501
|
Tokarz-Deptuła B, Palma J, Baraniecki Ł, Stosik M, Kołacz R, Deptuła W. What Function Do Platelets Play in Inflammation and Bacterial and Viral Infections? Front Immunol 2021; 12:770436. [PMID: 34970260 PMCID: PMC8713818 DOI: 10.3389/fimmu.2021.770436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/22/2021] [Indexed: 12/15/2022] Open
Abstract
The article presents the function of platelets in inflammation as well as in bacterial and viral infections, which are the result of their reaction with the endovascular environment, including cells of damaged vascular endothelium and cells of the immune system. This role of platelets is conditioned by biologically active substances present in their granules and in their specific structures - EV (extracellular vesicles).
Collapse
Affiliation(s)
| | - Joanna Palma
- Department of Biochemical Sciences, Pomeranian Medical University, Szczecin, Poland
| | | | - Michał Stosik
- Institute of Biological Science, Faculty of Biological Sciences, University of Zielona Góra, Zielona Góra, Poland
| | - Roman Kołacz
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | - Wiesław Deptuła
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Toruń, Poland
| |
Collapse
|
502
|
Lung histopathologic clusters in severe COVID-19: a link between clinical picture and tissue damage. Crit Care 2021; 25:423. [PMID: 34903264 PMCID: PMC8667540 DOI: 10.1186/s13054-021-03846-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/28/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Autoptic pulmonary findings have been described in severe COVID-19 patients, but evidence regarding the correlation between clinical picture and lung histopathologic patterns is still weak. METHODS This was a retrospective cohort observational study conducted at the referral center for infectious diseases in northern Italy. Full lung autoptic findings and clinical data of patients who died from COVID-19 were analyzed. Lung histopathologic patterns were scored according to the extent of tissue damage. To consider coexisting histopathologic patterns, hierarchical clustering of histopathologic findings was applied. RESULTS Whole pulmonary examination was available in 75 out of 92 full autopsies. Forty-eight hospitalized patients (64%), 44 from ICU and four from the medical ward, had complete clinical data. The histopathologic patterns had a time-dependent distribution with considerable overlap among patterns. Duration of positive-pressure ventilation (p < 0.0001), mean positive end-expiratory pressure (PEEP) (p = 0.007), worst serum albumin (p = 0.017), interleukin 6 (p = 0.047), and kidney SOFA (p = 0.001) differed among histopathologic clusters. The amount of PEEP for long-lasting ventilatory treatment was associated with the cluster showing the largest areas of early and late proliferative diffuse alveolar damage. No pharmacologic interventions or comorbidities affected the lung histopathology. CONCLUSIONS Our study draws a comprehensive link between the clinical and pulmonary histopathologic findings in a large cohort of COVID-19 patients. These results highlight that the positive end-expiratory pressures and the duration of the ventilatory treatment correlate with lung histopathologic patterns, providing new clues to the knowledge of the pathophysiology of severe SARS-CoV-2 pneumonia.
Collapse
|
503
|
Biering SB, de Sousa FTG, Tjang LV, Pahmeier F, Ruan R, Blanc SF, Patel TS, Worthington CM, Glasner DR, Castillo-Rojas B, Servellita V, Lo NT, Wong MP, Warnes CM, Sandoval DR, Clausen TM, Santos YA, Ortega V, Aguilar HC, Esko JD, Chui CY, Pak JE, Beatty PR, Harris E. SARS-CoV-2 Spike triggers barrier dysfunction and vascular leak via integrins and TGF-β signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.12.10.472112. [PMID: 34931188 PMCID: PMC8687463 DOI: 10.1101/2021.12.10.472112] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Severe COVID-19 is associated with epithelial and endothelial barrier dysfunction within the lung as well as in distal organs. While it is appreciated that an exaggerated inflammatory response is associated with barrier dysfunction, the triggers of this pathology are unclear. Here, we report that cell-intrinsic interactions between the Spike (S) glycoprotein of SARS-CoV-2 and epithelial/endothelial cells are sufficient to trigger barrier dysfunction in vitro and vascular leak in vivo , independently of viral replication and the ACE2 receptor. We identify an S-triggered transcriptional response associated with extracellular matrix reorganization and TGF-β signaling. Using genetic knockouts and specific inhibitors, we demonstrate that glycosaminoglycans, integrins, and the TGF-β signaling axis are required for S-mediated barrier dysfunction. Our findings suggest that S interactions with barrier cells are a contributing factor to COVID-19 disease severity and offer mechanistic insight into SARS-CoV-2 triggered vascular leak, providing a starting point for development of therapies targeting COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Scott B. Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | | | - Laurentia V. Tjang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Felix Pahmeier
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Richard Ruan
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Sophie F. Blanc
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Trishna S. Patel
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | | | - Dustin R. Glasner
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
| | - Bryan Castillo-Rojas
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Venice Servellita
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
| | - Nicholas T.N. Lo
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Marcus P. Wong
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Colin M. Warnes
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Daniel R. Sandoval
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| | - Thomas Mandel Clausen
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| | - Yale A. Santos
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
| | - Victoria Ortega
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Hector C. Aguilar
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Jeffrey D. Esko
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, USA
| | - Charles Y. Chui
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- UCSF-Abbott Viral Diagnostics and Discovery Center, San Francisco, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - John E. Pak
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - P. Robert Beatty
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Lead contact
| |
Collapse
|
504
|
Udovicic I, Stanojevic I, Djordjevic D, Zeba S, Rondovic G, Abazovic T, Lazic S, Vojvodic D, To K, Abazovic D, Khan W, Surbatovic M. Immunomonitoring of Monocyte and Neutrophil Function in Critically Ill Patients: From Sepsis and/or Trauma to COVID-19. J Clin Med 2021; 10:jcm10245815. [PMID: 34945111 PMCID: PMC8706110 DOI: 10.3390/jcm10245815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/27/2021] [Accepted: 12/01/2021] [Indexed: 12/15/2022] Open
Abstract
Immune cells and mediators play a crucial role in the critical care setting but are understudied. This review explores the concept of sepsis and/or injury-induced immunosuppression and immuno-inflammatory response in COVID-19 and reiterates the need for more accurate functional immunomonitoring of monocyte and neutrophil function in these critically ill patients. in addition, the feasibility of circulating and cell-surface immune biomarkers as predictors of infection and/or outcome in critically ill patients is explored. It is clear that, for critically ill, one size does not fit all and that immune phenotyping of critically ill patients may allow the development of a more personalized approach with tailored immunotherapy for the specific patient. In addition, at this point in time, caution is advised regarding the quality of evidence of some COVID-19 studies in the literature.
Collapse
Affiliation(s)
- Ivo Udovicic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
| | - Ivan Stanojevic
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
- Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Dragan Djordjevic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
| | - Snjezana Zeba
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
| | - Goran Rondovic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
| | - Tanja Abazovic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
| | - Srdjan Lazic
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
- Institute of Epidemiology, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Danilo Vojvodic
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
- Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia
| | - Kendrick To
- Division of Trauma & Orthopaedic Surgery, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 2QQ, UK; (K.T.); (W.K.)
| | - Dzihan Abazovic
- Emergency Medical Centar of Montenegro, Vaka Djurovica bb, 81000 Podgorica, Montenegro;
| | - Wasim Khan
- Division of Trauma & Orthopaedic Surgery, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 2QQ, UK; (K.T.); (W.K.)
| | - Maja Surbatovic
- Clinic of Anesthesiology and Intensive Therapy, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; (I.U.); (D.D.); (S.Z.); (G.R.); (T.A.)
- Faculty of Medicine of the Military Medical Academy, University of Defence, Crnotravska 17, 11000 Belgrade, Serbia; (I.S.); (S.L.); (D.V.)
- Correspondence: ; Tel.: +381-11-2665-125
| |
Collapse
|
505
|
Risk Factors for Venous Thromboembolism in Severe COVID-19: A Study-Level Meta-Analysis of 21 Studies. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182412944. [PMID: 34948552 PMCID: PMC8700787 DOI: 10.3390/ijerph182412944] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 02/07/2023]
Abstract
Venous thromboembolism (VTE) in patients with COVID-19 in intensive care units (ICU) is frequent, but risk factors (RF) remain unidentified. In this meta-analysis (CRD42020188764) we searched for observational studies from ICUs reporting the association between VTE and RF in Medline/Embase up to 15 April 2021. Reviewers independently extracted data in duplicate and assessed the certainty of the evidence using the GRADE approach. Analyses were conducted using the random-effects model and produced a non-adjusted odds ratio (OR). We analysed 83 RF from 21 studies (5296 patients). We found moderate-certainty evidence for an association between VTE and the D-dimer peak (OR 5.83, 95%CI 3.18–10.70), and length of hospitalization (OR 7.09, 95%CI 3.41–14.73) and intubation (OR 2.61, 95%CI 1.94–3.51). We identified low-certainty evidence for an association between VTE and CRP (OR 1.83, 95% CI 1.32–2.53), D-dimer (OR 4.58, 95% CI 2.52–8.50), troponin T (OR 8.64, 95% CI 3.25–22.97), and the requirement for inotropic drugs (OR 1.67, 95% CI 1.15–2.43). Traditional VTE RF (i.e., history of cancer, previous VTE events, obesity) were not found to be associated to VTE in COVID-19. Anticoagulation was not associated with a decreased VTE risk. VTE RF in severe COVID-19 correspond to individual illness severity, and inflammatory and coagulation parameters.
Collapse
|
506
|
Oba S, Hosoya T, Amamiya M, Mitsumura T, Kawata D, Sasaki H, Kamiya M, Yamamoto A, Ando T, Shimada S, Shirai T, Okamoto T, Tateishi T, Endo A, Aiboshi J, Nosaka N, Yamanouchi H, Ugawa T, Nagaoka E, Oi K, Tao S, Maejima Y, Tanaka Y, Tanimoto K, Takeuchi H, Tohda S, Hirakawa A, Sasano T, Arai H, Otomo Y, Miyazaki Y, Yasuda S. Arterial and Venous Thrombosis Complicated in COVID-19: A Retrospective Single Center Analysis in Japan. Front Cardiovasc Med 2021; 8:767074. [PMID: 34869681 PMCID: PMC8639692 DOI: 10.3389/fcvm.2021.767074] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/18/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Thrombosis is a characteristic complication in coronavirus disease 2019 (COVID-19). Since coagulopathy has been observed over the entire clinical course, thrombosis might be a clue to understanding the specific pathology in COVID-19. Currently, there is limited epidemiological data of COVID-19-associated thrombosis in the Japanese population and none regarding variant strains of SARS-CoV-2. Here, we elucidate the risk factors and the pattern of thrombosis in COVID-19 patients. Methods: The patients consecutively admitted to Tokyo Medical and Dental University Hospital with COVID-19 were retrospectively analyzed. SARS-CoV-2 variants of concern/interest (VOC/VOI) carrying the spike protein mutants E484K, N501Y, or L452R were identified by PCR-based analysis. All thrombotic events were diagnosed by clinical symptoms, ultrasonography, and/or radiological tests. Results: Among the 516 patients, 32 patients experienced 42 thromboembolic events. Advanced age, severe respiratory conditions, and several abnormal laboratory markers were associated with the development of thrombosis. While thrombotic events occurred in 13% of the patients with a severe respiratory condition, those events still occurred in 2.5% of the patients who did not require oxygen therapy. Elevated D-dimer and ferritin levels on admission were independent risk factors of thrombosis (adjusted odds ratio 9.39 and 3.11, 95% confidence interval 2.08-42.3, and 1.06-9.17, respectively). Of the thrombotic events, 22 were venous, whereas 20 were arterial. While patients with thrombosis received anticoagulation and antiinflammatory therapies with a higher proportion, the mortality rate, organ dysfunctions, and bleeding complications in these patients were higher than those without thrombosis. The incidence of thrombosis in COVID-19 became less frequent over time, such as during the replacement of the earlier strains of SARS-CoV-2 by VOC/VOI and during increased use of anticoagulatory therapeutics. Conclusion: This study elucidated that elevated D-dimer and ferritin levels are useful biomarkers of thrombosis in COVID-19 patients. The comparable incidence of arterial thrombosis with venous thrombosis and the development of thrombosis in less severe patients required further considerations for the management of Japanese patients with COVID-19. Further studies would be required to identify high-risk populations and establish appropriate interventions for thrombotic complications in COVID-19.
Collapse
Affiliation(s)
- Seiya Oba
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tadashi Hosoya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Miki Amamiya
- Department of Cardiovascular Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takahiro Mitsumura
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Daisuke Kawata
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hirokazu Sasaki
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Mari Kamiya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Akio Yamamoto
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takahiro Ando
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Sho Shimada
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tsuyoshi Shirai
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tsukasa Okamoto
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tomoya Tateishi
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Akira Endo
- Trauma and Acute Critical Care Medical Center, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Junichi Aiboshi
- Trauma and Acute Critical Care Medical Center, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Nobuyuki Nosaka
- Department of Intensive Care Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hideo Yamanouchi
- Department of Intensive Care Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Toyomu Ugawa
- Department of Intensive Care Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Eiki Nagaoka
- Department of Cardiovascular Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Keiji Oi
- Department of Cardiovascular Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Susumu Tao
- Department of Cardiovascular Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yasuhiro Maejima
- Department of Cardiovascular Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yukie Tanaka
- Research Core, Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kousuke Tanimoto
- Research Core, Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Genome Laboratory, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiroaki Takeuchi
- Department of Molecular Virology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shuji Tohda
- Clinical Laboratory, Tokyo Medical and Dental University (TMDU) Hospital, Tokyo, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tetsuo Sasano
- Department of Cardiovascular Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hirokuni Arai
- Department of Cardiovascular Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yasuhiro Otomo
- Trauma and Acute Critical Care Medical Center, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shinsuke Yasuda
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
507
|
Denson JL, Gillet AS, Zu Y, Brown M, Pham T, Yoshida Y, Mauvais-Jarvis F, Douglas IS, Moore M, Tea K, Wetherbie A, Stevens R, Lefante J, Shaffer JG, Armaignac DL, Belden KA, Kaufman M, Heavner SF, Danesh VC, Cheruku SR, St Hill CA, Boman K, Deo N, Bansal V, Kumar VK, Walkey AJ, Kashyap R. Metabolic Syndrome and Acute Respiratory Distress Syndrome in Hospitalized Patients With COVID-19. JAMA Netw Open 2021; 4:e2140568. [PMID: 34935924 PMCID: PMC8696573 DOI: 10.1001/jamanetworkopen.2021.40568] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
IMPORTANCE Obesity, diabetes, and hypertension are common comorbidities in patients with severe COVID-19, yet little is known about the risk of acute respiratory distress syndrome (ARDS) or death in patients with COVID-19 and metabolic syndrome. OBJECTIVE To determine whether metabolic syndrome is associated with an increased risk of ARDS and death from COVID-19. DESIGN, SETTING, AND PARTICIPANTS This multicenter cohort study used data from the Society of Critical Care Medicine Discovery Viral Respiratory Illness Universal Study collected from 181 hospitals across 26 countries from February 15, 2020, to February 18, 2021. Outcomes were compared between patients with metabolic syndrome (defined as ≥3 of the following criteria: obesity, prediabetes or diabetes, hypertension, and dyslipidemia) and a control population without metabolic syndrome. Participants included adult patients hospitalized for COVID-19 during the study period who had a completed discharge status. Data were analyzed from February 22 to October 5, 2021. EXPOSURES Exposures were SARS-CoV-2 infection, metabolic syndrome, obesity, prediabetes or diabetes, hypertension, and/or dyslipidemia. MAIN OUTCOMES AND MEASURES The primary outcome was in-hospital mortality. Secondary outcomes included ARDS, intensive care unit (ICU) admission, need for invasive mechanical ventilation, and length of stay (LOS). RESULTS Among 46 441 patients hospitalized with COVID-19, 29 040 patients (mean [SD] age, 61.2 [17.8] years; 13 059 [45.0%] women and 15713 [54.1%] men; 6797 Black patients [23.4%], 5325 Hispanic patients [18.3%], and 16 507 White patients [57.8%]) met inclusion criteria. A total of 5069 patients (17.5%) with metabolic syndrome were compared with 23 971 control patients (82.5%) without metabolic syndrome. In adjusted analyses, metabolic syndrome was associated with increased risk of ICU admission (adjusted odds ratio [aOR], 1.32 [95% CI, 1.14-1.53]), invasive mechanical ventilation (aOR, 1.45 [95% CI, 1.28-1.65]), ARDS (aOR, 1.36 [95% CI, 1.12-1.66]), and mortality (aOR, 1.19 [95% CI, 1.08-1.31]) and prolonged hospital LOS (median [IQR], 8.0 [4.2-15.8] days vs 6.8 [3.4-13.0] days; P < .001) and ICU LOS (median [IQR], 7.0 [2.8-15.0] days vs 6.4 [2.7-13.0] days; P < .001). Each additional metabolic syndrome criterion was associated with increased risk of ARDS in an additive fashion (1 criterion: 1147 patients with ARDS [10.4%]; P = .83; 2 criteria: 1191 patients with ARDS [15.3%]; P < .001; 3 criteria: 817 patients with ARDS [19.3%]; P < .001; 4 criteria: 203 patients with ARDS [24.3%]; P < .001). CONCLUSIONS AND RELEVANCE These findings suggest that metabolic syndrome was associated with increased risks of ARDS and death in patients hospitalized with COVID-19. The association with ARDS was cumulative for each metabolic syndrome criteria present.
Collapse
Affiliation(s)
- Joshua L Denson
- Section of Pulmonary Diseases, Critical Care, and Environmental Medicine, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | | | - Yuanhao Zu
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Margo Brown
- Section of Pulmonary Diseases, Critical Care, and Environmental Medicine, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Thaidan Pham
- Section of Pulmonary Diseases, Critical Care, and Environmental Medicine, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Yilin Yoshida
- Section of Endocrinology and Metabolism, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
- Southeast Louisiana Veterans Affairs Healthcare System, New Orleans
| | - Franck Mauvais-Jarvis
- Section of Endocrinology and Metabolism, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
- Southeast Louisiana Veterans Affairs Healthcare System, New Orleans
| | - Ivor S Douglas
- Division of Pulmonary Sciences & Critical Care Medicine, Denver Health Medical Center, Denver, Colorado
- University of Colorado, Anschutz School of Medicine, Aurora
| | - Mathew Moore
- Section of Pulmonary Diseases, Critical Care, and Environmental Medicine, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Kevin Tea
- Section of Pulmonary Diseases, Critical Care, and Environmental Medicine, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Andrew Wetherbie
- Section of Pulmonary Diseases, Critical Care, and Environmental Medicine, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Rachael Stevens
- Section of Pulmonary Diseases, Critical Care, and Environmental Medicine, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - John Lefante
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Jeffrey G Shaffer
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | | | - Katherine A Belden
- Division of Infectious Diseases, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | | | - Smith F Heavner
- Prisma Health Department of Medicine, Prisma Health Upstate, Greenville, South Carolina
| | - Valerie C Danesh
- Baylor Scott & White Health, Department of Nursing, Dallas, Texas
| | - Sreekanth R Cheruku
- Divisions of Cardiothoracic Anesthesiology and Critical Care Medicine, Department of Anesthesiology and Pain Management, UT Southwestern Medical Center, Dallas, Texas
| | | | - Karen Boman
- Society of Critical Care Medicine, Mount Prospect, Illinois
| | - Neha Deo
- Department of Anesthesia and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Vikas Bansal
- Department of Anesthesia and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Allan J Walkey
- The Pulmonary Center, Division of Pulmonary, Allergy, Sleep and Critical Care; Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Rahul Kashyap
- Department of Anesthesia and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
508
|
Al-Kuraishy HM, Al-Gareeb AI, Alzahrani KJ, Alexiou A, Batiha GES. Niclosamide for Covid-19: bridging the gap. Mol Biol Rep 2021; 48:8195-8202. [PMID: 34664162 PMCID: PMC8522539 DOI: 10.1007/s11033-021-06770-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/17/2021] [Indexed: 01/18/2023]
Abstract
AIM/PURPOSE Niclosamide (NCL) is an anthelminthic drug, which is widely used to treat various diseases due to its pleiotropic anti-inflammatory and antiviral activities. NCL modulates of uncoupling oxidative phosphorylation and different signaling pathways in human biological processes. The wide-spectrum antiviral effect of NCL makes it a possible candidate for recent pandemic SARS-CoV-2 infection and may reduce Covid-19 severity. Therefore, the aim of the present study was to review and clarify the potential role of NCL in Covid-19. METHODS This study reviewed and highlighted the protective role of NCL therapy in Covid-19. A related literature search in PubMed, Scopus, Web of Science, Google Scholar, and Science Direct was done. RESULTS NCL has noteworthy anti-inflammatory and antiviral effects. The primary antiviral mechanism of NCL is through neutralization of endosomal PH and inhibition of viral protein maturation. NCL acts as a proton carrier, inhibits homeostasis of endosomal PH, which limiting of viral proliferation and release. The anti-inflammatory effects of NCL are mediated by suppression of inflammatory signaling pathways and release of pro-inflammatory cytokines. However, the major limitation in using NCL is low aqueous solubility, which reduces oral bioavailability and therapeutic serum concentration that reducing the in vivo effect of NCL against SARS-CoV-2. CONCLUSIONS NCL has anti-inflammatory and immune regulatory effects by modulating the release of pro-inflammatory cytokines, inhibition of NF-κB /NLRP3 inflammasome and mTOR signaling pathway. NCL has an anti-SARS-CoV-2 effect via interruption of viral life-cycle and/or induction of cytopathic effect. Prospective clinical studies and clinical trials are mandatory to confirm the potential role of NCL in patients with Covid-19 concerning the severity and clinical outcomes.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, PO Box 11099, Taif, 21944, Saudi Arabia
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia.
- AFNP Med Austria, Wien, Austria.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, El Beheira, Egypt.
| |
Collapse
|
509
|
Terrier O, Si-Tahar M, Ducatez M, Chevalier C, Pizzorno A, Le Goffic R, Crépin T, Simon G, Naffakh N. Influenza viruses and coronaviruses: Knowns, unknowns, and common research challenges. PLoS Pathog 2021; 17:e1010106. [PMID: 34969061 PMCID: PMC8718010 DOI: 10.1371/journal.ppat.1010106] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The development of safe and effective vaccines in a record time after the emergence of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a remarkable achievement, partly based on the experience gained from multiple viral outbreaks in the past decades. However, the Coronavirus Disease 2019 (COVID-19) crisis also revealed weaknesses in the global pandemic response and large gaps that remain in our knowledge of the biology of coronaviruses (CoVs) and influenza viruses, the 2 major respiratory viruses with pandemic potential. Here, we review current knowns and unknowns of influenza viruses and CoVs, and we highlight common research challenges they pose in 3 areas: the mechanisms of viral emergence and adaptation to humans, the physiological and molecular determinants of disease severity, and the development of control strategies. We outline multidisciplinary approaches and technological innovations that need to be harnessed in order to improve preparedeness to the next pandemic.
Collapse
Affiliation(s)
- Olivier Terrier
- CNRS GDR2073 ResaFlu, Groupement de Recherche sur les Virus Influenza, France
- CIRI, Centre International de Recherche en Infectiologie (Team VirPath), Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - Mustapha Si-Tahar
- CNRS GDR2073 ResaFlu, Groupement de Recherche sur les Virus Influenza, France
- Inserm U1100, Research Center for Respiratory Diseases (CEPR), Université de Tours, Tours, France
| | - Mariette Ducatez
- CNRS GDR2073 ResaFlu, Groupement de Recherche sur les Virus Influenza, France
- IHAP, UMR1225, Université de Toulouse, ENVT, INRAE, Toulouse, France
| | - Christophe Chevalier
- CNRS GDR2073 ResaFlu, Groupement de Recherche sur les Virus Influenza, France
- Université Paris-Saclay, UVSQ, INRAE, VIM, Equipe Virus Influenza, Jouy-en-Josas, France
| | - Andrés Pizzorno
- CNRS GDR2073 ResaFlu, Groupement de Recherche sur les Virus Influenza, France
- CIRI, Centre International de Recherche en Infectiologie (Team VirPath), Inserm U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - Ronan Le Goffic
- CNRS GDR2073 ResaFlu, Groupement de Recherche sur les Virus Influenza, France
- Université Paris-Saclay, UVSQ, INRAE, VIM, Equipe Virus Influenza, Jouy-en-Josas, France
| | - Thibaut Crépin
- CNRS GDR2073 ResaFlu, Groupement de Recherche sur les Virus Influenza, France
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, Grenoble, France
| | - Gaëlle Simon
- CNRS GDR2073 ResaFlu, Groupement de Recherche sur les Virus Influenza, France
- Swine Virology Immunology Unit, Ploufragan-Plouzané-Niort Laboratory, ANSES, Ploufragan, France
| | - Nadia Naffakh
- CNRS GDR2073 ResaFlu, Groupement de Recherche sur les Virus Influenza, France
- RNA Biology and Influenza Virus Unit, Institut Pasteur, CNRS UMR3569, Université de Paris, Paris, France
| |
Collapse
|
510
|
Goswami J, MacArthur TA, Sridharan M, Tange J, Kirmse AJ, Lundell KA, Chen D, Auton MT, Chon TY, Hurt RT, Salonen BR, Ganesh R, Erben YM, Marquez CP, Dong JF, Kozar RA, Heller SF, Loomis EA, Johnstone AL, Bailey KR, Spears GM, Park MS. Biomarkers of thromboinflammation correlate to COVID-19 infection and admission status in emergency department patients. THROMBOSIS UPDATE 2021; 5:100090. [PMID: 38620680 PMCID: PMC8603399 DOI: 10.1016/j.tru.2021.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/09/2021] [Accepted: 11/18/2021] [Indexed: 11/14/2022] Open
Abstract
Background COVID-19-associated coagulopathy is incompletely understood. Objectives To characterize thrombin generation, Von Willebrand Factor (VWF), neutrophil extracellular traps (NETs), and their role in COVID-19 risk stratification in the emergency department (ED). Patients/methods Plasma samples from 67 ED COVID-19 patients were compared to 38 healthy volunteers (HVs). Thrombin generation (calibrated automated thrombogram, CAT) was expressed as lag time (LT, min), peak height (PH, min), and time to peak (ttPeak, min). Citrullinated nucleosomes and histones were quantified with ELISA, VWF antigen and activity (IU/dL) through latex immunoassay, Factor VIII (IU/dL) through one-stage optical clot detection, and VWF multimers with Western blot densitometry. Wilcoxon testing and multivariable logistic regression were performed. Results presented as median [Q1, Q3]; p < 0.05 significant. Results COVID-19 patients had longer LT (4.00 [3.26, 4.67]; 2.95 [2.67, 3.10], p < 0.001) and ttPeak (7.33 [6.33, 8.04]; 6.45 [6.00, 7.50], p = 0.004), greater VWF antigen (212 [158, 275]; 110 [91, 128], p < 0.001) and Factor VIII levels (148 [106, 190]; 106 [86, 129], p < 0.001), with decreased high molecular weight multimers (Normalized multimer ratio 0.807 [0.759, 0.869]; 0.891 [0.858, 0.966], p < 0.001), than HVs. COVID-19 patients requiring admission from the ED had longer LT and ttPeak with greater VWF antigen and Factor VIII levels than those not admitted. Two and three variable models of CAT parameters and VWF correlated with COVID-19 and admission status (C-statistics 0.677 to 0.922). Conclusions Thrombin generation kinetics and VWF levels, independent of NETs, may have a role in predicting admission need for COVID-19 patients.
Collapse
Affiliation(s)
- Julie Goswami
- Trauma, Critical Care, and General Surgery, Department of Surgery, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Taleen A MacArthur
- Trauma, Critical Care, and General Surgery, Department of Surgery, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Meera Sridharan
- Department of Hematology, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Julie Tange
- Department of Hematology, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Andrew J Kirmse
- Trauma, Critical Care, and General Surgery, Department of Surgery, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Kaitlin A Lundell
- Trauma, Critical Care, and General Surgery, Department of Surgery, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Dong Chen
- Division of Hematopathology, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Matthew T Auton
- Division of Biochemistry and Molecular Biology, Department of Hematology, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Tony Y Chon
- Department of General Internal Medicine, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Ryan T Hurt
- Department of General Internal Medicine, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Bradley R Salonen
- Department of General Internal Medicine, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Ravindra Ganesh
- Department of General Internal Medicine, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Young M Erben
- Department of Vascular and Endovascular Surgery, Mayo Clinic, 4500 San Pablo Road S., Jacksonville, FL, 32224, USA
| | - Christopher P Marquez
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 4500 San Pablo Road S., Jacksonville, FL, 32224, USA
| | - Jing-Fei Dong
- Division of Hematology, University of Washington School of Medicine, Bloodworks Research Institute, 1551 Eastlake Avenue E, Seattle, WA, 98102, USA
| | - Rosemary A Kozar
- Shock Trauma Center, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD, 21201, USA
| | - Stephanie F Heller
- Trauma, Critical Care, and General Surgery, Department of Surgery, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Erica A Loomis
- Trauma, Critical Care, and General Surgery, Department of Surgery, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | | | - Kent R Bailey
- Clinical Statistics and Biostatistics, Department of Health Sciences Research, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Grant M Spears
- Clinical Statistics and Biostatistics, Department of Health Sciences Research, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| | - Myung S Park
- Trauma, Critical Care, and General Surgery, Department of Surgery, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
- Department of Hematology, Mayo Clinic, 200 1st St. SW, Rochester, MN, 55905, USA
| |
Collapse
|
511
|
Koehler M, Ray A, Moreira RA, Juniku B, Poma AB, Alsteens D. Molecular insights into receptor binding energetics and neutralization of SARS-CoV-2 variants. Nat Commun 2021; 12:6977. [PMID: 34848718 PMCID: PMC8633007 DOI: 10.1038/s41467-021-27325-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Despite an unprecedented global gain in knowledge since the emergence of SARS-CoV-2, almost all mechanistic knowledge related to the molecular and cellular details of viral replication, pathology and virulence has been generated using early prototypic isolates of SARS-CoV-2. Here, using atomic force microscopy and molecular dynamics, we investigated how these mutations quantitatively affected the kinetic, thermodynamic and structural properties of RBD-ACE2 complex formation. We observed for several variants of concern a significant increase in the RBD-ACE2 complex stability. While the N501Y and E484Q mutations are particularly important for the greater stability, the N501Y mutation is unlikely to significantly affect antibody neutralization. This work provides unprecedented atomistic detail on the binding of SARS-CoV-2 variants and provides insight into the impact of viral mutations on infection-induced immunity.
Collapse
Affiliation(s)
- Melanie Koehler
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Ankita Ray
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Rodrigo A Moreira
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Pawińskiego 5B, 02-106, Warsaw, Poland
| | - Blinera Juniku
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Adolfo B Poma
- International Center for Research on Innovative Biobased Materials (ICRI-BioM)-International Research Agenda, Lodz University of Technology, Żeromskiego 116, 90-924, Lodz, Poland.
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Louvain-la-Neuve, Belgium.
- Walloon Excellence in Life sciences and Biotechnology (WELBIO), 1300, Wavre, Belgium.
| |
Collapse
|
512
|
Lovre D, Bateman K, Sherman M, Fonseca VA, Lefante J, Mauvais-Jarvis F. Acute estradiol and progesterone therapy in hospitalised adults to reduce COVID-19 severity: a randomised control trial. BMJ Open 2021; 11:e053684. [PMID: 34848523 PMCID: PMC8635865 DOI: 10.1136/bmjopen-2021-053684] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION As of November 2021, COVID-19 has killed more than 5 million people globally, including over 750 000 in the USA. Apart from corticosteroids, most available therapeutic options are at best marginally efficient in reducing disease severity and are extremely expensive. The systematic investigation of clinically approved drugs is a priority to determine what does mitigate disease severity. Oestradiol (E2) and progesterone (P4) produce a state of anti-inflammatory immune responses and immune tolerance, and enhanced antibody production. The goal of this trial is to evaluate the efficacy of a short E2 and P4 therapy, in addition to standard of care (SOC), in mitigating disease severity in COVID-19 hospitalised patients. METHODS AND ANALYSIS Phase 2, randomised, double blind, placebo-controlled, single-centre trial. Patients hospitalised for confirmed COVID-19, with scores 3-5 on the 9-point WHO ordinal scale are randomised between two arms: (1) Oestradiol cypionate intramuscular (IM) and micronised progesterone oral (PO), in addition to SOC, and (2) placebo, in addition to SOC. The primary outcome is the proportion of patients improving to scores 1 or 2 on the WHO scale through day 28. Secondary outcomes include length of hospital stay, duration of mechanical ventilation, cause of death, readmission rates, change in inflammatory biomarkers between admission and occurrence of primary endpoint, and adverse events. Study sample size will be up to 120 participants. The trial is currently recruiting subjects. ETHICS AND DISSEMINATION The sponsor of this study is the Center of Excellence in Sex-Based Biology & Medicine at Tulane University, New Orleans, Louisiana, USA. Ethical approval was obtained from the Tulane institutional review board on 14 May 2021. The study was reviewed by the US Food and Drug Administration and granted Investigational New Drug #152 499. Results of the study will be submitted for publication in a peer-reviewed journal. TRIAL REGISTRATION NUMBER NCT04865029; Pre-results.
Collapse
Affiliation(s)
- Dragana Lovre
- Section of Endocrinology and Metabolism, John W. Deming Department of Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
- Section of Endocrinology, Department of Medicine, Southeast Louisiana Veterans Health Care System, New Orleans, Louisiana, USA
- Tulane Center of Excellence in Sex-Based Biology & Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Kristin Bateman
- Section of General Internal Medicine and Geriatrics, John W. Deming Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Mya Sherman
- Clinical Translational Unit, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Vivian A Fonseca
- Section of Endocrinology and Metabolism, John W. Deming Department of Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
- Section of Endocrinology, Department of Medicine, Southeast Louisiana Veterans Health Care System, New Orleans, Louisiana, USA
| | - John Lefante
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Franck Mauvais-Jarvis
- Section of Endocrinology and Metabolism, John W. Deming Department of Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
- Section of Endocrinology, Department of Medicine, Southeast Louisiana Veterans Health Care System, New Orleans, Louisiana, USA
- Tulane Center of Excellence in Sex-Based Biology & Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| |
Collapse
|
513
|
Emadi-Baygi M, Ehsanifard M, Afrashtehpour N, Norouzi M, Joz-Abbasalian Z. Corona Virus Disease 2019 (COVID-19) as a System-Level Infectious Disease With Distinct Sex Disparities. Front Immunol 2021; 12:778913. [PMID: 34912345 PMCID: PMC8667725 DOI: 10.3389/fimmu.2021.778913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/11/2021] [Indexed: 01/08/2023] Open
Abstract
The current global pandemic of the Severe Acute Respiratory Syndrome CoronaVirus 2 (SARS-CoV-2) causing COVID-19, has infected millions of people and continues to pose a threat to many more. Angiotensin-Converting Enzyme 2 (ACE2) is an important player of the Renin-Angiotensin System (RAS) expressed on the surface of the lung, heart, kidney, neurons, and endothelial cells, which mediates SARS-CoV-2 entry into the host cells. The cytokine storms of COVID-19 arise from the large recruitment of immune cells because of the dis-synchronized hyperactive immune system, lead to many abnormalities including hyper-inflammation, endotheliopathy, and hypercoagulability that produce multi-organ dysfunction and increased the risk of arterial and venous thrombosis resulting in more severe illness and mortality. We discuss the aberrated interconnectedness and forthcoming crosstalks between immunity, the endothelium, and coagulation, as well as how sex disparities affect the severity and outcome of COVID-19 and harm men especially. Further, our conceptual framework may help to explain why persistent symptoms, such as reduced physical fitness and fatigue during long COVID, may be rooted in the clotting system.
Collapse
Affiliation(s)
- Modjtaba Emadi-Baygi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Mahsa Ehsanifard
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Najmeh Afrashtehpour
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Mahnaz Norouzi
- Department of Research and Development, Erythrogen Medical Genetics Lab, Isfahan, Iran
| | - Zahra Joz-Abbasalian
- Clinical Laboratory, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
514
|
Al-Gburi S, Beissert S, Günther C. Molecular mechanisms of vasculopathy and coagulopathy in COVID-19. Biol Chem 2021; 402:1505-1518. [PMID: 34657406 DOI: 10.1515/hsz-2021-0245] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/06/2021] [Indexed: 01/08/2023]
Abstract
COVID-19 primarily affects the respiratory system and may lead to severe systemic complications, such as acute respiratory distress syndrome (ARDS), multiple organ failure, cytokine storm, and thromboembolic events. Depending on the immune status of the affected individual early disease control can be reached by a robust type-I-interferon (type-I-IFN) response restricting viral replication. If type-I-IFN upregulation is impaired, patients develop severe COVID-19 that involves profound alveolitis, endothelitis, complement activation, recruitment of immune cells, as well as immunothrombosis. In patients with proper initial disease control there can be a second flare of type-I-IFN release leading to post-COVID manifestation such as chilblain-like lesions that are characterized by thrombosis of small vessels in addition to an inflammatory infiltrate resembling lupus erythematosus (LE). Mechanistically, SARS-CoV-2 invades pneumocytes and endothelial cells by acting on angiotensin-II-converting enzyme 2 (ACE2). It is hypothesized, that viral uptake might downregulate ACE2 bioavailability and enhance angiotensin-II-derived pro-inflammatory and pro-thrombotic state. Since ACE2 is encoded on the X chromosome these conditions might also be influenced by gender-specific regulation. Taken together, SARS-CoV-2 infection affects the vascular compartment leading to variable thrombogenic or inflammatory response depending on the individual immune response status.
Collapse
Affiliation(s)
- Suzan Al-Gburi
- University Hospital Carl Gustav Carus, Technical University of Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Stefan Beissert
- University Hospital Carl Gustav Carus, Technical University of Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| | - Claudia Günther
- University Hospital Carl Gustav Carus, Technical University of Dresden, Fetscherstr. 74, D-01307 Dresden, Germany
| |
Collapse
|
515
|
Al-Abani K, Kilhamn N, Maret E, Mårtensson J. Thrombosis and Bleeding After Implementation of an Intermediate-Dose Prophylactic Anticoagulation Protocol in ICU Patients With COVID-19: A Multicenter Screening Study. J Intensive Care Med 2021; 37:480-490. [PMID: 34821162 DOI: 10.1177/08850666211051960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Thrombosis and bleeding after implementation of an intermediate-dose prophylactic anticoagulation protocol in intensive care unit (ICU) patients with coronavirus disease 2019 (COVID-19): a multicenter screening study Background: Venous thromboembolism (VTE) is common among critically ill patients with COVID-19. Information regarding VTE prevalence and bleeding complications after implementation of intermediate-dose prophylactic anticoagulation in such patients is, however, limited. Methods: We performed a prospective, observational study in 6 ICUs in 2 university-affiliated teaching hospitals in Sweden. After implementation of an intermediate-dose prophylactic anticoagulation protocol, we performed ultrasound screening for proximal lower-extremity deep vein thrombosis (DVT) and collected routine computed tomography pulmonary angiography exam results. Results: A total of 100 COVID-19 patients were included from June 21, 2020, through February 18, 2021. During a median follow-up of 120 (IQR, 89-134) days, we found VTE in 37 patients with the majority (78.4%) being diagnosed after ICU arrival. Overall, 20 patients had proximal lower-extremity DVT with 95% being detected on ultrasound screening; 22 patients had pulmonary vascular thrombosis; and 4 patients had venous thrombosis at other sites. A total of 6 patients had both proximal lower-extremity DVT and pulmonary vascular thrombosis. On univariate logistic regression analysis of 14 baseline characteristics, only pre-existing heart failure was associated with VTE (OR 4.67, 95% CI 1.13-19.34). Major and non-major bleeding occurred in 10 and 18 patients, respectively. Conclusions: In our cohort of ICU patients with COVID-19, we observed a high prevalence of VTE and bleeding complications after implementation of intermediate-dose anticoagulation. In approximately half of patients, VTE was identified on screening ultrasound.
Collapse
Affiliation(s)
- Kais Al-Abani
- 59562Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Naima Kilhamn
- 59562Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Eva Maret
- 27106Department of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden.,27106Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Johan Mårtensson
- 59562Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden.,27106Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
516
|
Pandolfi S, Chirumbolo S, Ricevuti G, Valdenassi L, Bjørklund G, Lysiuk R, Doşa MD, Lenchyk L, Fazio S. Home pharmacological therapy in early COVID-19 to prevent hospitalization and reduce mortality: Time for a suitable proposal. Basic Clin Pharmacol Toxicol 2021; 130:225-239. [PMID: 34811895 PMCID: PMC9011697 DOI: 10.1111/bcpt.13690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/03/2021] [Accepted: 11/16/2021] [Indexed: 01/08/2023]
Abstract
The COVID‐19 pandemic is a highly dramatic concern for mankind. In Italy, the pandemic exerted its major impact throughout the period of February to June 2020. To date, the awkward amount of more than 134,000 deaths has been reported. Yet, post‐mortem autopsy was performed on a very modest number of patients who died from COVID‐19 infection, leading to a first confirmation of an immune‐thrombosis of the lungs as the major COVID‐19 pathogenesis, likewise for SARS. Since then (June–August 2020), no targeted early therapy considering this pathogenetic issue was approached. The patients treated with early anti‐inflammatory, anti‐platelet, anticoagulant and antibiotic therapy confirmed that COVID‐19 was an endothelial inflammation with immuno‐thrombosis. Patients not treated or scarcely treated with the most proper and appropriate therapy and in the earliest, increased the hospitalization rate in the intensive care units and also mortality, due to immune‐thrombosis from the pulmonary capillary district and alveoli. The disease causes widespread endothelial inflammation, which can induce damage to various organs and systems. Therapy must be targeted in this consideration, and in this review, we demonstrate how early anti‐inflammatory therapy may treat endothelia inflammation and immune‐thrombosis caused by COVID‐19, by using drugs we are going to recommend in this paper.
Collapse
Affiliation(s)
- Sergio Pandolfi
- High School of Oxygen Ozone Therapy, University of Pavia, Pavia, Italy.,Unit of Neurosurgery, Villa Mafalda Health Clinics, Rome, Italy
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Italy
| | | | - Luigi Valdenassi
- High School of Oxygen Ozone Therapy, University of Pavia, Pavia, Italy
| | - Geir Bjørklund
- Department of Direction Board, Council for Nutritional an Environmental Medicine (CONEM), Mo i Rana, Norway
| | - Roman Lysiuk
- CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Monica Daniela Doşa
- Department of Pharmacology, Faculty of Medicine, Ovidius University, Constanta, Romania
| | - Larysa Lenchyk
- CONEM Ukraine Pharmacognosy and Natural Product Chemistry Research Group, National University of Pharmacy, Kharkiv, Ukraine
| | - Serafino Fazio
- Department of Internal Medicine, University of Naples Federico II, Naples, Italy
| |
Collapse
|
517
|
Freiwald T, Afzali B. Renal diseases and the role of complement: Linking complement to immune effector pathways and therapeutics. Adv Immunol 2021; 152:1-81. [PMID: 34844708 PMCID: PMC8905641 DOI: 10.1016/bs.ai.2021.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The complement system is an ancient and phylogenetically conserved key danger sensing system that is critical for host defense against pathogens. Activation of the complement system is a vital component of innate immunity required for the detection and removal of pathogens. It is also a central orchestrator of adaptive immune responses and a constituent of normal tissue homeostasis. Once complement activation occurs, this system deposits indiscriminately on any cell surface in the vicinity and has the potential to cause unwanted and excessive tissue injury. Deposition of complement components is recognized as a hallmark of a variety of kidney diseases, where it is indeed associated with damage to the self. The provenance and the pathophysiological role(s) played by complement in each kidney disease is not fully understood. However, in recent years there has been a renaissance in the study of complement, with greater appreciation of its intracellular roles as a cell-intrinsic system and its interplay with immune effector pathways. This has been paired with a profusion of novel therapeutic agents antagonizing complement components, including approved inhibitors against complement components (C)1, C3, C5 and C5aR1. A number of clinical trials have investigated the use of these more targeted approaches for the management of kidney diseases. In this review we present and summarize the evidence for the roles of complement in kidney diseases and discuss the available clinical evidence for complement inhibition.
Collapse
Affiliation(s)
- Tilo Freiwald
- Immunoregulation Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, MD, United States; Department of Nephrology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany
| | - Behdad Afzali
- Department of Nephrology, University Hospital Frankfurt, Goethe-University, Frankfurt am Main, Germany.
| |
Collapse
|
518
|
Sattui SE, Conway R, Putman MS, Seet AM, Gianfrancesco MA, Beins K, Hill C, Liew D, Mackie SL, Mehta P, Neill L, Gomez G, Salinas MIH, Maldonado FN, Mariz HA, de Sousa Studart SA, Araujo NC, Knight A, Rozza D, Quartuccio L, Samson M, Bally S, Maria AT, Chazerain P, Hasseli R, Müller-Ladner U, Hoyer BF, Voll R, Torres RP, Luis M, Ribeirio SLE, Al-Emadi S, Sparks JA, Hsu TYT, D'Silva KM, Patel NJ, Wise L, Gilbert E, Almada MV, Duarte-García A, Ugarte-Gil M, Jacobsohn L, Izadi Z, Strangfeld A, Mateus EF, Hyrich KL, Gossec L, Carmona L, Lawson-Tovey S, Kearsley-Fleet L, Schaefer M, Sirotich E, Hausmann JS, Sufka P, Bhana S, Liew JW, Grainger R, Machado PM, Wallace ZS, Yazdany J, Robinson PC. Outcomes of COVID-19 in patients with primary systemic vasculitis or polymyalgia rheumatica from the COVID-19 Global Rheumatology Alliance physician registry: a retrospective cohort study. LANCET RHEUMATOLOGY 2021; 3:e855-e864. [PMID: 34778843 PMCID: PMC8570701 DOI: 10.1016/s2665-9913(21)00316-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background Patients with primary systemic vasculitis or polymyalgia rheumatica might be at a high risk for poor COVID-19 outcomes due to the treatments used, the potential organ damage cause by primary systemic vasculitis, and the demographic factors associated with these conditions. We therefore aimed to investigate factors associated with COVID-19 outcomes in patients with primary systemic vasculitis or polymyalgia rheumatica. Methods In this retrospective cohort study, adult patients (aged ≥18 years) diagnosed with COVID-19 between March 12, 2020, and April 12, 2021, who had a history of primary systemic vasculitis (antineutrophil cytoplasmic antibody [ANCA]-associated vasculitis, giant cell arteritis, Behçet's syndrome, or other vasculitis) or polymyalgia rheumatica, and were reported to the COVID-19 Global Rheumatology Alliance registry were included. To assess COVID-19 outcomes in patients, we used an ordinal COVID-19 severity scale, defined as: (1) no hospitalisation; (2) hospitalisation without supplemental oxygen; (3) hospitalisation with any supplemental oxygen or ventilation; or (4) death. Multivariable ordinal logistic regression analyses were used to estimate odds ratios (ORs), adjusting for age, sex, time period, number of comorbidities, smoking status, obesity, glucocorticoid use, disease activity, region, and medication category. Analyses were also stratified by type of rheumatic disease. Findings Of 1202 eligible patients identified in the registry, 733 (61·0%) were women and 469 (39·0%) were men, and their mean age was 63·8 years (SD 17·1). A total of 374 (31·1%) patients had polymyalgia rheumatica, 353 (29·4%) had ANCA-associated vasculitis, 183 (15·2%) had giant cell arteritis, 112 (9·3%) had Behçet's syndrome, and 180 (15·0%) had other vasculitis. Of 1020 (84·9%) patients with outcome data, 512 (50·2%) were not hospitalised, 114 (11·2%) were hospitalised and did not receive supplemental oxygen, 239 (23·4%) were hospitalised and received ventilation or supplemental oxygen, and 155 (15·2%) died. A higher odds of poor COVID-19 outcomes were observed in patients who were older (per each additional decade of life OR 1·44 [95% CI 1·31-1·57]), were male compared with female (1·38 [1·05-1·80]), had more comorbidities (per each additional comorbidity 1·39 [1·23-1·58]), were taking 10 mg/day or more of prednisolone compared with none (2·14 [1·50-3·04]), or had moderate, or high or severe disease activity compared with those who had disease remission or low disease activity (2·12 [1·49-3·02]). Risk factors varied among different disease subtypes. Interpretation Among patients with primary systemic vasculitis and polymyalgia rheumatica, severe COVID-19 outcomes were associated with variable and largely unmodifiable risk factors, such as age, sex, and number of comorbidities, as well as treatments, including high-dose glucocorticoids. Our results could be used to inform mitigation strategies for patients with these diseases. Funding American College of Rheumatology and the European Alliance of Associations for Rheumatology.
Collapse
Affiliation(s)
- Sebastian E Sattui
- Department of Medicine, Division of Rheumatology, Hospital for Special Surgery, New York, NY, USA
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Richard Conway
- Department of Rheumatology, St James's Hospital, Dublin, Ireland
| | | | - Andrea M Seet
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, CA, USA
| | - Milena A Gianfrancesco
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, CA, USA
| | | | - Catherine Hill
- Rheumatology Unit, The Queen Elizabeth Hospital, Woodville, SA, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - David Liew
- Department of Rheumatology, Austin Health, Melbourne, Australia
- Department of Medicine, University of Melbourne, VIC, Australia
| | - Sarah L Mackie
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Puja Mehta
- Centre for Inflammation and Tissue Repair, UCL Respiratory, University College London, UK
- Department of Rheumatology, University College London Hospitals NHS Foundation Trust, London, UK
| | - Lorna Neill
- Polymyalgia Rheumatica and Giant Cell Arteritis Scotland, Perth, Scotland, UK
| | - Gimena Gomez
- Research Unit Argentine Society of Rheumatology, Buenos Aires, Argentina
| | | | | | | | | | - Nafice Costa Araujo
- Instituto de Assistencia Medica ao Servidor Publico Estadual de Sao Paulo, Sao Paulo, Brazil
| | - Ann Knight
- Rheumatology, Institute of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Davide Rozza
- Epidemiology Research Unit, Italian Society for Rheumatology, Milan, Italy
| | - Luca Quartuccio
- Clinic of Rheumatology, Department of Medicine, University of Udine, School of Rheumatology, Santa Maria della Misericordia Academic Hospital, Udine, Italy
| | - Maxime Samson
- Department of Internal Medicine and Clinical Immunology, Dijon University Hospital, Dijon, France
| | - Stéphane Bally
- Nephrology and Dialysis Service, Metropole Savoie Hospital Center, Chambery, France
| | - Alexandre Tj Maria
- Department of Internal Medicine and Multi-Organic Diseases, Saint-Eloi University Hospital of Montpellier, Montpellier, France
| | - Pascal Chazerain
- Department of Rheumatology and Internal Medicine, Diaconesses Croix Saint Simon Hospital, Paris, France
| | - Rebecca Hasseli
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus Liebig University Giessen, Bad Nauheim, Germany
| | - Ulf Müller-Ladner
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus Liebig University Giessen, Bad Nauheim, Germany
| | - Bimba F Hoyer
- Department of Rheumatology and Clinical Immunology, Clinic for Internal Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Reinhard Voll
- Department of Rheumatology and Clinical Immunology, University Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Germany
| | - Rita Pinheiro Torres
- CEDOC, Nova Medical School, Lisbon, Portugal
- Rheumatology Service, Egas Moniz Hospital, Lisboa Occidental Hospital Centre, Lisbon, Portugal
| | - Mariana Luis
- Department of Rheumatology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- School of Medicine, Universidade de Coimbra, Coimbra, Portugal
| | | | | | - Jeffrey A Sparks
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tiffany Y-T Hsu
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristin M D'Silva
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Naomi J Patel
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Leanna Wise
- Los Angeles County Hospital, Los Angeles, CA, USA
- University of South California Medical Center, Los Angeles, CA, USA
| | - Emily Gilbert
- Division of Rheumatology, Mayo Clinic Health System, Jacksonville, FL, USA
| | | | - Alí Duarte-García
- Division of Rheumatology, Mayo Clinic Health System, Rochester, MN, USA
| | - Manuel Ugarte-Gil
- School of Medicine, University Cientifica del Sur, Lima, Peru
- Rheumatology Department, Hospital Guillermo Almenara Irigoyen, EsSalud, Lima, Peru
| | - Lindsay Jacobsohn
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, CA, USA
| | - Zara Izadi
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, CA, USA
| | - Anja Strangfeld
- German Rheumatism Research Center, Epidemiology and Health Care Research, Berlin, Germany
| | - Elsa F Mateus
- Portuguese League Against Rheumatic Diseases, Lisbon, Portugal
| | - Kimme L Hyrich
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- National Institute of Health Research Manchester Biomedical Research Centre, University of Manchester, Manchester, UK
- National Institute of Health Research Manchester Biomedical Research Centre, Manchester University NHS Trust, Manchester, UK
| | - Laure Gossec
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris France
- Department of Rheumatology, Pitié-Salpêtrière Hospital, AP-HP, Sorbonne Université, Paris, France
| | | | - Saskia Lawson-Tovey
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, University of Manchester, Manchester, UK
- Manchester Academic Health Science Centre, Manchester, UK
| | | | - Martin Schaefer
- German Rheumatism Research Center, Epidemiology and Health Care Research, Berlin, Germany
| | - Emily Sirotich
- McMaster University, Hamilton, ON, Canada
- Canadian Arthritis Patient Alliance, Toronto, ON, Canada
| | - Jonathan S Hausmann
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Boston Children's Hospital, Boston, MA, USA
| | | | | | - Jean W Liew
- Boston University School of Medicine, Boston, MA, USA
| | | | - Pedro M Machado
- Centre for Rheumatology and Department of Neuromuscular Diseases, University College London, UK
- National Institute for Health Research, University College London Hospitals Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK
- Department of Rheumatology, Northwick Park Hospital, London North West University Healthcare NHS Trust, London, UK
| | - Zachary S Wallace
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jinoos Yazdany
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, CA, USA
| | - Philip C Robinson
- University of Queensland, Brisbane, QLD, Australia
- Royal Brisbane and Women's Hospital, Metro North Hospital and Health Service, Herston, QLD, Australia
| |
Collapse
|
519
|
Assante R, D'Antonio A, Mannarino T, Gaudieri V, Zampella E, Mainolfi CG, Cantoni V, Green R, Caiazzo E, Nappi C, Criscuolo E, Bologna R, Zumbo G, Petretta M, Cuocolo A, Acampa W. Impact of COVID-19 infection on short-term outcome in patients referred to stress myocardial perfusion imaging. Eur J Nucl Med Mol Imaging 2021; 49:1544-1552. [PMID: 34773166 PMCID: PMC8589632 DOI: 10.1007/s00259-021-05619-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/05/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE We assessed the impact of COVID-19 infection on cardiovascular events in patients with suspected or known coronary artery disease (CAD) referred to stress single-photon emission computed tomography myocardial perfusion imaging (MPS). METHODS A total of 960 consecutive patients with suspected or known CAD were submitted by referring physicians to stress MPS for assessment of myocardial ischemia between January 2018 and June 2019. All patients underwent stress-optional rest MPS. Perfusion defects were quantitated as % of LV myocardium and expressed as total perfusion defect (TPD), representing the defect extent and severity. A TPD ≥ 5% was considered abnormal. RESULTS During a mean follow-up of 27 months (range 4-38) 31 events occurred. Moreover, 55 (6%) patients had a COVID-19 infection. The median time from index MPS to COVID-19 infection was 16 months (range 6-24). At Cox multivariable analysis, abnormal MPS and COVID-19 infection resulted as independent predictors of events. There were no significant differences in annualized event rate in COVID-19 patients with or without abnormal MPS (p = 0.56). Differently, in patients without COVID-19, the presence of abnormal MPS was associated with higher event rate (p < .001). Patients with infection compared to those without had a higher event rate in the presence of both normal and abnormal TPD. CONCLUSION In patients with suspected or known CAD, the presence of COVID-19 infection during a short-term follow-up was associated with a higher rate of cardiovascular events.
Collapse
Affiliation(s)
- Roberta Assante
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Adriana D'Antonio
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Teresa Mannarino
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Valeria Gaudieri
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy.
| | - Emilia Zampella
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Ciro Gabriele Mainolfi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Valeria Cantoni
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Roberta Green
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Elisa Caiazzo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Carmela Nappi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Emanuele Criscuolo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Roberto Bologna
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Giulia Zumbo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | | | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Wanda Acampa
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
520
|
Abstract
This article provides an overview of the history of the sepsis definitions as well as an overview of the current understanding of the pathogenesis of sepsis. The evolution of the treatment bundles is also presented.
Collapse
Affiliation(s)
- Michael H Ackerman
- Masters in Healthcare Innovation Program, The Ohio State University, Columbus, OH, USA.
| | - Thomas Ahrens
- Viven Health, 006 Woodbridge Creek Court, St Louis, MO 63129, USA
| | - Justin Kelly
- OSU Wexner Medical Center - The James, 460 West 10th Avenue, Room C1138, Columbus, OH, 43210, USA
| | - Anne Pontillo
- Nursing Education Department, James Cancer Hospital Solove Research Institute, 660 Ackerman Road, 5th Floor/Room 574, Columbus, OH 43202, USA
| |
Collapse
|
521
|
Khwatenge CN, Pate M, Miller LC, Sang Y. Immunometabolic Dysregulation at the Intersection of Obesity and COVID-19. Front Immunol 2021; 12:732913. [PMID: 34737743 PMCID: PMC8560738 DOI: 10.3389/fimmu.2021.732913] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/28/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity prevails worldwide to an increasing effect. For example, up to 42% of American adults are considered obese. Obese individuals are prone to a variety of complications of metabolic disorders including diabetes mellitus, hypertension, cardiovascular disease, and chronic kidney disease. Recent meta-analyses of clinical studies in patient cohorts in the ongoing coronavirus-disease 2019 (COVID-19) pandemic indicate that the presence of obesity and relevant disorders is linked to a more severe prognosis of COVID-19. Given the significance of obesity in COVID-19 progression, we provide a review of host metabolic and immune responses in the immunometabolic dysregulation exaggerated by obesity and the viral infection that develops into a severe course of COVID-19. Moreover, sequela studies of individuals 6 months after having COVID-19 show a higher risk of metabolic comorbidities including obesity, diabetes, and kidney disease. These collectively implicate an inter-systemic dimension to understanding the association between obesity and COVID-19 and suggest an interdisciplinary intervention for relief of obesity-COVID-19 complications beyond the phase of acute infection.
Collapse
Affiliation(s)
- Collins N Khwatenge
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, Nashville, TN, United States
| | - Marquette Pate
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, Nashville, TN, United States
| | - Laura C Miller
- Virus and Prion Research Unit, National Animal Disease Center, United States Department of Agriculture, Agricultural Research Service, Ames, IA, United States
| | - Yongming Sang
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, Nashville, TN, United States
| |
Collapse
|
522
|
Puhm F, Flamand L, Boilard E. Platelet extracellular vesicles in COVID-19: Potential markers and makers. J Leukoc Biol 2021; 111:63-74. [PMID: 34730839 PMCID: PMC8667644 DOI: 10.1002/jlb.3mir0221-100r] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Platelets and platelet extracellular vesicles (pEV) are at the crossroads of coagulation and immunity. Extracellular vesicles are messengers that not only transmit signals between cells, but also provide information about the status of their cell of origin. Thus, pEVs have potential as both biomarkers of platelet activation and contributors to pathology. Coronavirus Disease‐19 (COVID‐19), caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2), is a complex disease affecting multiple organs and is characterized by a high degree of inflammation and risk of thrombosis in some patients. In this review, we introduce pEVs as valuable biomarkers in disease with a special focus on their potential as predictors of and contributors to COVID‐19.
Collapse
Affiliation(s)
- Florian Puhm
- Department of Infectious Diseases and Immunity, Centre de recherche du CHU de Québec, Québec, Québec, Canada.,Université Laval and Centre de recherche ARThrite, Québec, Québec, Canada
| | - Louis Flamand
- Department of Infectious Diseases and Immunity, Centre de recherche du CHU de Québec, Québec, Québec, Canada.,Université Laval and Centre de recherche ARThrite, Québec, Québec, Canada
| | - Eric Boilard
- Department of Infectious Diseases and Immunity, Centre de recherche du CHU de Québec, Québec, Québec, Canada.,Université Laval and Centre de recherche ARThrite, Québec, Québec, Canada
| |
Collapse
|
523
|
Kikuchi K, Nangaku M, Ryuzaki M, Yamakawa T, Yoshihiro O, Hanafusa N, Sakai K, Kanno Y, Ando R, Shinoda T, Nakamoto H, Akizawa T. Survival and predictive factors in dialysis patients with COVID-19 in Japan: a nationwide cohort study. RENAL REPLACEMENT THERAPY 2021; 7:59. [PMID: 34697570 PMCID: PMC8529564 DOI: 10.1186/s41100-021-00378-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/07/2021] [Indexed: 11/30/2022] Open
Abstract
Background The Japanese Association of Dialysis Physicians, the Japanese Society for Dialysis Therapy, and the Japanese Society of Nephrology jointly established COVID-19 Task Force Committee and began surveying the number of newly infected patients. Methods This registry of the COVID-19 Task Force Committee was used to collect data of dialysis patients; a total of 1010 dialysis patients with COVID-19 were included in the analysis. Overall survival of patients was investigated with stratification by age group, complication status, and treatment. In addition, predictive factors for mortality were also investigated. The overall survival was estimated by Kaplan–Meier methods and compared by using log-rank test. Multivariate analysis was performed to identify the risk factor of mortality. For all statistical analyses, p < 0.05 was considered to be statistically significant. Results The mortality risk was increased with age (p < 0.001). The mortality risk was significantly higher in patients with peripheral arterial disease (HR: 1.49, 95% CI 1.05–2.10) and significantly lower in patients who were treated with remdesivir (HR: 0.60, 95% CI 0.37–0.98). Multivariate analysis showed increased risk of mortality with increment in BMI, and increment in CRP, and decreased risk with increment in albumin. Conclusion Dialysis patients have a high severity of illness and a high risk of mortality in cases of COVID-19. Treatment with remdesivir might be effective in shortening the duration of hospitalization and reducing the risk of mortality.
Collapse
Affiliation(s)
- Kan Kikuchi
- Division of Nephrology, Shimoochiai Clinic, 2-1-6 Shimoochiai, Shinjuku-ku, Tokyo, 161-0033 Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Munekazu Ryuzaki
- Department of Nephrology, Tokyo Saiseikai Central Hospital, Tokyo, Japan
| | | | | | - Norio Hanafusa
- Department of Blood Purification, Tokyo Women's Medical University, Tokyo, Japan
| | - Ken Sakai
- Department of Nephrology, Faculty of Medicine,, Toho University, Tokyo, Japan
| | - Yoshihiko Kanno
- Department of Nephrology, Tokyo Medical University, Tokyo, Japan
| | - Ryoichi Ando
- Department of Nephrology, Seishokai Memorial Hospital, Tokyo, Japan
| | - Toshio Shinoda
- Faculty of Medical and Health Sciences, Tsukuba International University, Tsuchiura, Japan
| | - Hidetomo Nakamoto
- Department of General Internal Medicine, Saitama Medical University, Iruma, Japan
| | - Tadao Akizawa
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
524
|
Abstract
In 2019 first reports about a new human coronavirus emerged, which causes common cold symptoms as well as acute respiratory distress syndrome. The virus was identified as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and severe thrombotic events including deep vein thrombosis, pulmonary embolism, and microthrombi emerged as additional symptoms. Heart failure, myocardial infarction, myocarditis, and stroke have also been observed. As main mediator of thrombus formation, platelets became one of the key aspects in SARS-CoV-2 research. Platelets may also directly interact with SARS-CoV-2 and have been shown to carry the SARS-CoV-2 virus. Platelets can also facilitate the virus uptake by secretion of the subtilisin-like proprotein convertase furin. Cleavage of the SARS-CoV-2 spike protein by furin enhances binding capabilities and virus entry into various cell types. In COVID-19 patients, platelet count differs between mild and serious infections. Patients with mild symptoms have a slightly increased platelet count, whereas thrombocytopenia is a hallmark of severe COVID-19 infections. Low platelet count can be attributed to platelet apoptosis and the incorporation of platelets into microthrombi (peripheral consumption) and severe thrombotic events. The observed excessive formation of thrombi is due to hyperactivation of platelets caused by the infection. Various factors have been suggested in the activation of platelets in COVID-19, such as hypoxia, vessel damage, inflammatory factors, NETosis, SARS-CoV-2 interaction, autoimmune reactions, and autocrine activation. COVID-19 does alter chemokine and cytokine plasma concentrations. Platelet chemokine profiles are altered in COVID-19 and contribute to the described chemokine storms observed in severely ill COVID-19 patients.
Collapse
Affiliation(s)
- Anne-Katrin Rohlfing
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls Universtität Tübingen, Tübingen, Germany
| | - Dominik Rath
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls Universtität Tübingen, Tübingen, Germany
| | - Tobias Geisler
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls Universtität Tübingen, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Cardiology and Angiology, University Hospital Tübingen, Eberhard Karls Universtität Tübingen, Tübingen, Germany
| |
Collapse
|
525
|
Riera-Mestre A, Jara-Palomares L, Lecumberri R, Trujillo-Santos J, Grau E, Blanco-Molina A, Piera Carbonell A, Jiménez S, Frías Vargas M, Fuset MP, Bellmunt-Montoya S, Monreal M, Jiménez D. PICO Questions and DELPHI Methodology for the Management of Venous Thromboembolism Associated with COVID-19. Viruses 2021; 13:2128. [PMID: 34834935 PMCID: PMC8624706 DOI: 10.3390/v13112128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/16/2022] Open
Abstract
Patients with coronavirus disease 2019 (COVID-19) have a higher risk of venous thromboembolic disease (VTE) than patients with other infectious or inflammatory diseases, both as macrothrombosis (pulmonar embolism and deep vein thrombosis) or microthrombosis. However, the use of anticoagulation in this scenario remains controversial. This is a project that used DELPHI methodology to answer PICO questions related to anticoagulation in patients with COVID-19. The objective was to reach a consensus among multidisciplinary VTE experts providing answers to those PICO questions. Seven PICO questions regarding patients with COVID-19 responded with a broad consensus: 1. It is recommended to avoid pharmacological thromboprophylaxis in most COVID-19 patients not requiring hospital admission; 2. In most hospitalized patients for COVID-19 who are receiving oral anticoagulants before admission, it is recommended to replace them by low molecular weight heparin (LMWH) at therapeutic doses; 3. Thromboprophylaxis with LMWH at standard doses is suggested for COVID-19 patients admitted to a conventional hospital ward; 4. Standard-doses thromboprophylaxis with LMWH is recommended for COVID-19 patients requiring admission to Intensive Care Unit; 5. It is recommended not to determine D-Dimer levels routinely in COVID-19 hospitalized patients to select those in whom VTE should be suspected, or as a part of the diagnostic algorithm to rule out or confirm a VTE event; 6. It is recommended to discontinue pharmacological thromboprophylaxis at discharge in most patients hospitalized for COVID-19; 7. It is recommended to withdraw anticoagulant treatment after 3 months in most patients with a VTE event associated with COVID-19. The combination of PICO questions and DELPHI methodology provides a consensus on different recommendations for anticoagulation management in patients with COVID-19.
Collapse
Affiliation(s)
- Antoni Riera-Mestre
- Internal Medicine Department, Hospital Universitari de Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), 08907 Barcelona, Spain
- Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907 Barcelona, Spain
| | - Luis Jara-Palomares
- Medical-Surgical Unit for Respiratory Diseases, Virgen del Rocío University Hospital, 28029 Sevilla, Spain;
- Center for Biomedical Research in Respiratory Diseases Network (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Ramón Lecumberri
- Hematology Department, Clínica Universidad de Navarra, 31008 Pamplona, Spain;
- Center for Biomedical Research Network on Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Trujillo-Santos
- Internal Medicine Department, Hospital General Universitario Santa Lucía, 30204 Cartagena, Spain;
- Faculty of Health Sciences, Universidad Católica San Antonio de Murcia (UCAM), 30107 Murcia, Spain;
| | - Enric Grau
- Hematology Department, Lluis Alcanyis de Xativa Hospital, 46800 Valencia, Spain;
| | | | | | - Sonia Jiménez
- Emergency Area, Hospital Clínic, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
| | | | - Mari Paz Fuset
- Intensive Medicine Department, Hospital Universitari de Bellvitge, Bellvitge Biomedical Research Institute (IDIBELL), 08907 Barcelona, Spain;
| | - Sergi Bellmunt-Montoya
- Angiology and Vascular Surgery Department, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain;
- Surgery Department, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain
| | - Manuel Monreal
- Faculty of Health Sciences, Universidad Católica San Antonio de Murcia (UCAM), 30107 Murcia, Spain;
- Internal Medicine Department, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - David Jiménez
- Center for Biomedical Research in Respiratory Diseases Network (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Pulmonology Department, Ramón y Cajal Hospital (IRYCIS), 28034 Madrid, Spain
- Department of Medicine, Alcala University, 28805 Madrid, Spain
| | | |
Collapse
|
526
|
Zhou Y, Xu X, Wei H. Complex Pathophysiological Mechanisms and the Propose of the Three-Dimensional Schedule For Future COVID-19 Treatment. Front Immunol 2021; 12:716940. [PMID: 34745094 PMCID: PMC8564179 DOI: 10.3389/fimmu.2021.716940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 10/05/2021] [Indexed: 11/13/2022] Open
Abstract
At present, the global COVID-19 epidemic is still in a state of anxiety, and increasing the cure rate of critically ill patients is an important means to defeat the virus. From an immune perspective, ARDS driven by an inflammatory storm is still the direct cause of death in severe COVID-19 patients. Although some experience has been gained in the treatment of COVID-19, and intensive COVID-19 vaccination has been carried out recently, it is still effective to save lives to develop more effective programs to alleviate the inflammatory storm and ARDS in patients with SARS-CoV-2 or emerging variants of SARS-CoV-2. In reorganizing the ARDS-related inflammatory storm formation program in COVID-19 patients, we highlighted the importance of the vicious circle of inflammatory cytokines and inflammatory cell death, which is aggravated by blood circulation to form multi-system inflammation. Summarizes the interlocking and crisscrossing of inflammatory response and inflammatory cell death mechanisms including NETs, pyrolysis, apoptosis and PANoptosis in severe COVID-19. More importantly, in response to the inflammatory storm formation program we described, and on the premise of following ethical and clinical experimental norms, we propose a three-dimensional integrated program for future research based on boosting antiviral immune response at the initial stage, inhibiting inflammatory cytokine signaling at the exacerbation stage and inhibiting cell death before it's worse to prevent and alleviate ARDS.
Collapse
Affiliation(s)
- Yonggang Zhou
- Institute of Gerontology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Laboratory for Physical Sciences at Microscale, The Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xiuxiu Xu
- Hefei National Laboratory for Physical Sciences at Microscale, The Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- Institute of Gerontology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Laboratory for Physical Sciences at Microscale, The Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| |
Collapse
|
527
|
Xu Z, Huang Y, Zhou J, Deng X, He W, Liu X, Li Y, Zhong N, Sang L. Current Status of Cell-Based Therapies for COVID-19: Evidence From Mesenchymal Stromal Cells in Sepsis and ARDS. Front Immunol 2021; 12:738697. [PMID: 34659231 PMCID: PMC8517471 DOI: 10.3389/fimmu.2021.738697] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/13/2021] [Indexed: 12/29/2022] Open
Abstract
The severe respiratory consequences of the coronavirus disease 2019 (COVID-19) pandemic have prompted the urgent need for novel therapies. Cell-based therapies, primarily using mesenchymal stromal cells (MSCs), have demonstrated safety and potential efficacy in the treatment of critical illness, particularly sepsis and acute respiratory distress syndrome (ARDS). However, there are limited preclinical data for MSCs in COVID-19. Recent studies have shown that MSCs could decrease inflammation, improve lung permeability, enhance microbe and alveolar fluid clearance, and promote lung epithelial and endothelial repair. In addition, MSC-based therapy has shown promising effects in preclinical studies and phase 1 clinical trials in sepsis and ARDS. Here, we review recent advances related to MSC-based therapy in the context of sepsis and ARDS and evaluate the potential value of MSCs as a therapeutic strategy for COVID-19.
Collapse
Affiliation(s)
- Zhiheng Xu
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Medical University, Guangzhou, China
| | - Yongbo Huang
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Medical University, Guangzhou, China
| | - Jianmeng Zhou
- School of Public Health, Southern Medical University, Guangzhou, China
| | - Xiumei Deng
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Medical University, Guangzhou, China
| | - Weiqun He
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Medical University, Guangzhou, China
| | - Xiaoqing Liu
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Medical University, Guangzhou, China
| | - Yimin Li
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Medical University, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Medical University, Guangzhou, China
| | - Ling Sang
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Medical University, Guangzhou, China.,Guangzhou Laboratory, Guangzhou, China
| |
Collapse
|
528
|
Ai J, Hong W, Wu M, Wei X. Pulmonary vascular system: A vulnerable target for COVID-19. MedComm (Beijing) 2021; 2:531-547. [PMID: 34909758 PMCID: PMC8662299 DOI: 10.1002/mco2.94] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 02/05/2023] Open
Abstract
The number of coronavirus disease 2019 (COVID‐19) cases has been increasing significantly, and the disease has evolved into a global pandemic, posing an unprecedented challenge to the healthcare community. Angiotensin‐converting enzyme 2, the binding and entry receptor of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) in hosts, is also expressed on pulmonary vascular endothelium; thus, pulmonary vasculature is a potential target in COVID‐19. Indeed, pulmonary vascular thickening is observed by early clinical imaging, implying a tropism of SARS‐CoV‐2 for pulmonary vasculature. Recent studies reported that COVID‐19 is associated with vascular endothelial damage and dysfunction along with inflammation, coagulopathy, and microthrombosis; all of these pathologic changes are the hallmarks of pulmonary vascular diseases. Notwithstanding the not fully elucidated effects of COVID‐19 on pulmonary vasculature, the vascular endotheliopathy that occurs after infection is attributed to direct infection and indirect damage mainly caused by renin‐angiotensin‐aldosterone system imbalance, coagulation cascade, oxidative stress, immune dysregulation, and intussusceptive angiogenesis. Degradation of endothelial glycocalyx exposes endothelial cell (EC) surface receptors to the vascular lumen, which renders pulmonary ECs more susceptible to SARS‐CoV‐2 infection. The present article reviews the potential pulmonary vascular pathophysiology and clinical presentations in COVID‐19 to provide a basis for clinicians and scientists, providing insights into the development of therapeutic strategies targeting pulmonary vasculature.
Collapse
Affiliation(s)
- Jiayuan Ai
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu Sichuan PR China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu Sichuan PR China
| | - Min Wu
- Department of Biomedical Sciences School of Medicine and Health Sciences University of North Dakota Grand Forks North Dakota USA
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu Sichuan PR China
| |
Collapse
|
529
|
Hertanto DM, Sutanto H, Adi S. Case Report: Diabetic nephropathy aggravates the progression and prognosis of COVID-19-associated acute limb ischemia. F1000Res 2021; 10:584. [PMID: 34642600 PMCID: PMC8430448 DOI: 10.12688/f1000research.54193.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/10/2021] [Indexed: 12/23/2022] Open
Abstract
Hypercoagulation is a hallmark of both the novel coronavirus disease (COVID-19) and type 2 diabetes mellitus (T2DM). It increases the risk for vascular thrombosis, including peripheral artery diseases. Among others, acute limb ischemia (ALI) is one of most common complications that requires immediate and prompt treatments to reduce morbidity and mortality. However, the complex interplay between COVID-19, T2DM and its complications (e.g., diabetic nephropathy), and ALI creates a great challenge in the management of the disease. Here, we present a case of a 59-year-old diabetic female with progressive pain in her left leg in the last five years, which was significantly intensified following COVID-19 diagnosis. Bluish coloration, numbness and functional impairments were observed during examinations with no palpable pulsation on left posterior tibial and dorsalis pedis arteries. The patient also had diabetic nephropathy (stage III), hypoalbuminemia, anemia and a urinary tract infection that complicated the management of the disease. Due to the excruciating pain and the worsening of the limb conditions, right leg revascularization and left leg amputation were performed at day 14 after admission. Following the surgeries, no more pain was observed and patient was discharged for further follow-up at the outpatient clinic.
Collapse
Affiliation(s)
- Decsa Medika Hertanto
- Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya, Jawa Timur, 60132, Indonesia.,Department of Internal Medicine, Dr. Soetomo Hospital, Surabaya, Jawa Timur, 60286, Indonesia
| | - Henry Sutanto
- Department of Cardiology, Maastricht University, Maastricht, Limburg, 6229ER, The Netherlands.,Department of Physiology and Pharmacology, SUNY Downstate Medical Center, Brooklyn, New York, 11203, USA
| | - Soebagijo Adi
- Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya, Jawa Timur, 60132, Indonesia.,Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Dr. Soetomo Hospital, Surabaya, Jawa Timur, 60286, Indonesia
| |
Collapse
|
530
|
SARS-CoV-2 and Atherosclerosis: Should COVID-19 Be Recognized as a New Predisposing Cardiovascular Risk Factor? J Cardiovasc Dev Dis 2021; 8:jcdd8100130. [PMID: 34677199 PMCID: PMC8539652 DOI: 10.3390/jcdd8100130] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/27/2021] [Accepted: 10/08/2021] [Indexed: 12/18/2022] Open
Abstract
At the beginning of the COVID-19 pandemic, the lung was recognized as the main target organ; now, new evidence suggests that SARS-CoV-2 infection leads to vascular disease. In a previous review, we supposed a bidirectional link between endothelial dysfunction and COVID-19, identifying atherosclerosis as having a crucial role in its pathogenesis. Atherosclerosis with an existing endothelial dysfunction may worsen COVID-19 manifestations, leading to adverse outcomes, as largely reported. However, COVID-19 may be the trigger factor in the progression of the atherosclerotic process up to making it clinically manifest. The thrombotic complications can involve not only the atherosclerotic plaque, but also the durability of the surgical device implanted to treat a pre-existing coronary artery disease as recently reported. The burden of the disease makes necessary a long-term stratification of patients, revising drastically targeted therapy among others.
Collapse
|
531
|
Varela AA, Cheng S, Werren JH. Novel ACE2 protein interactions relevant to COVID-19 predicted by evolutionary rate correlations. PeerJ 2021; 9:e12159. [PMID: 34616619 PMCID: PMC8449537 DOI: 10.7717/peerj.12159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is the cell receptor that the coronavirus SARS-CoV-2 binds to and uses to enter and infect human cells. COVID-19, the pandemic disease caused by the coronavirus, involves diverse pathologies beyond those of a respiratory disease, including micro-thrombosis (micro-clotting), cytokine storms, and inflammatory responses affecting many organ systems. Longer-term chronic illness can persist for many months, often well after the pathogen is no longer detected. A better understanding of the proteins that ACE2 interacts with can reveal information relevant to these disease manifestations and possible avenues for treatment. We have undertaken an approach to predict candidate ACE2 interacting proteins which uses evolutionary inference to identify a set of mammalian proteins that “coevolve” with ACE2. The approach, called evolutionary rate correlation (ERC), detects proteins that show highly correlated evolutionary rates during mammalian evolution. Such proteins are candidates for biological interactions with the ACE2 receptor. The approach has uncovered a number of key ACE2 protein interactions of potential relevance to COVID-19 pathologies. Some proteins have previously been reported to be associated with severe COVID-19, but are not currently known to interact with ACE2, while additional predicted novel ACE2 interactors are of potential relevance to the disease. Using reciprocal rankings of protein ERCs, we have identified strongly interconnected ACE2 associated protein networks relevant to COVID-19 pathologies. ACE2 has clear connections to coagulation pathway proteins, such as Coagulation Factor V and fibrinogen components FGA, FGB, and FGG, the latter possibly mediated through ACE2 connections to Clusterin (which clears misfolded extracellular proteins) and GPR141 (whose functions are relatively unknown). ACE2 also connects to proteins involved in cytokine signaling and immune response (e.g. XCR1, IFNAR2 and TLR8), and to Androgen Receptor (AR). The ERC prescreening approach has elucidated possible functions for relatively uncharacterized proteins and possible new functions for well-characterized ones. Suggestions are made for the validation of ERC-predicted ACE2 protein interactions. We propose that ACE2 has novel protein interactions that are disrupted during SARS-CoV-2 infection, contributing to the spectrum of COVID-19 pathologies.
Collapse
Affiliation(s)
- Austin A Varela
- Department of Biology, University of Rochester, Rochester, New York, United States
| | - Sammy Cheng
- Department of Biology, University of Rochester, Rochester, New York, United States
| | - John H Werren
- Department of Biology, University of Rochester, Rochester, New York, United States
| |
Collapse
|
532
|
Vecchié A, Batticciotto A, Tangianu F, Bonaventura A, Pennella B, Abenante A, Corso R, Grazioli S, Mumoli N, Para O, Maresca AM, Dalla Gasperina D, Dentali F. High-dose dexamethasone treatment for COVID-19 severe acute respiratory distress syndrome: a retrospective study. Intern Emerg Med 2021; 16:1913-1919. [PMID: 34275096 PMCID: PMC8286167 DOI: 10.1007/s11739-021-02800-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/27/2021] [Indexed: 12/15/2022]
Abstract
Low-dose dexamethasone reduces mortality in patients with coronavirus disease 2019 (COVID-19)-related acute respiratory distress syndrome (ARDS). We retrospectively analyzed the efficacy of high-dose dexamethasone in patients with COVID-19-related ARDS and evaluated factors affecting the composite outcome (death or invasive mechanical ventilation). From March 4th to April 1st 2020, 98 patients with COVID-19 pneumonia were included. Those who after at least 7 days from symptom onset presented a worsening of the respiratory function or of inflammatory biomarkers were started on intravenous high-dose dexamethasone (20 mg daily for 5 days, followed by 10 mg daily for 5 days). Most patients were males (62%) with a mean age of 69 years. Hypertension and cardiovascular disease (CVD) were prevalent. Following dexamethasone treatment, a significant improvement in PaO2/FiO2 (277.41 [178.5-374.8] mmHg vs. 146.75 [93.62-231.16] mmHg, p < 0.001), PaO2 (88.15 [76.62-112.0] mmHg vs. 65.65 [57.07-81.22] mmHg, p < 0.001), and SpO2 (96 [95-98]% vs. 94 [90-96]%, p < 0.001) was observed. A concomitant decrease in C-reactive protein and ferritin levels was found (132.25 [82.27-186.5] mg/L vs. 7.3 [3.3-24.2] mg/L and 1169 [665-2056] ng/mL vs. 874.0 [569.5-1434] ng/mL, respectively; p < 0.001 for both vs. baseline). CVD was found to increase the risk of the composite outcome (RR 7.64, 95% CI 1.24-47.06, p = 0.028). In hospitalized patients with COVID-19-related ARDS, high-dose dexamethasone rapidly improves the clinical status and decreases inflammatory biomarkers. CVD was found to increase the risk of the composite outcome. These data support the importance of randomized clinical trials with high-dose dexamethasone in COVID-19 patients.
Collapse
Affiliation(s)
| | | | - Flavio Tangianu
- Department of Internal Medicine, ASST Sette Laghi, Varese, Italy
| | - Aldo Bonaventura
- Department of Internal Medicine, ASST Sette Laghi, Varese, Italy
| | | | - Alessia Abenante
- Department of Internal Medicine, ASST Sette Laghi, Varese, Italy
| | - Rossana Corso
- Department of Internal Medicine, ASST Sette Laghi, Varese, Italy
| | - Stefano Grazioli
- Department of Internal Medicine, ASST Sette Laghi, Varese, Italy
| | - Nicola Mumoli
- Department of Internal Medicine, Ospedale di Magenta, ASST Ovest Milanese, Legnano, Italy
| | - Ombretta Para
- SOD Medicina Interna I, Azienda Ospedaliero-Universitaria Careggi, Firenze, Italy
| | | | | | - Francesco Dentali
- Department of Medicine and Surgery, Insubria University, Varese, Italy.
| |
Collapse
|
533
|
Endothelial Activation and Stress Index (EASIX) as an Early Predictor for Mortality and Overall Survival in Hematological and Non-Hematological Patients with COVID-19: Multicenter Cohort Study. J Clin Med 2021; 10:jcm10194373. [PMID: 34640390 PMCID: PMC8509351 DOI: 10.3390/jcm10194373] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/27/2022] Open
Abstract
COVID-19, as a disease involving the endothelium of multiple organs, is characterized by high mortality rates among hospitalized patients. Patients with hematological malignancies are particularly at risk of an unfavorable course of COVID-19. The endothelial activation and stress index (EASIX) score has been used as a simple predictor of overall survival (OS) in specific groups of hematological cancer patients. EASIX, as a biomarker of endothelial dysfunction, might play a prognostic role in patients with COVID-19. Here, we performed a comprehensive retrospective analysis of the EASIX score in 523 hospitalized COVID-19 patients with or without coexisting hematological cancer. Hematological cancer COVID-19 patients had higher EASIX scores compared to the overall population with COVID-19. In hematological patients, EASIX was a strong predictor of the occurrence of sepsis during COVID-19. Our findings demonstrated EASIX as a strong predictor of intensive care unit admission, in-hospital mortality, the occurrence of acute renal failure and the need for hemodialysis, both in hematological and non-hematological COVID-19 patients. Patients with a high EASIX score on COVID-19 diagnosis had significantly inferior OS compared to patients with low EASIX. We showed for the first time that EASIX might serve as a simple, universal prognostic tool of OS in both hematological and non-hematological COVID-19 patients.
Collapse
|
534
|
Philadelphia-Negative Chronic Myeloproliferative Neoplasms during the COVID-19 Pandemic: Challenges and Future Scenarios. Cancers (Basel) 2021; 13:cancers13194750. [PMID: 34638236 PMCID: PMC8507529 DOI: 10.3390/cancers13194750] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/09/2021] [Accepted: 09/17/2021] [Indexed: 12/30/2022] Open
Abstract
An outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) started in December 2019 in China and then become pandemic in February 2020. Several publications investigated the possible increased rate of COVID-19 infection in hematological malignancies. Based on the published data, strategies for the management of chronic Philadelphia-negative chronic myeloproliferative neoplasms (MPNs) are provided. The risk of severe COVID-19 seems high in MPN, particularly in patients with essential thrombocythemia, but not negligible in myelofibrosis. MPN patients are at high risk of both thrombotic and hemorrhagic complications and this must be accounted in the case of COVID-19 deciding on a case-by-case basis. There are currently no data to suggest that hydroxyurea or interferon may influence the risk or severity of COVID-19 infection. Conversely, while the immunosuppressive activity of ruxolitinib might pose increased risk of infection, its abrupt discontinuation during COVID-19 syndrome is associated with worse outcome. All MPN patients should receive vaccine against COVID-19; reassuring data are available on efficacy of mRNA vaccines in MPNs.
Collapse
|
535
|
Bortolotti D, Gentili V, Rizzo S, Schiuma G, Beltrami S, Spadaro S, Strazzabosco G, Campo G, Carosella ED, Papi A, Rizzo R, Contoli M. Increased sHLA-G Is Associated with Improved COVID-19 Outcome and Reduced Neutrophil Adhesion. Viruses 2021; 13:1855. [PMID: 34578436 PMCID: PMC8473385 DOI: 10.3390/v13091855] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/17/2022] Open
Abstract
Human leukocyte antigen (HLA) is a group of molecules involved in inflammatory and infective responses. We evaluated blood sHLA-E and sHLA-G levels in hospitalized COVID-19 patients with respiratory failure and their relationship with clinical evolution, changes in endothelial activation biomarker profile, and neutrophil adhesion. sHLA-E, sHLA-G, and endothelial activation biomarkers were quantified by ELISA assay in plasma samples. Neutrophil adhesion to endothelium was assessed in the presence/absence of patients' plasma samples. At admission, plasma levels of sHLA-G and sHLA-E were significantly higher in COVID-19 patients with respiratory failure compared to controls. COVID-19 clinical improvement was associated with increased sHLA-G plasma levels. In COVID-19, but not in control patients, an inverse correlation was found between serum sICAM-1 and E-selectin levels and plasma sHLA-G values. The in vitro analysis of activated endothelial cells confirmed the ability of HLA-G molecules to control sICAM-1 and sE-selectin expression via CD160 interaction and FGF2 induction and consequently neutrophil adhesion. We suggest a potential role for sHLA-G in improving COVID-19 patients' clinical condition related to the control of neutrophil adhesion to activated endothelium.
Collapse
Affiliation(s)
- Daria Bortolotti
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy; (D.B.); (V.G.); (S.R.); (G.S.); (S.B.); (G.S.)
| | - Valentina Gentili
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy; (D.B.); (V.G.); (S.R.); (G.S.); (S.B.); (G.S.)
| | - Sabrina Rizzo
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy; (D.B.); (V.G.); (S.R.); (G.S.); (S.B.); (G.S.)
| | - Giovanna Schiuma
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy; (D.B.); (V.G.); (S.R.); (G.S.); (S.B.); (G.S.)
| | - Silvia Beltrami
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy; (D.B.); (V.G.); (S.R.); (G.S.); (S.B.); (G.S.)
| | - Savino Spadaro
- Intensive Care Unit, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy;
| | - Giovanni Strazzabosco
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy; (D.B.); (V.G.); (S.R.); (G.S.); (S.B.); (G.S.)
| | - Gianluca Campo
- Cardiology Unit, Azienda Ospedaliero Universitaria di Ferrara, Cona, 44124 Ferrara, Italy;
| | - Edgardo D. Carosella
- CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI), Research Division in Hematology and Immunology (SRHI), Saint-Louis Hospital, 75001 Paris, France;
| | - Alberto Papi
- Respiratory Section, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (A.P.); (M.C.)
- Respiratory Unit, Azienda Ospedaliera Universitaria Ferrara, Cona, 44124 Ferrara, Italy
| | - Roberta Rizzo
- Department of Chemical, Pharmaceutical and Agricultural Science, University of Ferrara, 44121 Ferrara, Italy; (D.B.); (V.G.); (S.R.); (G.S.); (S.B.); (G.S.)
- Industrial Research and Technology Transfer Laboratory (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Marco Contoli
- Respiratory Section, Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (A.P.); (M.C.)
- Respiratory Unit, Azienda Ospedaliera Universitaria Ferrara, Cona, 44124 Ferrara, Italy
| |
Collapse
|
536
|
Alavi P, Rathod AM, Jahroudi N. Age-Associated Increase in Thrombogenicity and Its Correlation with von Willebrand Factor. J Clin Med 2021; 10:4190. [PMID: 34575297 PMCID: PMC8472522 DOI: 10.3390/jcm10184190] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023] Open
Abstract
Endothelial cells that cover the lumen of all blood vessels have the inherent capacity to express both pro and anticoagulant molecules. However, under normal physiological condition, they generally function to maintain a non-thrombogenic surface for unobstructed blood flow. In response to injury, certain stimuli, or as a result of dysfunction, endothelial cells release a highly adhesive procoagulant protein, von Willebrand factor (VWF), which plays a central role in formation of platelet aggregates and thrombus generation. Since VWF expression is highly restricted to endothelial cells, regulation of its levels is among the most important functions of endothelial cells for maintaining hemostasis. However, with aging, there is a significant increase in VWF levels, which is concomitant with a significant rise in thrombotic events. It is not yet clear why and how aging results in increased VWF levels. In this review, we have aimed to discuss the age-related increase in VWF, its potential mechanisms, and associated coagulopathies as probable consequences.
Collapse
Affiliation(s)
| | | | - Nadia Jahroudi
- Department of Medicine, University of Alberta, Edmonton, AB T6G 2S2, Canada; (P.A.); (A.M.R.)
| |
Collapse
|
537
|
Pandolfi S, Chirumbolo S. Home therapy of COVID-19 at the earliest may greatly prevent hospitalization. Basic Clin Pharmacol Toxicol 2021; 129:395-396. [PMID: 34494369 PMCID: PMC8652880 DOI: 10.1111/bcpt.13650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 12/18/2022]
Affiliation(s)
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
538
|
Francischetti IM, Toomer K, Zhang Y, Jani J, Siddiqui Z, Brotman DJ, Hooper JE, Kickler TS. Upregulation of pulmonary tissue factor, loss of thrombomodulin and immunothrombosis in SARS-CoV-2 infection. EClinicalMedicine 2021; 39:101069. [PMID: 34377969 PMCID: PMC8342934 DOI: 10.1016/j.eclinm.2021.101069] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND SARS-CoV-2 infection is associated with thrombotic and microvascular complications. The cause of coagulopathy in the disease is incompletely understood. METHODS A single-center cross-sectional study including 66 adult COVID-19 patients (40 moderate, 26 severe disease), and 9 controls, performed between 04/2020 and 10/2020. Markers of coagulation, endothelial cell function [angiopoietin-1,-2, P-selectin, von Willebrand Factor Antigen (WF:Ag), von Willebrand Factor Ristocetin Cofactor, ADAMTS13, thrombomodulin, soluble Endothelial cell Protein C Receptor (sEPCR), Tissue Factor Pathway Inhibitor], neutrophil activation (elastase, citrullinated histones) and fibrinolysis (tissue-type plasminogen activator, plasminogen activator inhibitor-1) were evaluated using ELISA. Tissue Factor (TF) was estimated by antithrombin-FVIIa complex (AT/FVIIa) and microparticles-TF (MP-TF). We correlated each marker and determined its association with severity. Expression of pulmonary TF, thrombomodulin and EPCR was determined by immunohistochemistry in 9 autopsies. FINDINGS Comorbidities were frequent in both groups, with older age associated with severe disease. All patients were on prophylactic anticoagulants. Three patients (4.5%) developed pulmonary embolism. Mortality was 7.5%. Patients presented with mild alterations in the coagulogram (compensated state). Biomarkers of endothelial cell, neutrophil activation and fibrinolysis were elevated in severe vs moderate disease; AT/FVIIa and MP-TF levels were higher in severe patients. Logistic regression revealed an association of D-dimers, angiopoietin-1, vWF:Ag, thrombomodulin, white blood cells, absolute neutrophil count (ANC) and hemoglobin levels with severity, with ANC and vWF:Ag identified as independent factors. Notably, postmortem specimens demonstrated epithelial expression of TF in the lung of fatal COVID-19 cases with loss of thrombomodulin staining, implying in a shift towards a procoagulant state. INTERPRETATION Coagulation dysregulation has multifactorial etiology in SARS-Cov-2 infection. Upregulation of pulmonary TF with loss of thrombomodulin emerge as a potential link to immunothrombosis, and therapeutic targets in the disease. FUNDING John Hopkins University School of Medicine.
Collapse
Key Words
- ADAMTS13, a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13
- ALC, absolute lymphocyte count
- ALI, Acute Lung Injury
- AMC, absolute monocyte count
- ANC, absolute neutrophil count
- AT/VIIa, antithrombin-FVIIa complex
- Coagulation
- ELISA, enzyme-linked immunosorbent assay
- Hb, hemoglobin
- Hemostasis
- ICU, intensive care unit
- Ixolaris
- LMWH, low molecular weight heparin
- MP-TF, Microparticles-Tissue Factor
- PAI-1, plasminogen activator inhibitor-1
- PAR, protease-activated receptor
- TF, Tissue Factor
- TFPI, Tissue Factor Pathway Inhibitor
- Thrombosis
- WBC, white blood cells
- sEPCR, soluble Endothelial cell Protein C Receptor
- t-PA, tissue-type plasminogen activator
- vWF, von Willebrand Factor
- vWF:Ag, von Willebrand Factor Antigen
- vWF:RCo, von Willebrand Factor Ristocetin Cofactor
Collapse
Affiliation(s)
- Ivo M.B. Francischetti
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Corresponding author.
| | - Kevin Toomer
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yifan Zhang
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Jayesh Jani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zishan Siddiqui
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniel J. Brotman
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jody E. Hooper
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Thomas S. Kickler
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
539
|
Ahearn‐Ford S, Lunjani N, McSharry B, MacSharry J, Fanning L, Murphy G, Everard C, Barry A, McGreal A, Lawati SM, Lapthorne S, Sherlock C, McKeogh A, Jackson A, Faller E, Horgan M, Sadlier C, O'Mahony L. Long-term disruption of cytokine signalling networks is evident in patients who required hospitalization for SARS-CoV-2 infection. Allergy 2021; 76:2910-2913. [PMID: 34028045 PMCID: PMC8222932 DOI: 10.1111/all.14953] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 12/31/2022]
Affiliation(s)
| | | | - Brian McSharry
- APC Microbiome Ireland University College Cork Cork Ireland
- School of Microbiology University College Cork Cork Ireland
| | - John MacSharry
- APC Microbiome Ireland University College Cork Cork Ireland
- School of Microbiology University College Cork Cork Ireland
- Department of Medicine University College Cork Cork Ireland
| | - Liam Fanning
- APC Microbiome Ireland University College Cork Cork Ireland
- Department of Medicine University College Cork Cork Ireland
| | - Gerard Murphy
- Department of Infectious Diseases Cork University Hospital Cork Ireland
| | - Cormac Everard
- Department of Infectious Diseases Cork University Hospital Cork Ireland
| | - Aoife Barry
- Department of Infectious Diseases Cork University Hospital Cork Ireland
| | - Aimee McGreal
- Department of Infectious Diseases Cork University Hospital Cork Ireland
| | | | - Susan Lapthorne
- Department of Infectious Diseases Cork University Hospital Cork Ireland
| | - Colin Sherlock
- Department of Infectious Diseases Cork University Hospital Cork Ireland
| | - Anna McKeogh
- Department of Infectious Diseases Cork University Hospital Cork Ireland
| | - Arthur Jackson
- Department of Infectious Diseases Cork University Hospital Cork Ireland
| | - Eamonn Faller
- Department of Infectious Diseases Cork University Hospital Cork Ireland
| | - Mary Horgan
- Department of Medicine University College Cork Cork Ireland
- Department of Infectious Diseases Cork University Hospital Cork Ireland
| | - Corinna Sadlier
- Department of Infectious Diseases Cork University Hospital Cork Ireland
| | - Liam O'Mahony
- APC Microbiome Ireland University College Cork Cork Ireland
- School of Microbiology University College Cork Cork Ireland
- Department of Medicine University College Cork Cork Ireland
| |
Collapse
|
540
|
Taquet M, Husain M, Geddes JR, Luciano S, Harrison PJ. Cerebral venous thrombosis and portal vein thrombosis: A retrospective cohort study of 537,913 COVID-19 cases. EClinicalMedicine 2021; 39:101061. [PMID: 34368663 PMCID: PMC8324974 DOI: 10.1016/j.eclinm.2021.101061] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND There are concerns about a link between the ChAdOx1 nCoV-19 and Ad26.COV2.S vaccines against COVID-19 and cerebral venous thrombosis (CVT) and other thrombotic events. One key missing component of the risk-benefit analysis of using such vaccines is the risk of these severe thrombotic events following COVID-19. METHODS Using a retrospective cohort study based on electronic health records primarily in the USA, the absolute risks of CVT and portal vein thrombosis (PVT) in the two weeks following a diagnosis of COVID-19 (made between January 20, 2020 and March 25, 2021) were calculated. The risks were compared to cohorts of patients with influenza (diagnosed within the same period) and people receiving an mRNA vaccine (i.e. not the ChAdOx1 nCoV-19 and Ad26.COV2.S vaccines) against COVID-19 (matched for demographics and the main risk factors for CVT and PVT). FINDINGS A total of 537,913 patients with a COVID-19 diagnosis were included. The incidence of CVT in the two weeks after a COVID-19 diagnosis was 42.8 per million people (95% CI 28.5-64.2). This was significantly higher than in a matched cohort of people who received an mRNA vaccine (RR = 6.33, 95% CI 1.87-21.40, P = 0.00014) and patients with influenza (RR = 2.67, 95% CI 1.04-6.81, P = 0.031). The incidence of PVT after COVID-19 diagnosis was 392.3 per million people (95% CI 342.8-448.9). This was significantly higher than in a matched cohort of people who received an mRNA vaccine (RR=4.46, 95% CI 3.12-6.37, P < 0.0001) and patients with influenza (RR=1.43, 95% CI 1.10-1.88, P = 0.0094).
Collapse
Affiliation(s)
- Maxime Taquet
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Health NHS Foundation Trust, Oxford, United Kingdom
- Corresponding author.
| | - Masud Husain
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - John R Geddes
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| | | | - Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
- Oxford Health NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
541
|
García de Guadiana-Romualdo L, Martínez Martínez M, Rodríguez Mulero MD, Esteban-Torrella P, Hernández Olivo M, Alcaraz García MJ, Campos-Rodríguez V, Sancho-Rodríguez N, Galindo Martínez M, Alcaraz A, Ros Braquehais MS, Báguena Perez-Crespo C, Ramos Arenas V, Tomás Jiménez C, Consuegra-Sánchez L, Conesa-Hernandez A, Piñera-Salmerón P, Bernal-Morell E. Circulating MR-proADM levels, as an indicator of endothelial dysfunction, for early risk stratification of mid-term mortality in COVID-19 patients. Int J Infect Dis 2021; 111:211-218. [PMID: 34461254 PMCID: PMC8400460 DOI: 10.1016/j.ijid.2021.08.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 12/23/2022] Open
Abstract
Objectives Thromboinflammation, resulting from a complex interaction between thrombocytopathy, coagulopathy, and endotheliopathy, contributes to increased mortality in COVID-19 patients. MR-proADM, as a surrogate of adrenomedullin system disruption, leading to endothelial damage, has been reported as a promising biomarker for short-term prognosis. We evaluated the role of MR-proADM in the mid-term mortality in COVID-19 patients. Methods A prospective, observational study enrolling COVID-19 patients from August to October 2020. A blood sample for laboratory test analysis was drawn on arrival in the emergency department. The primary endpoint was 90-day mortality. The area under the curve (AUC) and Cox regression analyses were used to assess discriminatory ability and association with the endpoint. Results A total of 359 patients were enrolled, and the 90-day mortality rate was 8.9%. ROC AUC for MR-proADM predicting 90-day mortality was 0.832. An optimal cutoff of 0.80 nmol/L showed a sensitivity of 96.9% and a specificity of 58.4%, with a negative predictive value of 99.5%. Circulating MR-proADM levels (inverse transformed), after adjusting by a propensity score including eleven potential confounders, were an independent predictor of 90-day mortality (HR: 0.162 [95% CI: 0.043-0.480]) Conclusions Our data confirm that MR-proADM has a role in the mid-term prognosis of COVID-19 patients and might assist physicians with risk stratification.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Antonia Alcaraz
- Infectious Disease Unit, Hospital General Universitario Reina Sofía, Murcia, Spain
| | | | | | - Verónica Ramos Arenas
- Laboratory Medicine Department, Hospital Universitario Santa Lucía, Cartagena, Spain
| | | | | | | | | | - Enrique Bernal-Morell
- Infectious Disease Unit, Hospital General Universitario Reina Sofía, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB).
| |
Collapse
|
542
|
Chao WC, Wu CL, Huang JA, Chai JW, Teng CL, Lee WL, Fu YC, Chen SA. Association between Early Absolute Neutrophil Count and Level of D-Dimer among Patients with COVID-19 Infection in Central Taiwan. J Clin Med 2021; 10:jcm10173891. [PMID: 34501339 PMCID: PMC8432119 DOI: 10.3390/jcm10173891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/17/2021] [Accepted: 08/27/2021] [Indexed: 12/24/2022] Open
Abstract
Thromboembolism is a critical event in patients with coronavirus disease (COVID)-19 infection and highly associated with neutrophil extracellular traps. D-dimer has been found to be an essential thromboembolism-associated biomarker; however, the association between absolute neutrophil count (ANC) and level of D-dimer in patients with COVID-19 infection remains unclear. In this study, we enrolled consecutive patients with COVID-19 admitted to Taichung Veterans General Hospital (TCVGH), a referral center in central Taiwan with 20 airborne infection isolation rooms. Spearman correlation was used to determine the association between ANC and level of D-dimer in distinct time periods. A total of 28 consecutive patients with COVID-19 infection were enrolled, and 32.1% (9/28) of them required mechanical ventilation. Patients requiring mechanical ventilation had a higher ANC (8225 vs. 3427/µL, p < 0.01) and levels of D-dimer (6.0 vs. 0.6 mg/L, p < 0.01) compared with those without mechanical ventilation. Notably, we identified five patients with image-proven thromboembolic events during the hospital course, with the number of patients with pulmonary embolism, venous thrombosis and acute ischemic stroke were 2, 1, and 2, respectively. We found that ANC within 4 days correlated with the level of D-dimer to a moderate level (r = 0.71, p < 0.05), and the association between ANC and D-dimer no longer exist after day 5. In conclusion, we found highly prevalent thromboembolic events among patients with severe COVID-19 infection in central Taiwan and identified the association between early ANC and D-dimer. More studies are warranted to elucidate the underlying mechanism.
Collapse
Affiliation(s)
- Wen-Cheng Chao
- Department of Critical Care Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan; (W.-C.C.); (C.-L.W.)
- Department of Computer Science, Tunghai University, Taichung 407224, Taiwan
- Department of Automatic Control Engineering, Feng Chia University, Taichung 407802, Taiwan
- Big Data Center, National Chung Hsing University, Taichung 40227, Taiwan
| | - Chieh-Liang Wu
- Department of Critical Care Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan; (W.-C.C.); (C.-L.W.)
- Department of Computer Science, Tunghai University, Taichung 407224, Taiwan
- Department of Automatic Control Engineering, Feng Chia University, Taichung 407802, Taiwan
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung 407224, Taiwan
| | - Jin-An Huang
- Department of Neurological Institute, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
| | - Jyh-Wen Chai
- Department of Radiology, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
| | - Chieh-Lin Teng
- Division of Hematology and Medical Oncology, Department of Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
- Department of Life Science, Tunghai University, Taichung 407224, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Wen-Lieng Lee
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
- Department of Medicine, National Yang-Ming Chiou Tung University, Taipei 112, Taiwan
- Correspondence: (W.-L.L.); (Y.-C.F.)
| | - Yun-Ching Fu
- Section of Pediatric Cardiology, Department of Pediatrics, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiou Tung University, Taipei 112, Taiwan
- Correspondence: (W.-L.L.); (Y.-C.F.)
| | - Shih-Ann Chen
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
- Institute of Clinical Medicine, National Yang Ming Chiou Tung University, Taipei 112, Taiwan
| |
Collapse
|
543
|
Strich JR, Tian X, Samour M, King CS, Shlobin O, Reger R, Cohen J, Ahmad K, Brown AW, Khangoora V, Aryal S, Migdady Y, Kyte JJ, Joo J, Hays R, Collins AC, Battle E, Valdez J, Rivero J, Kim IH, Erb-Alvarez J, Shalhoub R, Chakraborty M, Wong S, Colton B, Ramos-Benitez MJ, Warner S, Chertow DS, Olivier KN, Aue G, Davey RT, Suffredini AF, Childs RW, Nathan SD. Fostamatinib for the Treatment of Hospitalized Adults With Coronavirus Disease 2019: A Randomized Trial. Clin Infect Dis 2021; 75:e491-e498. [PMID: 34467402 PMCID: PMC9890443 DOI: 10.1093/cid/ciab732] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) requiring hospitalization is characterized by robust antibody production, dysregulated immune response, and immunothrombosis. Fostamatinib is a novel spleen tyrosine kinase inhibitor that we hypothesize will ameliorate Fc activation and attenuate harmful effects of the anti-COVID-19 immune response. METHODS We conducted a double-blind, randomized, placebo-controlled trial in hospitalized adults requiring oxygen with COVID-19 where patients receiving standard of care were randomized to receive fostamatinib or placebo. The primary outcome was serious adverse events by day 29. RESULTS A total of 59 patients underwent randomization (30 to fostamatinib and 29 to placebo). Serious adverse events occurred in 10.5% of patients in the fostamatinib group compared with 22% in placebo (P = .2). Three deaths occurred by day 29, all receiving placebo. The mean change in ordinal score at day 15 was greater in the fostamatinib group (-3.6 ± 0.3 vs -2.6 ± 0.4, P = .035) and the median length in the intensive care unit was 3 days in the fostamatinib group vs 7 days in placebo (P = .07). Differences in clinical improvement were most evident in patients with severe or critical disease (median days on oxygen, 10 vs 28, P = .027). There were trends toward more rapid reductions in C-reactive protein, D-dimer, fibrinogen, and ferritin levels in the fostamatinib group. CONCLUSION For COVID-19 requiring hospitalization, the addition of fostamatinib to standard of care was safe and patients were observed to have improved clinical outcomes compared with placebo. These results warrant further validation in larger confirmatory trials. CLINICAL TRIALS REGISTRATION Clinicaltrials.gov, NCT04579393.
Collapse
Affiliation(s)
- Jeffrey R Strich
- Correspondence: J. R. Strich, Critical Care Medicine Department, National Institutes of Health Clinical Center, 10 Center Drive, 2C145, Bethesda, MD 20892 ()
| | - Xin Tian
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mohamed Samour
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christopher S King
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Oksana Shlobin
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Robert Reger
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jonathan Cohen
- Adventist Healthcare Shady Grove Medical Center, Rockville, Maryland, USA
| | - Kareem Ahmad
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - A Whitney Brown
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Vikramjit Khangoora
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Shambhu Aryal
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Yazan Migdady
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jennifer Jo Kyte
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jungnam Joo
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Rebecca Hays
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - A Claire Collins
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Edwinia Battle
- Advanced Lung Disease and Lung Transplant Program, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Janet Valdez
- United States Public Health Service Commissioned Corps, Rockville, Maryland, USA,National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Josef Rivero
- United States Public Health Service Commissioned Corps, Rockville, Maryland, USA,National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ick Ho Kim
- United States Public Health Service Commissioned Corps, Rockville, Maryland, USA,National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Julie Erb-Alvarez
- United States Public Health Service Commissioned Corps, Rockville, Maryland, USA,National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ruba Shalhoub
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mala Chakraborty
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Susan Wong
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Benjamin Colton
- Pharmacy Department, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Marcos J Ramos-Benitez
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, Maryland, USA,Postdoctoral Research Associate Training Program, National Institute of General Medical Sciences, Bethesda, Maryland, USA
| | - Seth Warner
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Daniel S Chertow
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, Maryland, USA,United States Public Health Service Commissioned Corps, Rockville, Maryland, USA,National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kenneth N Olivier
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Georg Aue
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Richard T Davey
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Anthony F Suffredini
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | | | | |
Collapse
|
544
|
The roles of platelets in COVID-19-associated coagulopathy and vaccine-induced immune thrombotic thrombocytopenia. Trends Cardiovasc Med 2021; 32:1-9. [PMID: 34455073 PMCID: PMC8390120 DOI: 10.1016/j.tcm.2021.08.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/06/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023]
Abstract
In coronavirus disease 2019 (COVID-19), multiple thromboinflammatory events contribute to the pathophysiology, including coagulation system activation, suppressed fibrinolysis, vascular endothelial cell injury, and prothrombotic alterations in immune cells such as macrophages and neutrophils. Although thrombocytopenia is not an initial presentation as an infectious coagulopathy, recent studies have demonstrated the vital role of platelets in COVID-19-associated coagulopathy SARS-CoV-2 and its spike protein have been known to directly or indirectly promote release of prothrombotic and inflammatory mediators that lead to COVID-19-associated coagulopathy. Although clinical features of vaccine-induced immune thrombotic thrombocytopenia include uncommon locations of thrombosis, including cerebral venous sinus, we speculate coronavirus spike-protein-initiated prothrombotic pathways are involved in the pathogenesis of vaccine-induced immune thrombotic thrombocytopenia, as current evidence suggests that the spike protein is the promotor and other cofactors such as perturbed immune response and inflammatory reaction enhance the production of anti-platelet factor 4 antibody.
Collapse
|
545
|
Kaklamanos A, Belogiannis K, Skendros P, Gorgoulis VG, Vlachoyiannopoulos PG, Tzioufas AG. COVID-19 Immunobiology: Lessons Learned, New Questions Arise. Front Immunol 2021; 12:719023. [PMID: 34512643 PMCID: PMC8427766 DOI: 10.3389/fimmu.2021.719023] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/09/2021] [Indexed: 12/17/2022] Open
Abstract
There is strong evidence that COVID-19 pathophysiology is mainly driven by a spatiotemporal immune deregulation. Both its phenotypic heterogeneity, spanning from asymptomatic to severe disease/death, and its associated mortality, are dictated by and linked to maladaptive innate and adaptive immune responses against SARS-CoV-2, the etiologic factor of the disease. Deregulated interferon and cytokine responses, with the contribution of immune and cellular stress-response mediators (like cellular senescence or uncontrolled inflammatory cell death), result in innate and adaptive immune system malfunction, endothelial activation and inflammation (endothelitis), as well as immunothrombosis (with enhanced platelet activation, NET production/release and complement hyper-activation). All these factors play key roles in the development of severe COVID-19. Interestingly, another consequence of this immune deregulation, is the production of autoantibodies and the subsequent development of autoimmune phenomena observed in some COVID-19 patients with severe disease. These new aspects of the disease that are now emerging (like autoimmunity and cellular senescence), could offer us new opportunities in the field of disease prevention and treatment. Simultaneously, lessons already learned from the immunobiology of COVID-19 could offer new insights, not only for this disease, but also for a variety of chronic inflammatory responses observed in autoimmune and (auto)inflammatory diseases.
Collapse
Affiliation(s)
- Aimilios Kaklamanos
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Institute for Autoimmune Systemic and Neurological Diseases, Athens, Greece
| | - Konstantinos Belogiannis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Skendros
- First Department of Internal Medicine and Laboratory of Molecular Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| | - Vassilis G. Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
- Basic Research Center, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
- Center for New Biotechnologies and Precision Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Panayiotis G. Vlachoyiannopoulos
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Institute for Autoimmune Systemic and Neurological Diseases, Athens, Greece
| | - Athanasios G. Tzioufas
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Institute for Autoimmune Systemic and Neurological Diseases, Athens, Greece
| |
Collapse
|
546
|
Rainer Q, Molina M, Ibrahim E, Saltzman R, Masterson T, Ramasamy R. Peyronie's disease in a patient after COVID-19 infection: A case report. Andrologia 2021; 53:e14219. [PMID: 34397121 PMCID: PMC8420294 DOI: 10.1111/and.14219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/05/2021] [Indexed: 12/27/2022] Open
Abstract
Coronavirus disease 2019 (COVID‐19) is an emerging infectious disease caused by a novel coronavirus (SARS‐CoV‐2), which demonstrates the ability to invade endothelial cells and cause systemic inflammation. Many possible long‐term sequelae of COVID‐19 remain unidentified. We describe a case of a man who developed Peyronie's disease after a resolved COVID‐19 infection. Erectile dysfunction was confirmed by the International Index of Erectile Function‐15(IIEF) and Sexual Health Inventory for Men(SHIM) scores. A diagnosis was Peyronie's disease was confirmed on ultrasound. Furthermore, he was found to have low endothelial progenitor cells colony‐forming units and low brachial artery flow‐mediated vasodilation, both of that are indicative of endothelial dysfunction. This case suggests Peyronie's disease should be considered as a possible sequela of COVID‐19 infection and providers should inquire about a history of COVID‐19 infection in patients presenting with Peyronie's disease.
Collapse
Affiliation(s)
- Quinn Rainer
- University of Miami Miller School of Medicine, Miami, FL, USA
| | - Manuel Molina
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Emad Ibrahim
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Russell Saltzman
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Thomas Masterson
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ranjith Ramasamy
- Department of Urology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
547
|
Chen R, Zhang S, Su S, Ye H, Shu H. Interactions Between Specific Immune Status of Pregnant Women and SARS-CoV-2 Infection. Front Cell Infect Microbiol 2021; 11:721309. [PMID: 34458162 PMCID: PMC8387674 DOI: 10.3389/fcimb.2021.721309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 07/29/2021] [Indexed: 12/18/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogen responsible for the Coronavirus Disease 2019 (COVID-19) global pandemic. Because it is a new and highly contagious coronavirus, most people, especially pregnant women, lack immunity. It is therefore important to understand the interaction between why pregnant women are susceptible to SARS-CoV-2 and the specific immune systems of pregnant women. Here, we provide an overview of the changes that occur in the immune system during pregnancy, the activation and response of the immune system in pregnant women with COVID-19, adverse pregnancy outcomes in pregnant women with COVID-19, and the treatment and prevention of COVID-19 in this population.
Collapse
Affiliation(s)
- Ruirong Chen
- Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Shaofen Zhang
- Department of Gynaecology and Obstetrics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Sheng Su
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Haiyan Ye
- Department of Gynaecology and Obstetrics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Haihua Shu
- Department of Anesthesiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
548
|
Mumoli N, Bonaventura A, Colombo A, Vecchié A, Cei M, Vitale J, Pavan L, Mazzone A, Dentali F. Lung function and symptoms in post-COVID-19 patients: a single-center experience. Mayo Clin Proc Innov Qual Outcomes 2021; 5:907-915. [PMID: 34396048 PMCID: PMC8352649 DOI: 10.1016/j.mayocpiqo.2021.08.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Objective To address the lack of information about clinical sequelae of coronavirus disease 2019 (COVID-19). Patients and Methods Previously hospitalized COVID-19 patients who were attending the outpatient clinic for post-COVID-19 patients (ASST Ovest Milanese, Magenta, Italy) were included in this retrospective study. They underwent blood draw for complete blood count, C-reactive protein (CRP), ferritin, D-dimer, and arterial blood gas analysis (ABG) and chest high-resolution computed tomography (HRCT) scan. The primary endpoint was the assessment of blood gas exchanges after 3 months. Other endpoints included the assessment of symptoms and chest HRCT scan abnormalities and changes in inflammatory biomarkers after 3 months from hospital admission. Results Eighty-eight patients (men 73.9%) were included. Admission arterial ABG analysis showed hypoxia and hypocapnia and a PaO2/FiO2 of 271.4 (238-304.7) mmHg, that greatly improved after 3 months (426.19 [395.24-461.90] mmHg, P<.001). A 40% of patients was still hypocapnic after 3 months. Inflammatory biomarkers dramatically improved after 3 months from hospitalization. Fever, resting dyspnea, and cough were common at hospital admission and improved after 3 months, when dyspnea on exertion and arthralgias arose. On chest HRCT scan, more than half of individuals still presented interstitial involvement. Positive correlations between the interstitial pattern at 3 months and dyspnea on admission were found. CRP at admission was positively associated with the presence of interstitial involvement at follow-up. The persistence of cough was associated with presence of bronchiectasis and consolidation on follow-up chest HRCT scan. Conclusion While inflammatory biomarker levels normalized after 3 months, signs of lung damage persist for a longer period. These findings support the need for implementing post-COVID-19 outpatient clinics to closely follow-up COVID-19 patients after hospitalization.
Collapse
Key Words
- ABG, arterial blood gas
- ARDS, acute respiratory distress syndrome
- COVID-19
- COVID-19, coronavirus disease 2019
- CPAP, continuous positive airway pressure
- CRP, C-reactive protein
- CT, computed tomography
- CVD, cardiovascular disease
- DOE, dyspnea on exertion
- GGO, ground-glass opacity
- HRCT, high-resolution computed tomography
- IQR, interquartile range
- PFT, pulmonary function test
- PaCO2, arterial partial pressure of carbon dioxide
- PaO2, arterial partial pressure of oxygen
- PaO2/FiO2, ratio of arterial partial pressure of oxygen to fractional inspired oxygen
- SARS-CoV-2
- SARS-CoV-2, severe acute respiratory syndrome coronavirus-2
- SD, standard deviation
- STROBE, Strengthening the Reporting of Observational Studies in Epidemiology
- SpO2, peripheral capillary oxygen saturation
- V/Q, ventilation/perfusion ratio
- chest CT scan
- hypocapnia
- inflammation
Collapse
Affiliation(s)
- Nicola Mumoli
- Department of Internal Medicine, ASST Ovest Milanese, Magenta MI, Italy
| | - Aldo Bonaventura
- Department of Internal Medicine, ASST Sette Laghi, Varese, Italy
| | | | | | - Marco Cei
- Department of Internal Medicine, Cecina Hospital, Cecina Livorno), Italy
| | - José Vitale
- Department of Internal Medicine, ASST Ovest Milanese, Magenta MI, Italy
| | - Luca Pavan
- Department of Internal Medicine, ASST Ovest Milanese, Magenta MI, Italy
| | - Antonino Mazzone
- Department of Internal Medicine, ASST Ovest Milanese, Magenta MI, Italy
| | - Francesco Dentali
- Department of Medicine and Surgery, Insubria University, Varese, Italy
| |
Collapse
|
549
|
Shaw RJ, Bradbury C, Abrams ST, Wang G, Toh CH. COVID-19 and immunothrombosis: emerging understanding and clinical management. Br J Haematol 2021; 194:518-529. [PMID: 34114204 DOI: 10.1111/bjh.17664] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic has been the most significant health crisis in recent global history. Early studies from Wuhan highlighted COVID-19-associated coagulopathy and a significant association with mortality was soon recognised. As research continues across the world, more evidence is emerging of the cross-talk between the innate immune system, coagulation activation and inflammation. Immunothrombosis has been demonstrated to play a key role in the pathophysiology of severe COVID-19, with extracellular histones and neutrophil extracellular traps detected in the plasma and cardiopulmonary tissues of critically ill patients. Targeting the components of immunothrombosis is becoming an important factor in the treatment of patients with COVID-19 infection. Recent studies report outcomes of intermediate and therapeutic anticoagulation in hospitalised patients with varying severities of COVID-19 disease, including optimal dosing and associated bleeding risks. Immunomodulatory therapies, including corticosteroids and IL-6 receptor antagonists, have been demonstrated to significantly reduce mortality in COVID-19 patients. As the pandemic continues, more studies are required to understand the driving factors and upstream mechanisms for coagulopathy and immunothrombosis in COVID-19, and thus potentially develop more targeted therapies for SARS-CoV-2 infection, both in the acute phase and in those who develop longer-term symptom burden.
Collapse
Affiliation(s)
- Rebecca J Shaw
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
- The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | | | - Simon T Abrams
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
| | - Guozheng Wang
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
| | - Cheng-Hock Toh
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
- The Roald Dahl Haemostasis and Thrombosis Centre, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
550
|
Luo MH, Qian YQ, Huang DL, Luo JC, Su Y, Wang H, Yu SJ, Liu K, Tu GW, Luo Z. Tailoring glucocorticoids in patients with severe COVID-19: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1261. [PMID: 34532398 PMCID: PMC8421952 DOI: 10.21037/atm-21-1783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 06/10/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To discuss the pathogenesis of severe coronavirus disease 2019 (COVID-19) infection and the pharmacological effects of glucocorticoids (GCs) toward this infection. To review randomized controlled trials (RCTs) using GCs to treat patients with severe COVID-19, and investigate whether GC timing, dosage, or duration affect clinical outcomes. Finally. to discuss the use of biological markers, respiratory parameters, and radiological evidence to select patients for improved GC therapeutic precision. BACKGROUND COVID-19 has become an unprecedented global challenge. As GCs have been used as key immunomodulators to treat inflammation-related diseases, they may play key roles in limiting disease progression by modulating immune responses, cytokine production, and endothelial function in patients with severe COVID-19, who often experience excessive cytokine production and endothelial and renin-angiotensin system (RAS) dysfunction. Current clinical trials have partially proven this efficacy, but GC timing, dosage, and duration vary greatly, with no unifying consensus, thereby creating confusion. METHODS Publications through March 2021 were retrieved from the Web of Science and PubMed. Results from cited references in published articles were also included. CONCLUSIONS GCs play key roles in treating severe COVID-19 infections. Pharmacologically, GCs could modulate immune cells, reduce cytokine and chemokine, and improve endothelial functions in patients with severe COVID-19. Benefits of GCs have been observed in multiple clinical trials, but the timing, dosage and duration vary across studies. Tapering as an option is not widely accepted. However, early initiation of treatment, a tailored dosage with appropriate tapering may be of particular importance, but evidence is inconclusive and more investigations are needed. Biological markers, respiratory parameters, and radiological evidence could also help select patients for specific tailored treatments.
Collapse
Affiliation(s)
- Ming-Hao Luo
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Qi Qian
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dan-Lei Huang
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing-Chao Luo
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Su
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huan Wang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shen-Ji Yu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kai Liu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guo-Wei Tu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhe Luo
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Critical Care Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| |
Collapse
|