for timely resolution of inflammation and return to homeostasis. Accumulating evidence challenges the classical view that neutrophils represent a homogeneous population and that halting neutrophil influx is sufficient to explain their rapid decline within inflamed loci during the resolution of protective inflammation. Hence, understanding the mechanisms that govern neutrophil functions and their removal from the inflammatory locus is critical for minimizing damage to the surrounding tissue and for return to homeostasis. In this review, we briefly address recent advances in characterizing neutrophil phenotypic and functional heterogeneity and the molecular mechanisms that determine the fate of neutrophils within inflammatory loci and the outcome of the inflammatory response. We also discuss how these mechanisms may be harnessed as potential therapeutic targets to facilitate resolution of inflammation.
Collapse
Affiliation(s)
- János G Filep
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - Amiram Ariel
- Departmentof Biology and Human Biology, University of Haifa, Haifa, Israel
| |
Collapse
|
273
|
Schönrich G, Raftery MJ, Samstag Y. Devilishly radical NETwork in COVID-19: Oxidative stress, neutrophil extracellular traps (NETs), and T cell suppression. Adv Biol Regul 2020; 77:100741. [PMID: 32773102 PMCID: PMC7334659 DOI: 10.1016/j.jbior.2020.100741] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Pandemic coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and poses an unprecedented challenge to healthcare systems due to the lack of a vaccine and specific treatment options. Accordingly, there is an urgent need to understand precisely the pathogenic mechanisms underlying this multifaceted disease. There is increasing evidence that the immune system reacts insufficiently to SARS-CoV-2 and thus contributes to organ damage and to lethality. In this review, we suggest that the overwhelming production of reactive oxygen species (ROS) resulting in oxidative stress is a major cause of local or systemic tissue damage that leads to severe COVID-19. It increases the formation of neutrophil extracellular traps (NETs) and suppresses the adaptive arm of the immune system, i.e. T cells that are necessary to kill virus-infected cells. This creates a vicious cycle that prevents a specific immune response against SARS-CoV-2. The key role of oxidative stress in the pathogenesis of severe COVID-19 implies that therapeutic counterbalancing of ROS by antioxidants such as vitamin C or NAC and/or by antagonizing ROS production by cells of the mononuclear phagocyte system (MPS) and neutrophil granulocytes and/or by blocking of TNF-α can prevent COVID-19 from becoming severe. Controlled clinical trials and preclinical models of COVID-19 are needed to evaluate this hypothesis.
Collapse
Affiliation(s)
- Günther Schönrich
- Institute of Virology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany.
| | - Martin J Raftery
- Institute of Virology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Yvonne Samstag
- Section Molecular Immunology, Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
274
|
Whyte CS, Morrow GB, Mitchell JL, Chowdary P, Mutch NJ. Fibrinolytic abnormalities in acute respiratory distress syndrome (ARDS) and versatility of thrombolytic drugs to treat COVID-19. J Thromb Haemost 2020; 18:1548-1555. [PMID: 32329246 PMCID: PMC7264738 DOI: 10.1111/jth.14872] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 01/10/2023]
Abstract
The global pandemic of coronavirus disease 2019 (COVID-19) is associated with the development of acute respiratory distress syndrome (ARDS), which requires ventilation in critically ill patients. The pathophysiology of ARDS results from acute inflammation within the alveolar space and prevention of normal gas exchange. The increase in proinflammatory cytokines within the lung leads to recruitment of leukocytes, further propagating the local inflammatory response. A consistent finding in ARDS is the deposition of fibrin in the air spaces and lung parenchyma. COVID-19 patients show elevated D-dimers and fibrinogen. Fibrin deposits are found in the lungs of patients due to the dysregulation of the coagulation and fibrinolytic systems. Tissue factor (TF) is exposed on damaged alveolar endothelial cells and on the surface of leukocytes promoting fibrin deposition, while significantly elevated levels of plasminogen activator inhibitor 1 (PAI-1) from lung epithelium and endothelial cells create a hypofibrinolytic state. Prophylaxis treatment of COVID-19 patients with low molecular weight heparin (LMWH) is important to limit coagulopathy. However, to degrade pre-existing fibrin in the lung it is essential to promote local fibrinolysis. In this review, we discuss the repurposing of fibrinolytic drugs, namely tissue-type plasminogen activator (tPA), to treat COVID-19 associated ARDS. tPA is an approved intravenous thrombolytic treatment, and the nebulizer form has been shown to be effective in plastic bronchitis and is currently in Phase II clinical trial. Nebulizer plasminogen activators may provide a targeted approach in COVID-19 patients to degrade fibrin and improving oxygenation in critically ill patients.
Collapse
Affiliation(s)
- Claire S Whyte
- Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Aberdeen, UK
| | - Gael B Morrow
- Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Aberdeen, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Joanne L Mitchell
- Institute of Cardiovascular and Metabolic Sciences, School of Biological Sciences, University of Reading, Reading, UK
| | - Pratima Chowdary
- Katharine Dormandy Haemophilia and Thrombosis Centre Royal Free Hospital, London, UK
| | - Nicola J Mutch
- Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences and Nutrition, Aberdeen, UK
| |
Collapse
|
275
|
Jiao Y, Li W, Wang W, Tong X, Xia R, Fan J, Du J, Zhang C, Shi X. Platelet-derived exosomes promote neutrophil extracellular trap formation during septic shock. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:380. [PMID: 32600436 PMCID: PMC7322900 DOI: 10.1186/s13054-020-03082-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 06/09/2020] [Indexed: 12/16/2022]
Abstract
Background Platelets have been demonstrated to be potent activators of neutrophil extracellular trap (NET) formation during sepsis. However, the mediators and molecular pathways involved in human platelet-mediated NET generation remain poorly defined. Circulating plasma exosomes mostly originating from platelets may induce vascular apoptosis and myocardial dysfunction during sepsis; however, their role in NET formation remains unclear. This study aimed to detect whether platelet-derived exosomes could promote NET formation during septic shock and determine the potential mechanisms involved. Methods Polymorphonuclear neutrophils (PMNs) were cocultured with exosomes isolated from the plasma of healthy controls and septic shock patients or the supernatant of human platelets stimulated ex vivo with phosphate buffer saline (PBS) or lipopolysaccharide (LPS). A lethal cecal ligation and puncture (CLP) mouse model was used to mimic sepsis in vivo; then, NET formation and molecular pathways were detected. Results NET components (dsDNA and MPO-DNA complexes) were significantly increased in response to treatment with septic shock patient-derived exosomes and correlated positively with disease severity and outcome. In the animal CLP model, platelet depletion reduced plasma exosome concentration, NET formation, and lung injury. Mechanistic studies demonstrated that exosomal high-mobility group protein 1 (HMGB1) and/or miR-15b-5p and miR-378a-3p induced NET formation through the Akt/mTOR autophagy pathway. Furthermore, the results suggested that IκB kinase (IKK) controls platelet-derived exosome secretion in septic shock. Conclusions Platelet-derived exosomes promote excessive NET formation in sepsis and subsequent organ injury. This finding suggests a previously unidentified role of platelet-derived exosomes in sepsis and may lead to new therapeutic approaches.
Collapse
Affiliation(s)
- Yang Jiao
- Department of Anesthesiology and Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Weiwei Li
- Department of Anesthesiology and Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Wei Wang
- Department of Anesthesiology and Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xingyu Tong
- Department of Anesthesiology and Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Ran Xia
- Department of Anesthesiology and Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Jianer Du
- Department of Anesthesiology and Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Chengmi Zhang
- Department of Anesthesiology and Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Xueyin Shi
- Department of Anesthesiology and Intensive Care Unit, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
276
|
Ross JT, Nesseler N, Leligdowicz A, Zemans RL, Mahida RY, Minus E, Langelier C, Gotts JE, Matthay MA. The ex vivo perfused human lung is resistant to injury by high-dose S. pneumoniae bacteremia. Am J Physiol Lung Cell Mol Physiol 2020; 319:L218-L227. [PMID: 32519893 DOI: 10.1152/ajplung.00053.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Few patients with bacteremia from a nonpulmonary source develop acute respiratory distress syndrome (ARDS). However, the mechanisms that protect the lung from injury in bacteremia have not been identified. We simulated bacteremia by adding Streptococcus pneumoniae to the perfusate of the ex vivo perfused human lung model. In contrast to a pneumonia model in which bacteria were instilled into the distal air spaces of one lobe, injection of high doses of S. pneumoniae into the perfusate was not associated with alveolar epithelial injury as demonstrated by low protein permeability of the alveolar epithelium, intact alveolar fluid clearance, and the absence of alveolar edema. Unexpectedly, the ex vivo human lung rapidly cleared large quantities of S. pneumoniae even though the perfusate had very few intravascular phagocytes and lacked immunoglobulins or complement. The bacteria were cleared in part by the small number of neutrophils in the perfusate, alveolar macrophages in the airspaces, and probably by interstitial pathways. Together, these findings identify one mechanism by which the lung and the alveolar epithelium are protected from injury in bacteremia.
Collapse
Affiliation(s)
- James T Ross
- Department of Surgery, University of California San Francisco, San Francisco, California
| | - Nicolas Nesseler
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California.,Department of Anesthesia and Critical Care, Pontchaillou, University Hospital of Rennes, Rennes, France.,University Rennes, CHU de Rennes, Inra, Inserm, Institut NUMECAN-UMR_A 1341, UMR_S 1241, Rennes, France.,University Rennes, CHU Rennes, Inserm, CIC 1414 (Centre d'Investigation Clinique de Rennes), Rennes, France
| | - Aleksandra Leligdowicz
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, California.,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Rachel L Zemans
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Rahul Y Mahida
- Birmingham Acute Care Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham, United Kingdom
| | | | - Chaz Langelier
- Division of Infectious Diseases, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Jeffrey E Gotts
- Department of Medicine, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, California
| |
Collapse
|
277
|
Zuo Y, Yalavarthi S, Shi H, Gockman K, Zuo M, Madison JA, Blair C, Weber A, Barnes BJ, Egeblad M, Woods RJ, Kanthi Y, Knight JS. Neutrophil extracellular traps in COVID-19. JCI Insight 2020; 5:138999. [PMID: 32329756 PMCID: PMC7308057 DOI: 10.1172/jci.insight.138999] [Citation(s) in RCA: 778] [Impact Index Per Article: 155.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023] Open
Abstract
In severe cases of coronavirus disease 2019 (COVID-19), viral pneumonia progresses to respiratory failure. Neutrophil extracellular traps (NETs) are extracellular webs of chromatin, microbicidal proteins, and oxidant enzymes that are released by neutrophils to contain infections. However, when not properly regulated, NETs have the potential to propagate inflammation and microvascular thrombosis - including in the lungs of patients with acute respiratory distress syndrome. We now report that sera from patients with COVID-19 have elevated levels of cell-free DNA, myeloperoxidase-DNA (MPO-DNA), and citrullinated histone H3 (Cit-H3); the latter 2 are specific markers of NETs. Highlighting the potential clinical relevance of these findings, cell-free DNA strongly correlated with acute-phase reactants, including C-reactive protein, D-dimer, and lactate dehydrogenase, as well as absolute neutrophil count. MPO-DNA associated with both cell-free DNA and absolute neutrophil count, while Cit-H3 correlated with platelet levels. Importantly, both cell-free DNA and MPO-DNA were higher in hospitalized patients receiving mechanical ventilation as compared with hospitalized patients breathing room air. Finally, sera from individuals with COVID-19 triggered NET release from control neutrophils in vitro. Future studies should investigate the predictive power of circulating NETs in longitudinal cohorts and determine the extent to which NETs may be novel therapeutic targets in severe COVID-19.
Collapse
Affiliation(s)
- Yu Zuo
- 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
| | - Hui Shi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Division of Rheumatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kelsey Gockman
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Melanie Zuo
- Division of Geriatric and Palliative Medicine and
| | - Jacqueline A. Madison
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Blair
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrew Weber
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Northwell Health, New York, New York, USA
| | - Betsy J. Barnes
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Departments of Molecular Medicine and Pediatrics, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Robert J. Woods
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yogendra Kanthi
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Division of Cardiology, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
278
|
Barnes BJ, Adrover JM, Baxter-Stoltzfus A, Borczuk A, Cools-Lartigue J, Crawford JM, Daßler-Plenker J, Guerci P, Huynh C, Knight JS, Loda M, Looney MR, McAllister F, Rayes R, Renaud S, Rousseau S, Salvatore S, Schwartz RE, Spicer JD, Yost CC, Weber A, Zuo Y, Egeblad M. Targeting potential drivers of COVID-19: Neutrophil extracellular traps. J Exp Med 2020; 217:e20200652. [PMID: 32302401 PMCID: PMC7161085 DOI: 10.1084/jem.20200652] [Citation(s) in RCA: 1096] [Impact Index Per Article: 219.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/09/2020] [Accepted: 04/13/2020] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a novel, viral-induced respiratory disease that in ∼10-15% of patients progresses to acute respiratory distress syndrome (ARDS) triggered by a cytokine storm. In this Perspective, autopsy results and literature are presented supporting the hypothesis that a little known yet powerful function of neutrophils-the ability to form neutrophil extracellular traps (NETs)-may contribute to organ damage and mortality in COVID-19. We show lung infiltration of neutrophils in an autopsy specimen from a patient who succumbed to COVID-19. We discuss prior reports linking aberrant NET formation to pulmonary diseases, thrombosis, mucous secretions in the airways, and cytokine production. If our hypothesis is correct, targeting NETs directly and/or indirectly with existing drugs may reduce the clinical severity of COVID-19.
Collapse
Affiliation(s)
- Betsy J. Barnes
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research & Departments of Molecular Medicine and Pediatrics, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY
| | | | | | - Alain Borczuk
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | | | - James M. Crawford
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, East Garden City, NY
| | | | | | - Caroline Huynh
- Division of Thoracic and Upper GI Surgery, Department of Surgery, Montreal, Canada
- Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Roni Rayes
- Division of Thoracic and Upper GI Surgery, Department of Surgery, Montreal, Canada
- Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | | | - Simon Rousseau
- Department of Medicine, McGill University & The Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Steven Salvatore
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Robert E. Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Jonathan D. Spicer
- Division of Thoracic and Upper GI Surgery, Department of Surgery, Montreal, Canada
- Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Christian C. Yost
- Department of Pediatrics, Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, UT
| | - Andrew Weber
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Northwell Health, Manhasset, NY
| | - Yu Zuo
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | | |
Collapse
|
279
|
Lv D, Xu Y, Cheng H, Ke Y, Zhang X, Ying K. A novel cell-based assay for dynamically detecting neutrophil extracellular traps-induced lung epithelial injuries. Exp Cell Res 2020; 394:112101. [PMID: 32474064 PMCID: PMC7256615 DOI: 10.1016/j.yexcr.2020.112101] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
Acute lung injury (ALI) and its more severe form, acute respiratory distress syndrome (ARDS) are common lung disorders characterized by alveolar-capillary barrier disruption and dyspnea, which can cause substantial morbidity and mortality. Currently, a cluster of acute respiratory illnesses, known as novel coronavirus (2019-nCoV)-infected pneumonia (NCIP), which allegedly originally occurred in Wuhan, China, has increased rapidly worldwide. The critically ill patients with ARDS have high mortality in subjects with comorbidities. Previously, the excessive recruitment and activation of neutrophils (polymorphonuclear leukocytes [PMNs]), accompanied by neutrophil extracellular traps (NETs) formation were reported being implicated in the pathogenesis of ALI/ARDS. However, the direct visualization of lung epithelial injuries caused by NETs, and the qualitative and quantitative evaluations of this damage are still lacking. Additionally, those already reported methods are limited for their neglect of the pathological role exerted by NETs and focusing only on the morphological features of NETosis. Therefore, we established a cell-based assay for detecting NETs during lung epithelial cells-neutrophils co-culture using the xCELLigence system, a recognized real-time, dynamic, label-free, sensitive, and high-throughput apparatus. Our results demonstrated that lung epithelial injuries, reflected by declines in cell index (CI) values, could be induced by lipopolysaccharide (LPS)-activated PMNs, or NETs in a time and dose-dependent manner. NETs generation was verified to be the major contributor to the cytotoxicity of activated PMNs; protein components of NETs were the prevailing cytotoxic mediators. Moreover, this cell-based assay identified that PMNs from severe pneumonia patients had a high NETs formative potential. Additionally, acetylsalicylic acid (ASA) and acetaminophen (APAP) were discovered alleviating NETs formation. Thus, this study not only presents a new methodology for detecting the pathophysiologic role of NETs but also lays down a foundation for exploring therapeutic interventions in an effort to cure ALI/ARDS in the clinical setting of severe pneumonia, including the emerging of NCIP. A real-time, dynamical and label-free assay for detecting NETs is established using the xCELLigence system. This establishment relies on the co-culture of lung epithelia and neutrophils, focusing on evaluating NETs’ effects. This cell-based assay has feasibility and practicality in clinical applications. This methodology builds a solid foundation for exploring therapies for ALI/ARDS, including the emerging NCIP.
Collapse
Affiliation(s)
- Dandan Lv
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
| | - Yiming Xu
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Yuehai Ke
- Department of Pathology and Pathophysiology and Department of Respiratory Medicine at Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology and Department of Respiratory Medicine at Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
| | - Kejing Ying
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China.
| |
Collapse
|
280
|
Fox SE, Akmatbekov A, Harbert JL, Li G, Quincy Brown J, Vander Heide RS. Pulmonary and cardiac pathology in African American patients with COVID-19: an autopsy series from New Orleans. THE LANCET RESPIRATORY MEDICINE 2020; 8:681-686. [PMID: 32473124 PMCID: PMC7255143 DOI: 10.1016/s2213-2600(20)30243-5] [Citation(s) in RCA: 895] [Impact Index Per Article: 179.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/07/2020] [Accepted: 05/12/2020] [Indexed: 12/21/2022]
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spread rapidly across the USA, causing extensive morbidity and mortality, particularly in the African American community. Autopsy can considerably contribute to our understanding of many disease processes and could provide crucial information to guide management of patients with coronavirus disease 2019 (COVID-19). We report on the relevant cardiopulmonary findings in, to our knowledge, the first autopsy series of ten African American decedents, with the cause of death attributed to COVID-19. Methods Autopsies were performed on ten African American decedents aged 44–78 years with cause of death attributed to COVID-19, reflective of the dominant demographic of deaths following COVID-19 diagnosis in New Orleans. Autopsies were done with consent of the decedents' next of kin. Pulmonary and cardiac features were examined, with relevant immunostains to characterise the inflammatory response, and RNA labelling and electron microscopy on representative sections. Findings Important findings include the presence of thrombosis and microangiopathy in the small vessels and capillaries of the lungs, with associated haemorrhage, that significantly contributed to death. Features of diffuse alveolar damage, including hyaline membranes, were present, even in patients who had not been ventilated. Cardiac findings included individual cell necrosis without lymphocytic myocarditis. There was no evidence of secondary pulmonary infection by microorganisms. Interpretation We identify key pathological states, including thrombotic and microangiopathic pathology in the lungs, that contributed to death in patients with severe COVID-19 and decompensation in this demographic. Management of these patients should include treatment to target these pathological mechanisms. Funding None.
Collapse
Affiliation(s)
- Sharon E Fox
- Department of Pathology, Louisiana State University Health Sciences Center, New Orleans, LA, USA; Pathology and Laboratory Medicine Service, Southeast Louisiana Veterans Healthcare System, New Orleans, LA, USA.
| | - Aibek Akmatbekov
- Department of Pathology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Jack L Harbert
- Department of Pathology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Guang Li
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - J Quincy Brown
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Richard S Vander Heide
- Department of Pathology, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| |
Collapse
|
281
|
Zuo Y, Yalavarthi S, Shi H, Gockman K, Zuo M, Madison JA, Blair C, Weber A, Barnes BJ, Egeblad M, Woods RJ, Kanthi Y, Knight JS. Neutrophil extracellular traps (NETs) as markers of disease severity in COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020. [PMID: 32511633 DOI: 10.1101/2020.04.09.20059626] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In severe cases of coronavirus disease 2019 (COVID-19), viral pneumonia progresses to respiratory failure. Neutrophil extracellular traps (NETs) are extracellular webs of chromatin, microbicidal proteins, and oxidant enzymes that are released by neutrophils to contain infections. However, when not properly regulated, NETs have potential to propagate inflammation and microvascular thrombosis, including in the lungs of patients with acute respiratory distress syndrome. While elevated levels of blood neutrophils predict worse outcomes in COVID-19, the role of NETs has not been investigated. We now report that sera from patients with COVID-19 (n=50 patients, n=84 samples) have elevated levels of cell-free DNA, myeloperoxidase(MPO)-DNA, and citrullinated histone H3 (Cit-H3); the latter two are highly specific markers of NETs. Highlighting the potential clinical relevance of these findings, cell-free DNA strongly correlated with acute phase reactants including C-reactive protein, D-dimer, and lactate dehydrogenase, as well as absolute neutrophil count. MPO-DNA associated with both cell-free DNA and absolute neutrophil count, while Cit-H3 correlated with platelet levels. Importantly, both cell-free DNA and MPO-DNA were higher in hospitalized patients receiving mechanical ventilation as compared with hospitalized patients breathing room air. Finally, sera from individuals with COVID-19 triggered NET release from control neutrophils in vitro. In summary, these data reveal high levels of NETs in many patients with COVID-19, where they may contribute to cytokine release and respiratory failure. Future studies should investigate the predictive power of circulating NETs in longitudinal cohorts, and determine the extent to which NETs may be novel therapeutic targets in severe COVID-19.
Collapse
|
282
|
Lawrence SM, Corriden R, Nizet V. How Neutrophils Meet Their End. Trends Immunol 2020; 41:531-544. [PMID: 32303452 DOI: 10.1016/j.it.2020.03.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/10/2020] [Accepted: 03/18/2020] [Indexed: 12/28/2022]
Abstract
Neutrophil death can transpire via diverse pathways and is regulated by interactions with commensal and pathogenic microorganisms, environmental exposures, and cell age. At steady state, neutrophil turnover and replenishment are continually maintained via a delicate balance between host-mediated responses and microbial forces. Disruptions in this equilibrium directly impact neutrophil numbers in circulation, cell trafficking, antimicrobial defenses, and host well-being. How neutrophils meet their end is physiologically important and can result in different immunologic consequences. Whereas nonlytic forms of neutrophil death typically elicit anti-inflammatory responses and promote healing, pathways ending with cell membrane rupture may incite deleterious proinflammatory responses, which can exacerbate local tissue injury, lead to chronic inflammation, or precipitate autoimmunity. This review seeks to provide a contemporary analysis of mechanisms of neutrophil death.
Collapse
Affiliation(s)
- Shelley M Lawrence
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, College of Medicine, University of California, San Diego, CA, USA; Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, College of Medicine, University of California, San Diego, CA, USA.
| | - Ross Corriden
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, College of Medicine, University of California, San Diego, CA, USA; Department of Pharmacology, University of California, San Diego, CA, USA
| | - Victor Nizet
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, College of Medicine, University of California, San Diego, CA, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, USA
| |
Collapse
|
283
|
Ojima M, Yamamoto N, Hirose T, Hamaguchi S, Tasaki O, Kojima T, Tomono K, Ogura H, Shimazu T. Serial change of neutrophil extracellular traps in tracheal aspirate of patients with acute respiratory distress syndrome: report of three cases. J Intensive Care 2020; 8:25. [PMID: 32308985 PMCID: PMC7149861 DOI: 10.1186/s40560-020-00444-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/01/2020] [Indexed: 01/09/2023] Open
Abstract
Background Neutrophil extracellular traps (NETs) are fibrous structures released from activated neutrophils. NET formation has been reported to be associated with acute respiratory distress syndrome (ARDS). However, there are no reports dealing with serial changes of NET formation in tracheal aspirate of ARDS patients. Case presentation We report three cases of ARDS. Case 1 is a 69-year-old man with necrotizing fasciitis of the buttocks, case 2 is a 49-year-old woman with extensive burns (80% of total body surface), and case 3 is a 73-year-old woman with severe bacterial pneumonia. We found abundant expression of citrullinated histone H3 (Cit-H3) and the formation of NETs at the onset of ARDS in all cases. The amounts of Cit-H3 and NETs decreased with the amelioration of respiratory failure in cases 1 and 2. In case 2, the amounts of Cit-H3 and NETs increased with aggravation of infection and respiratory status. In case 3, the abundant expression of Cit-H3 and NETs persisted; the patient did not recover from ARDS and eventually died. Cit-H3 and NETs were found in tracheal aspirates even if the patients had no direct injury to the lung as in cases 1 and 2. Conclusions In these three cases, the formation of NETs was observed in tracheal aspirate of patients with ARDS by either direct or indirect insults to the lung. The amount of NET formation changed dynamically over the clinical course of each patient.
Collapse
Affiliation(s)
- Masahiro Ojima
- 1Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Norihisa Yamamoto
- 2Division of Infection Control and Prevention, Osaka University Hospital, Suita, Osaka, Japan
| | - Tomoya Hirose
- 1Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shigeto Hamaguchi
- 2Division of Infection Control and Prevention, Osaka University Hospital, Suita, Osaka, Japan
| | - Osamu Tasaki
- 3Acute and Critical Care Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Takashi Kojima
- 4Central Laboratory for Clinical Investigation, Osaka University Hospital, Suita, Osaka, Japan
| | - Kazunori Tomono
- 2Division of Infection Control and Prevention, Osaka University Hospital, Suita, Osaka, Japan
| | - Hiroshi Ogura
- 1Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takeshi Shimazu
- 1Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
284
|
Lehman HK, Segal BH. The role of neutrophils in host defense and disease. J Allergy Clin Immunol 2020; 145:1535-1544. [PMID: 32283205 DOI: 10.1016/j.jaci.2020.02.038] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/15/2020] [Accepted: 02/05/2020] [Indexed: 12/21/2022]
Abstract
Neutrophils, the most abundant circulating leukocyte, are critical for host defense. Granulopoiesis is under the control of transcriptional factors and culminates in mature neutrophils with a broad armamentarium of antimicrobial pathways. These pathways include nicotinamide adenine dinucleotide phosphate oxidase, which generates microbicidal reactive oxidants, and nonoxidant pathways that target microbes through several mechanisms. Activated neutrophils can cause or worsen tissue injury, underscoring the need for calibration of activation and resolution of inflammation when infection has been cleared. Acquired neutrophil disorders are typically caused by cytotoxic chemotherapy or immunosuppressive agents. Primary neutrophil disorders typically result from disabling mutations of individual genes that result in impaired neutrophil number or function, and provide insight into basic mechanisms of neutrophil biology. Neutrophils can also be activated by noninfectious causes, including trauma and cellular injury, and can have off-target effects in which pathways that typically defend against infection exacerbate injury and disease. These off-target effects include acute organ injury, autoimmunity, and variable effects on the tumor microenvironment that can limit or worsen tumor progression. A greater understanding of neutrophil plasticity in these conditions is likely to pave the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Heather K Lehman
- Division of Allergy/Immunology & Rheumatology, Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Brahm H Segal
- Roswell Park Comprehensive Cancer Center, University at Buffalo, Buffalo, NY.
| |
Collapse
|
285
|
Matuszak J, Tabuchi A, Kuebler WM. Ventilation and Perfusion at the Alveolar Level: Insights From Lung Intravital Microscopy. Front Physiol 2020; 11:291. [PMID: 32308629 PMCID: PMC7145899 DOI: 10.3389/fphys.2020.00291] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/16/2020] [Indexed: 01/13/2023] Open
Abstract
Intravital microscopy (IVM) offers unique possibilities for the observation of biological processes and disease related mechanisms in vivo. Especially for anatomically complex and dynamic organs such as the lung and its main functional unit, the alveolus, IVM provides exclusive advantages in terms of spatial and temporal resolution. By the use of lung windows, which have advanced and improved over time, direct access to the lung surface is provided. In this review we will discuss two main topics, namely alveolar dynamics and perfusion from the perspective of IVM-based studies. Of special interest are unanswered questions regarding alveolar dynamics such as: What are physiologic alveolar dynamics? How do these dynamics change under pathologic conditions and how do those changes contribute to ventilator-induced lung injury? How can alveolar dynamics be targeted in a beneficial way? With respect to alveolar perfusion IVM has propelled our understanding of the pulmonary microcirculation and its perfusion, as well as pulmonary vasoreactivity, permeability and immunological aspects. Whereas the general mechanism behind these processes are understood, we still lack a proper understanding of the complex, multidimensional interplay between alveolar ventilation and microvascular perfusion, capillary recruitment, or vascular immune responses under physiologic and pathologic conditions. These are only part of the unanswered questions and problems, which we still have to overcome. IVM as the tool of choice might allow us to answer part of these questions within the next years or decades. As every method, IVM has advantages as well as limitations, which have to be taken into account for data analysis and interpretation, which will be addressed in this review.
Collapse
Affiliation(s)
- Jasmin Matuszak
- Institute of Physiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Arata Tabuchi
- The Keenan Research Centre for Biomedical Science at St. Michael’s, Toronto, ON, Canada
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Departments of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
286
|
Hu L, Zhao T, Sun Y, Chen Y, Bai K, Xu F. Bioinformatic identification of hub genes and key pathways in neutrophils of patients with acute respiratory distress syndrome. Medicine (Baltimore) 2020; 99:e19820. [PMID: 32282748 PMCID: PMC7220668 DOI: 10.1097/md.0000000000019820] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized as a neutrophil-dominant disorder without effective pharmacological interventions. Knowledge of neutrophils in ARDS patients at the transcriptome level is still limited. We aimed to identify the hub genes and key pathways in neutrophils of patients with ARDS. The transcriptional profiles of neutrophils from ARDS patients and healthy volunteers were obtained from the GSE76293 dataset. The differentially expressed genes (DEGs) between ARDS and healthy samples were screened using the limma R package. Subsequently, functional and pathway enrichment analyses were performed based on the database for annotation, visualization, and integrated discovery (DAVID). The construction of a protein-protein interaction network was carried out using the search tool for the retrieval of interacting genes (STRING) database and the network was visualized by Cytoscape software. The Cytoscape plugins cytoHubba and MCODE were used to identify hub genes and significant modules. Finally, 136 upregulated genes and 95 downregulated genes were identified. Gene ontology analyses revealed MHC class II plays a major role in functional annotations. SLC11A1, ARG1, CHI3L1, HP, LCN2, and MMP8 were identified as hub genes, and they were all involved in the neutrophil degranulation pathway. The MAPK and neutrophil degranulation pathways in neutrophils were considered as key pathways in the pathogenesis of ARDS. This study improves our understanding of the biological characteristics of neutrophils and the mechanisms underlying ARDS, and key pathways and hub genes identified in this work can serve as targets for novel ARDS treatment strategies.
Collapse
Affiliation(s)
- Lan Hu
- Department of Intensive Care Unit, Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation base of Child development and Critical Disorders; Children's Hospital of Chongqing Medical University
- Chongqing Key Laboratory of Pediatrics
- Department of Outpatient, Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Tianxin Zhao
- Department of Intensive Care Unit, Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation base of Child development and Critical Disorders; Children's Hospital of Chongqing Medical University
- Chongqing Key Laboratory of Pediatrics
| | - Yuelin Sun
- Department of Intensive Care Unit, Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation base of Child development and Critical Disorders; Children's Hospital of Chongqing Medical University
- Chongqing Key Laboratory of Pediatrics
| | - Yingfu Chen
- Department of Intensive Care Unit, Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation base of Child development and Critical Disorders; Children's Hospital of Chongqing Medical University
- Chongqing Key Laboratory of Pediatrics
| | - Ke Bai
- Department of Intensive Care Unit, Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation base of Child development and Critical Disorders; Children's Hospital of Chongqing Medical University
- Chongqing Key Laboratory of Pediatrics
| | - Feng Xu
- Department of Intensive Care Unit, Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders (Chongqing); China International Science and Technology Cooperation base of Child development and Critical Disorders; Children's Hospital of Chongqing Medical University
- Chongqing Key Laboratory of Pediatrics
| |
Collapse
|
287
|
Ortiz-Muñoz G, Yu MA, Lefrançais E, Mallavia B, Valet C, Tian JJ, Ranucci S, Wang KM, Liu Z, Kwaan N, Dawson D, Kleinhenz ME, Khasawneh FT, Haggie PM, Verkman AS, Looney MR. Cystic fibrosis transmembrane conductance regulator dysfunction in platelets drives lung hyperinflammation. J Clin Invest 2020; 130:2041-2053. [PMID: 31961827 PMCID: PMC7108932 DOI: 10.1172/jci129635] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 01/14/2020] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF) lung disease is characterized by an inflammatory response that can lead to terminal respiratory failure. The cystic fibrosis transmembrane conductance regulator (CFTR) is mutated in CF, and we hypothesized that dysfunctional CFTR in platelets, which are key participants in immune responses, is a central determinant of CF inflammation. We found that deletion of CFTR in platelets produced exaggerated acute lung inflammation and platelet activation after intratracheal LPS or Pseudomonas aeruginosa challenge. CFTR loss of function in mouse or human platelets resulted in agonist-induced hyperactivation and increased calcium entry into platelets. Inhibition of the transient receptor potential cation channel 6 (TRPC6) reduced platelet activation and calcium flux, and reduced lung injury in CF mice after intratracheal LPS or Pseudomonas aeruginosa challenge. CF subjects receiving CFTR modulator therapy showed partial restoration of CFTR function in platelets, which may be a convenient approach to monitoring biological responses to CFTR modulators. We conclude that CFTR dysfunction in platelets produces aberrant TRPC6-dependent platelet activation, which is a major driver of CF lung inflammation and impaired bacterial clearance. Platelets and TRPC6 are what we believe to be novel therapeutic targets in the treatment of CF lung disease.
Collapse
Affiliation(s)
| | - Michelle A. Yu
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Emma Lefrançais
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Beñat Mallavia
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Colin Valet
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Serena Ranucci
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Kristin M. Wang
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Zhe Liu
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Nicholas Kwaan
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Diana Dawson
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Fadi T. Khasawneh
- School of Pharmacy, University of Texas, El Paso, El Paso, Texas, USA
| | - Peter M. Haggie
- Department of Medicine, UCSF, San Francisco, California, USA
- Department of Physiology and
| | - Alan S. Verkman
- Department of Medicine, UCSF, San Francisco, California, USA
- Department of Physiology and
| | - Mark R. Looney
- Department of Medicine, UCSF, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| |
Collapse
|
288
|
Metzemaekers M, Gouwy M, Proost P. Neutrophil chemoattractant receptors in health and disease: double-edged swords. Cell Mol Immunol 2020; 17:433-450. [PMID: 32238918 PMCID: PMC7192912 DOI: 10.1038/s41423-020-0412-0] [Citation(s) in RCA: 307] [Impact Index Per Article: 61.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/05/2020] [Indexed: 02/08/2023] Open
Abstract
Neutrophils are frontline cells of the innate immune system. These effector leukocytes are equipped with intriguing antimicrobial machinery and consequently display high cytotoxic potential. Accurate neutrophil recruitment is essential to combat microbes and to restore homeostasis, for inflammation modulation and resolution, wound healing and tissue repair. After fulfilling the appropriate effector functions, however, dampening neutrophil activation and infiltration is crucial to prevent damage to the host. In humans, chemoattractant molecules can be categorized into four biochemical families, i.e., chemotactic lipids, formyl peptides, complement anaphylatoxins and chemokines. They are critically involved in the tight regulation of neutrophil bone marrow storage and egress and in spatial and temporal neutrophil trafficking between organs. Chemoattractants function by activating dedicated heptahelical G protein-coupled receptors (GPCRs). In addition, emerging evidence suggests an important role for atypical chemoattractant receptors (ACKRs) that do not couple to G proteins in fine-tuning neutrophil migratory and functional responses. The expression levels of chemoattractant receptors are dependent on the level of neutrophil maturation and state of activation, with a pivotal modulatory role for the (inflammatory) environment. Here, we provide an overview of chemoattractant receptors expressed by neutrophils in health and disease. Depending on the (patho)physiological context, specific chemoattractant receptors may be up- or downregulated on distinct neutrophil subsets with beneficial or detrimental consequences, thus opening new windows for the identification of disease biomarkers and potential drug targets.
Collapse
Affiliation(s)
- Mieke Metzemaekers
- Laboratory of Molecular Immunology, Rega Institute, KU Leuven, Herestraat 49 bus 1042, B-3000, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Rega Institute, KU Leuven, Herestraat 49 bus 1042, B-3000, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Rega Institute, KU Leuven, Herestraat 49 bus 1042, B-3000, Leuven, Belgium.
| |
Collapse
|
289
|
Shi Y, Liu T, Nieman DC, Cui Y, Li F, Yang L, Shi H, Chen P. Aerobic Exercise Attenuates Acute Lung Injury Through NET Inhibition. Front Immunol 2020; 11:409. [PMID: 32265910 PMCID: PMC7096358 DOI: 10.3389/fimmu.2020.00409] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction: Aerobic exercise improves lung inflammation in acute lung injury (ALI), but its mechanism remains unknown. Neutrophil extracellular traps (NETs) play an important role in LPS-induced ALI, and a positive correlation exists between NET formation and proinflammatory macrophage polarization. This study investigated whether aerobic exercise reduces the pro-inflammatory polarization of alveolar macrophages (AMs) by inhibiting the excessive release of NETs and then alleviating the inflammatory response of ALI. Methods: C57BL/6 male mice were randomly divided into four groups: sedentary group (CON), sedentary and extra-pulmonary LPS injection group (LPS), 5-weeks aerobic training intervention and LPS injection group (EXE+LPS), and DNase I plus LPS injection group (DNase+LPS). Twenty-four hours after drug injection, bronchoalveolar lavage fluid (BALF), AM, and lung tissues were obtained to detect inflammatory responses, NET formation, macrophage polarization, and protein activation. In the in vitro study, a murine AM cell line, designated MH-S, was stimulated with LPS, purified NETs, and NETs plus DNase I. Results: EXE+LPS and DNase+LPS mice exhibited reduced neutrophil infiltration, decreased NET release, and lower pro-inflammatory polarization of AM compared with LPS mice. Subsequently, Western blot showed inhibition of the phosphorylation of MAPK and NF-κB proteins of AMs in EXE+LPS and DNase+LPS mice compared with LPS mice. Lastly, stimulation of MH-S cells by NETs revealed a trend for pro-inflammatory cell polarization, with NF-κB protein activation at 8 h and ERK1/2 activation at 1, 2, and 8 h. Conclusions: Aerobic exercise alleviated ALI through NET-induced AM pro-inflammatory polarization involving ERK1/2 and NF-κB signaling.
Collapse
Affiliation(s)
- Yue Shi
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Tingting Liu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - David C Nieman
- North Carolina Research Campus, Appalachian State University, Kannapolis, NC, United States
| | - Yanqiu Cui
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Fei Li
- School of Physical Education and Sport Training, Shanghai University of Sport, Shanghai, China
| | - Luyu Yang
- Department of General Surgery, Cancer Metastasis Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Hui Shi
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peijie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
290
|
Hudock KM, Collins MS, Imbrogno M, Snowball J, Kramer EL, Brewington JJ, Gollomp K, McCarthy C, Ostmann AJ, Kopras EJ, Davidson CR, Srdiharan A, Arumugam P, Sengupta S, Xu Y, Worthen GS, Trapnell BC, Clancy JP. Neutrophil extracellular traps activate IL-8 and IL-1 expression in human bronchial epithelia. Am J Physiol Lung Cell Mol Physiol 2020; 319:L137-L147. [PMID: 32159969 DOI: 10.1152/ajplung.00144.2019] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Neutrophil extracellular traps (NETs) provide host defense but can contribute to the pathobiology of diverse human diseases. We sought to determine the extent and mechanism by which NETs contribute to human airway cell inflammation. Primary normal human bronchial epithelial cells (HBEs) grown at air-liquid interface and wild-type (wt)CFBE41o- cells (expressing wtCFTR) were exposed to cell-free NETs from unrelated healthy volunteers for 18 h in vitro. Cytokines were measured in the apical supernatant by Luminex, and the effect on the HBE transcriptome was assessed by RNA sequencing. NETs consistently stimulated IL-8, TNF-α, and IL-1α secretion by HBEs from multiple donors, with variable effects on other cytokines (IL-6, G-CSF, and GM-CSF). Expression of HBE RNAs encoding IL-1 family cytokines, particularly IL-36 subfamily members, was increased in response to NETs. NET exposure in the presence of anakinra [recombinant human IL-1 receptor antagonist (rhIL-1RA)] dampened NET-induced changes in IL-8 and TNF-α proteins as well as IL-36α RNA. rhIL-36RA limited the increase in expression of proinflammatory cytokine RNAs in HBEs exposed to NETs. NETs selectively upregulate an IL-1 family cytokine response in HBEs, which enhances IL-8 production and is limited by rhIL-1RA. The present findings describe a unique mechanism by which NETs may contribute to inflammation in human lung disease in vivo. NET-driven IL-1 signaling may represent a novel target for modulating inflammation in diseases characterized by a substantial NET burden.
Collapse
Affiliation(s)
- Kristin M Hudock
- Division of Adult Pulmonary & Critical Care Medicine, University of Cincinnati, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Margaret S Collins
- Division of Adult Pulmonary & Critical Care Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Michelle Imbrogno
- Division of Adult Pulmonary & Critical Care Medicine, University of Cincinnati, Cincinnati, Ohio
| | - John Snowball
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Elizabeth L Kramer
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - John J Brewington
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kandace Gollomp
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Cormac McCarthy
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Alicia J Ostmann
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Elizabeth J Kopras
- Division of Adult Pulmonary & Critical Care Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Cynthia R Davidson
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Anusha Srdiharan
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Paritha Arumugam
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Shaon Sengupta
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Yan Xu
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - G Scott Worthen
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Bruce C Trapnell
- Division of Adult Pulmonary & Critical Care Medicine, University of Cincinnati, Cincinnati, Ohio.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - John Paul Clancy
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
291
|
Ueki H, Wang IH, Zhao D, Gunzer M, Kawaoka Y. Multicolor two-photon imaging of in vivo cellular pathophysiology upon influenza virus infection using the two-photon IMPRESS. Nat Protoc 2020; 15:1041-1065. [PMID: 31996843 PMCID: PMC7086515 DOI: 10.1038/s41596-019-0275-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/03/2019] [Indexed: 12/14/2022]
Abstract
In vivo two-photon imaging is a valuable technique for studies of viral pathogenesis and host responses to infection in vivo. In this protocol, we describe a methodology for analyzing influenza virus-infected lung in vivo by two-photon imaging microscopy. We describe the surgical procedure, how to stabilize the lung, and an approach to analyzing the data. Further, we provide a database of fluorescent dyes, antibodies, and reporter mouse lines that can be used in combination with a reporter influenza virus (Color-flu) for multicolor analysis. Setup of this model typically takes ~30 min and enables the observation of influenza virus-infected lungs for >4 h during the acute phase of the inflammation and at least 1 h in the lethal phase. This imaging system, which we termed two-photon IMPRESS (imaging pathophysiology research system), is broadly applicable to analyses of other respiratory pathogens and reveals disease progression at the cellular level in vivo.
Collapse
Affiliation(s)
- Hiroshi Ueki
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - I-Hsuan Wang
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Dongming Zhao
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
292
|
Mallavia B, Liu F, Lefrançais E, Cleary SJ, Kwaan N, Tian JJ, Magnen M, Sayah DM, Soong A, Chen J, Saggar R, Shino MY, Ross DJ, Derhovanessian A, Lynch JP, Ardehali A, Weigt SS, Belperio JA, Hays SR, Golden JA, Leard LE, Shah RJ, Kleinhenz ME, Venado A, Kukreja J, Singer JP, Looney MR. Mitochondrial DNA Stimulates TLR9-Dependent Neutrophil Extracellular Trap Formation in Primary Graft Dysfunction. Am J Respir Cell Mol Biol 2020; 62:364-372. [PMID: 31647878 PMCID: PMC7055700 DOI: 10.1165/rcmb.2019-0140oc] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022] Open
Abstract
The immune system is designed to robustly respond to pathogenic stimuli but to be tolerant to endogenous ligands to not trigger autoimmunity. Here, we studied an endogenous damage-associated molecular pattern, mitochondrial DNA (mtDNA), during primary graft dysfunction (PGD) after lung transplantation. We hypothesized that cell-free mtDNA released during lung ischemia-reperfusion triggers neutrophil extracellular trap (NET) formation via TLR9 signaling. We found that mtDNA increases in the BAL fluid of experimental PGD (prolonged cold ischemia followed by orthotopic lung transplantation) and not in control transplants with minimal warm ischemia. The adoptive transfer of mtDNA into the minimal warm ischemia graft immediately before lung anastomosis induces NET formation and lung injury. TLR9 deficiency in neutrophils prevents mtDNA-induced NETs, and TLR9 deficiency in either the lung donor or recipient decreases NET formation and lung injury in the PGD model. Compared with human lung transplant recipients without PGD, severe PGD was associated with high levels of BAL mtDNA and NETs, with evidence of relative deficiency in DNaseI. We conclude that mtDNA released during lung ischemia-reperfusion triggers TLR9-dependent NET formation and drives lung injury. In PGD, DNaseI therapy has a potential dual benefit of neutralizing a major NET trigger (mtDNA) in addition to dismantling pathogenic NETs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Abbas Ardehali
- Department of Surgery, University of California, Los Angeles, Los Angeles, California
| | | | | | | | | | | | | | | | | | | | | | - Mark R. Looney
- Department of Medicine
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, California; and
| |
Collapse
|
293
|
Resolvin D4 attenuates the severity of pathological thrombosis in mice. Blood 2020; 134:1458-1468. [PMID: 31300403 DOI: 10.1182/blood.2018886317] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 07/03/2019] [Indexed: 12/31/2022] Open
Abstract
Deep vein thrombosis (DVT) is a common cardiovascular disease with a major effect on quality of life, and safe and effective therapeutic measures to efficiently reduce existent thrombus burden are scarce. Using a comprehensive targeted liquid chromatography-tandem mass spectrometry-based metabololipidomics approach, we established temporal clusters of endogenously biosynthesized specialized proresolving mediators (SPMs) and proinflammatory and prothrombotic lipid mediators during DVT progression in mice. Administration of resolvin D4 (RvD4), an SPM that was enriched at the natural onset of thrombus resolution, significantly reduced thrombus burden, with significantly less neutrophil infiltration and more proresolving monocytes in the thrombus, as well as an increased number of cells in an early apoptosis state. Moreover, RvD4 promoted the biosynthesis of other D-series resolvins involved in facilitating resolution of inflammation. Neutrophils from RvD4-treated mice were less susceptible to an ionomycin-induced release of neutrophil extracellular traps (NETs), a meshwork of decondensed chromatin lined with histones and neutrophil proteins critical for DVT development. These results suggest that delivery of SPMs, specifically RvD4, modulates the severity of thrombo-inflammatory disease in vivo and improves thrombus resolution.
Collapse
|
294
|
Zhou X, Yang L, Fan X, Zhao X, Chang N, Yang L, Li L. Neutrophil Chemotaxis and NETosis in Murine Chronic Liver Injury via Cannabinoid Receptor 1/ Gα i/o/ ROS/ p38 MAPK Signaling Pathway. Cells 2020; 9:cells9020373. [PMID: 32033504 PMCID: PMC7072548 DOI: 10.3390/cells9020373] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022] Open
Abstract
Neutrophils play an essential role in the control of inflammatory diseases. However, whether cannabinoid receptors (CBs) play a role in neutrophil chemotaxis and NETosis in sterile liver inflammation remains unknown. The expression of marker genes on neutrophils was characterized by FACS, immunofluorescence, qRT-PCR, and Western blot. The amount of neutrophils was significantly elevated from 7 days and reached the peak at 2 weeks in carbon tetrachloride (CCl4)-treated mouse liver. The mRNA expression of neutrophil marker Ly6G had positive correlation with CB1 and CB2 expression in injured liver. In vitro CBs were abundantly expressed in isolated neutrophils and CB1 agonist ACEA promoted the chemotaxis and cytoskeletal remodeling, which can be suppressed by CB1 antagonist AM281. Moreover, ACEA induced NETosis, myeloperoxidase release from lysosome and ROS burst, indicating neutrophil activation, via Gαi/o. Conversely, CB2 agonist JWH133 had no effect on neutrophil function. ROS and p38 MAPK signaling pathways were involved in CB1-mediated neutrophil function, and ROS was upstream of p38 MAPK. CB1 blockade in vivo significantly attenuated neutrophil infiltration and liver inflammation in CCl4-treated mice. Taken together, CB1 mediates neutrophil chemotaxis and NETosis via Gαi/o/ROS/p38 MAPK signaling pathway in liver inflammation, which represents an effective therapeutic strategy for liver diseases.
Collapse
|
295
|
Oleic acid-based nanosystems for mitigating acute respiratory distress syndrome in mice through neutrophil suppression: how the particulate size affects therapeutic efficiency. J Nanobiotechnology 2020; 18:25. [PMID: 32005196 PMCID: PMC6995149 DOI: 10.1186/s12951-020-0583-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 01/21/2020] [Indexed: 12/20/2022] Open
Abstract
Background Oleic acid (OA) is reported to show anti-inflammatory activity toward activated neutrophils. It is also an important material in nanoparticles for increased stability and cellular internalization. We aimed to evaluate the anti-inflammatory activity of injectable OA-based nanoparticles for treating lung injury. Different sizes of nanocarriers were prepared to explore the effect of nanoparticulate size on inflammation inhibition. Results The nanoparticles were fabricated with the mean diameters of 105, 153, and 225 nm. The nanocarriers were ingested by isolated human neutrophils during a 5-min period, with the smaller sizes exhibiting greater uptake. The size reduction led to the decrease of cell viability and the intracellular calcium level. The OA-loaded nanosystems dose-dependently suppressed the superoxide anion and elastase produced by the stimulated neutrophils. The inhibition level was comparable for the nanoparticles of different sizes. In the ex vivo biodistribution study, the pulmonary accumulation of nanoparticles increased following the increase of particle size. The nanocarriers were mainly excreted by the liver and bile clearance. Mice were exposed to intratracheal lipopolysaccharide (LPS) to induce acute respiratory distress syndrome (ARDS), like lung damage. The lipid-based nanocarriers mitigated myeloperoxidase (MPO) and cytokines more effectively as compared to OA solution. The larger nanoparticles displayed greater reduction on MPO, TNF-α, and IL-6 than the smaller ones. The histology confirmed the decreased pulmonary neutrophil recruitment and lung-architecture damage after intravenous administration of larger nanoparticles. Conclusions Nanoparticulate size, an essential property governing the anti-inflammatory effect and lung-injury therapy, had different effects on activated neutrophil inhibition and in vivo therapeutic efficacy.
Collapse
|
296
|
Boeddha NP, Bycroft T, Nadel S, Hazelzet JA. The Inflammatory and Hemostatic Response in Sepsis and Meningococcemia. Crit Care Clin 2020; 36:391-399. [PMID: 32172820 DOI: 10.1016/j.ccc.2019.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Meningococcemia is notorious for evasion of the host immune system and its rapid progression to fulminant disease, and serves as a unique model for pediatric sepsis. Illness severity is determined by complex interplays among host, pathogen, and environment. The inflammatory host response, including proinflammatory and anti-inflammatory responses in innate and adaptive immunity, skews toward a proinflammatory state. This leads to endothelial dysfunction and activation of the hemostatic response, which may lead to disseminated intravascular coagulation. This article reviews the pathogenesis of sepsis, in particular the inflammatory and hemostatic response in meningococcal sepsis.
Collapse
Affiliation(s)
- Navin P Boeddha
- Department of Pediatrics, Erasmus MC-Sophia Children's Hospital, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Thomas Bycroft
- St Mary's Hospital, Imperial College Healthcare NHS Trust, Praed Street, W21NY London, UK
| | - Simon Nadel
- St Mary's Hospital, Imperial College Healthcare NHS Trust, Praed Street, W21NY London, UK; Department of Paediatrics, Faculty of Medicine, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Jan A Hazelzet
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands.
| |
Collapse
|
297
|
Kashiouris MG, L’Heureux M, Cable CA, Fisher BJ, Leichtle SW, Fowler AA. The Emerging Role of Vitamin C as a Treatment for Sepsis. Nutrients 2020; 12:nu12020292. [PMID: 31978969 PMCID: PMC7070236 DOI: 10.3390/nu12020292] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/26/2022] Open
Abstract
Sepsis, a life-threatening organ dysfunction due to a dysregulated host response to infection, is a leading cause of morbidity and mortality worldwide. Decades of research have failed to identify any specific therapeutic targets outside of antibiotics, infectious source elimination, and supportive care. More recently, vitamin C has emerged as a potential therapeutic agent to treat sepsis. Vitamin C has been shown to be deficient in septic patients and the administration of high dose intravenous as opposed to oral vitamin C leads to markedly improved and elevated serum levels. Its physiologic role in sepsis includes attenuating oxidative stress and inflammation, improving vasopressor synthesis, enhancing immune cell function, improving endovascular function, and epigenetic immunologic modifications. Multiple clinical trials have demonstrated the safety of vitamin C and two recent studies have shown promising data on mortality improvement. Currently, larger randomized controlled studies are underway to validate these findings. With further study, vitamin C may become standard of care for the treatment of sepsis, but given its safety profile, current treatment can be justified with compassionate use.
Collapse
Affiliation(s)
- Markos G. Kashiouris
- Department of Internal Medicine, Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University School of Medicine, 1200 E Broad St., P.O. Box 980050, Richmond, VA 23298, USA; (M.L.); (C.A.C.); (B.J.F.); (A.A.F.)
- Correspondence: ; Tel.: +1-(804)-828-9893
| | - Michael L’Heureux
- Department of Internal Medicine, Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University School of Medicine, 1200 E Broad St., P.O. Box 980050, Richmond, VA 23298, USA; (M.L.); (C.A.C.); (B.J.F.); (A.A.F.)
| | - Casey A. Cable
- Department of Internal Medicine, Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University School of Medicine, 1200 E Broad St., P.O. Box 980050, Richmond, VA 23298, USA; (M.L.); (C.A.C.); (B.J.F.); (A.A.F.)
| | - Bernard J. Fisher
- Department of Internal Medicine, Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University School of Medicine, 1200 E Broad St., P.O. Box 980050, Richmond, VA 23298, USA; (M.L.); (C.A.C.); (B.J.F.); (A.A.F.)
| | - Stefan W. Leichtle
- Department of Surgery, Division of Acute Care Surgical Services, Virginia Commonwealth University School of Medicine, 1200 E Broad St., P.O. Box 980454, Richmond, VA 23298, USA;
| | - Alpha A. Fowler
- Department of Internal Medicine, Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University School of Medicine, 1200 E Broad St., P.O. Box 980050, Richmond, VA 23298, USA; (M.L.); (C.A.C.); (B.J.F.); (A.A.F.)
| |
Collapse
|
298
|
Abaricia JO, Shah AH, Musselman RM, Olivares-Navarrete R. Hydrophilic titanium surfaces reduce neutrophil inflammatory response and NETosis. Biomater Sci 2020; 8:2289-2299. [DOI: 10.1039/c9bm01474h] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neutrophils are sensitive to biomaterial surface properties, controlling activation and inflammatory microenvironment, revealing a novel target for enhancing biomaterial integration.
Collapse
Affiliation(s)
- Jefferson O. Abaricia
- Department of Biomedical Engineering
- School of Engineering
- Virginia Commonwealth University
- Richmond
- USA
| | - Arth H. Shah
- Department of Biomedical Engineering
- School of Engineering
- Virginia Commonwealth University
- Richmond
- USA
| | - Ryan M. Musselman
- Department of Biomedical Engineering
- School of Engineering
- Virginia Commonwealth University
- Richmond
- USA
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering
- School of Engineering
- Virginia Commonwealth University
- Richmond
- USA
| |
Collapse
|
299
|
Granger V, Peyneau M, Chollet-Martin S, de Chaisemartin L. Neutrophil Extracellular Traps in Autoimmunity and Allergy: Immune Complexes at Work. Front Immunol 2019; 10:2824. [PMID: 31849989 PMCID: PMC6901596 DOI: 10.3389/fimmu.2019.02824] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/15/2019] [Indexed: 12/17/2022] Open
Abstract
Neutrophil extracellular traps (NETs) have been initially described as main actors in host defense owing to their ability to immobilize and sometimes kill microorganisms. Subsequent studies have demonstrated their implication in the pathophysiology of various diseases, due to the toxic effects of their main components on surrounding tissues. Several distinct NETosis pathways have been described in response to various triggers. Among these triggers, IgG immune complexes (IC) play an important role since they induce robust NET release upon binding to activating FcγRs on neutrophils. Few in vitro studies have documented the mechanisms of IC-induced NET release and evidence about the partners involved is controversial. In vivo, animal models and clinical studies have strongly suggested the importance of IgG IC-induced NET release for autoimmunity and anaphylaxis. In this review, we will focus on two autoimmune diseases in which NETs are undoubtedly major players, systemic lupus erythematosus (SLE), and rheumatoid arthritis (RA). We will also discuss anaphylaxis as another example of disease recently associated with IC-induced NET release. Understanding the role of IC-induced NETs in these settings will pave the way for new diagnostic tools and therapeutic strategies.
Collapse
Affiliation(s)
- Vanessa Granger
- Département d'Immunologie et d'Hématologie, UF Auto-immunité et Hypersensibilités, HUPNVS, Hôpital Bichat, Paris, France.,Inflammation Chimiokines et Immunopathologie, INSERM UMR996, Faculté de Pharmacie, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Marine Peyneau
- Département d'Immunologie et d'Hématologie, UF Auto-immunité et Hypersensibilités, HUPNVS, Hôpital Bichat, Paris, France.,Inflammation Chimiokines et Immunopathologie, INSERM UMR996, Faculté de Pharmacie, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Sylvie Chollet-Martin
- Département d'Immunologie et d'Hématologie, UF Auto-immunité et Hypersensibilités, HUPNVS, Hôpital Bichat, Paris, France.,Inflammation Chimiokines et Immunopathologie, INSERM UMR996, Faculté de Pharmacie, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Luc de Chaisemartin
- Département d'Immunologie et d'Hématologie, UF Auto-immunité et Hypersensibilités, HUPNVS, Hôpital Bichat, Paris, France.,Inflammation Chimiokines et Immunopathologie, INSERM UMR996, Faculté de Pharmacie, Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| |
Collapse
|
300
|
Understanding the Role of Neutrophil Extracellular Traps in Patients With Severe Pneumonia and ARDS. Chest 2019; 156:1278-1280. [DOI: 10.1016/j.chest.2019.08.2179] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/03/2019] [Indexed: 11/23/2022] Open
|