1
|
Lee Y, Lee MK, Lee HR, Kim B, Kim M, Jung S. 3D-printed airway model as a platform for SARS-CoV-2 infection and antiviral drug testing. Biomaterials 2024; 311:122689. [PMID: 38944967 DOI: 10.1016/j.biomaterials.2024.122689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 05/20/2024] [Accepted: 06/24/2024] [Indexed: 07/02/2024]
Abstract
We present a bioprinted three-layered airway model with a physiologically relevant microstructure for the study of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection dynamics. This model exhibited clear cell-cell junctions and mucus secretion with an efficient expression of angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2). Having infected air-exposed epithelial cells in the upper layer with a minimum multiplicity of infection of 0.01, the airway model showed a marked susceptibility to SARS-CoV-2 within one-day post-infection (dpi). Furthermore, the unique longevity allowed the observation of cytopathic effects and barrier degradation for 21 dpi. The in-depth transcriptomic analysis revealed dramatic changes in gene expression affecting the infection pathway, viral proliferation, and host immune response which are consistent with COVID-19 patient data. Finally, the treatment of antiviral agents, such as remdesivir and molnupiravir, through the culture medium underlying the endothelium resulted in a marked inhibition of viral replication within the epithelium. The bioprinted airway model can be used as a manufacturable physiological platform to study disease pathogeneses and drug efficacy.
Collapse
Affiliation(s)
- Yunji Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Myoung Kyu Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Hwa-Rim Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| | - Byungil Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Meehyein Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea; Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Sungjune Jung
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
2
|
Lai HY, Fan KC, Lee YH, Lew WZ, Lai WY, Lee SY, Chang WJ, Huang HM. Using a static magnetic field to attenuate the severity in COVID-19-invaded lungs. Sci Rep 2024; 14:16830. [PMID: 39039227 PMCID: PMC11263632 DOI: 10.1038/s41598-024-67806-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024] Open
Abstract
Two important factors affecting the progress of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are the S-protein binding function of ACE2 receptors and the membrane fluidity of host cells. This study aimed to evaluate the effect of static magnetic field (SMF) on S-protein/ACE2 binding and cellular membrane fluidity of lung cells, and was performed in vitro using a Calu-3 cell model and in vivo using an animal model. The ability of ACE2 receptors to bind to SARS-CoV-2 spike protein on host cell surfaces under SMF stimulation was evaluated using fluorescence images. Host lung cell membrane fluidity was tested using fluorescence polarization to determine the effects of SMF. Our results indicate that 0.4 T SMF can affect binding between S-protein and ACE2 receptors and increase Calu-3 cell membrane fluidity, and that SMF exposure attenuates LPS-induced alveolar wall thickening in mice. These results may be of value for developing future non-contact, non-invasive, and low side-effect treatments to reduce disease severity in COVID-19-invaded lungs.
Collapse
Affiliation(s)
- Hsuan-Yu Lai
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kuo-Cheng Fan
- Department of Dentistry, Taipei Medical University Wan Fang Hospital, 11696, Taipei, Taiwan
| | - Yen-Hua Lee
- Department of Animal Science, National Pingtung University of Science and Technology, 912301, Pingtung, Taiwan
| | - Wei-Zhen Lew
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Wei-Yi Lai
- Department of Medical Research, Taipei Veterans General Hospital, 112062, Taipei, Taiwan
| | - Sheng-Yang Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Dentistry, Taipei Medical University Wan Fang Hospital, 11696, Taipei, Taiwan
| | - Wei-Jen Chang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Haw-Ming Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
3
|
Krivdić Dupan Z, Periša V, Suver Stević M, Mihalj M, Tolušić Levak M, Guljaš S, Salha T, Loinjak D, Kos M, Šapina M, Canjko I, Šambić Penc M, Štefančić M, Nešković N. The Impact of Pentraxin 3 Serum Levels and Angiotensin-Converting Enzyme Polymorphism on Pulmonary Infiltrates and Mortality in COVID-19 Patients. Biomedicines 2024; 12:1618. [PMID: 39062191 PMCID: PMC11275229 DOI: 10.3390/biomedicines12071618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
OBJECTIVES The aim of this study was to examine the impact of the pentraxin 3 (PTX3) serum level and angiotensin-converting enzyme (ACE) gene insertion/deletion (I/D) polymorphism on the severity of radiographic pulmonary infiltrates and the clinical outcomes of COVID-19. METHODS The severity of COVID-19 pulmonary infiltrates was evaluated within a week of admission by analyzing chest X-rays (CXR) using the modified Brixia (MBrixa) scoring system. The insertion (I)/deletion (D) polymorphism of the ACE gene and the serum levels of PTX3 were determined for all patients included in the study. RESULTS This study included 80 patients. Using a cut-off serum level of PTX3 ≥ 2.765 ng/mL, the ROC analysis (AUC 0.871, 95% CI 0.787-0.954, p < 0.001) showed a sensitivity of 85.7% and specificity of 78.8% in predicting severe MBrixa scores. Compared to ACE I/I polymorphism, D/D polymorphism significantly increased the risk of severe CXR infiltrates, OR 7.7 (95% CI: 1.9-30.1), and p = 0.002. Significant independent predictors of severe CXR infiltrates include hypertension (OR 7.71), PTX3 (OR 1.20), and ACE D/D polymorphism (OR 18.72). Hypertension (OR 6.91), PTX3 (OR 1.47), and ACE I/I polymorphism (OR 0.09) are significant predictors of poor outcomes. CONCLUSION PTX3 and ACE D/D polymorphism are significant predictors of the severity of COVID-19 pneumonia. PTX3 is a significant predictor of death.
Collapse
Affiliation(s)
- Zdravka Krivdić Dupan
- Department of Radiology, Osijek University Hospital, 31000 Osijek, Croatia
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Vlatka Periša
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Hematology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Mirjana Suver Stević
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Transfusion Medicine, Osijek University Hospital, 31000 Osijek, Croatia
| | - Martina Mihalj
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Dermatology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Maja Tolušić Levak
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Dermatology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Silva Guljaš
- Department of Radiology, Osijek University Hospital, 31000 Osijek, Croatia
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Tamer Salha
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Faculty of Dental Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Domagoj Loinjak
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Internal Medicine, Osijek University Hospital, 31000 Osijek, Croatia
| | - Martina Kos
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Pediatrics, Osijek University Hospital, 31000 Osijek, Croatia
| | - Matej Šapina
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Pediatrics, Osijek University Hospital, 31000 Osijek, Croatia
| | - Ivana Canjko
- Department of Radiotherapy and Oncology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Mirela Šambić Penc
- Department of Radiotherapy and Oncology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Marin Štefančić
- Department of Radiology, National Memorial Hospital Vukovar, 32000 Vukovar, Croatia
| | - Nenad Nešković
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- International Medical Center Priora, 31431 Cepin, Croatia
| |
Collapse
|
4
|
Starshinova A, Borozinets A, Kulpina A, Sereda V, Rubinstein A, Kudryavtsev I, Kudlay D. Bronchial Asthma and COVID-19: Etiology, Pathological Triggers, and Therapeutic Considerations. PATHOPHYSIOLOGY 2024; 31:269-287. [PMID: 38921725 PMCID: PMC11206645 DOI: 10.3390/pathophysiology31020020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Bronchial asthma (BA) continues to be a difficult disease to diagnose. Various factors have been described in the development of BA, but to date, there is no clear evidence for the etiology of this chronic disease. The emergence of COVID-19 has contributed to the pandemic course of asthma and immunologic features. However, there are no unambiguous data on asthma on the background and after COVID-19. There is correlation between various trigger factors that provoke the development of bronchial asthma. It is now obvious that the SARS-CoV-2 virus is one of the provoking factors. COVID-19 has affected the course of asthma. Currently, there is no clear understanding of whether asthma progresses during or after COVID-19 infection. According to the results of some studies, a significant difference was identified between the development of asthma in people after COVID-19. Mild asthma and moderate asthma do not increase the severity of COVID-19 infection. Nevertheless, oral steroid treatment and hospitalization for severe BA were associated with higher COVID-19 severity. The influence of SARS-CoV-2 infection is one of the protective factors. It causes the development of severe bronchial asthma. The accumulated experience with omalizumab in patients with severe asthma during COVID-19, who received omalizumab during the pandemic, has strongly suggested that continued treatment with omalizumab is safe and may help prevent the severe course of COVID-19. Targeted therapy for asthma with the use of omalizumab may also help to reduce severe asthma associated with COVID-19. However, further studies are needed to prove the effect of omalizumab. Data analysis should persist, based on the results of the course of asthma after COVID-19 with varying degrees of severity.
Collapse
Affiliation(s)
- Anna Starshinova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
| | - Anastasia Borozinets
- Medical Department, I.M. Sechenov First Moscow State Medical University, 197022 Moscow, Russia
| | - Anastasia Kulpina
- Medical Department, Saint Petersburg State Pediatric Medical University, 194100 St. Petersburg, Russia;
| | - Vitaliy Sereda
- Medical Department, Saint Petersburg State University, 199034 St. Petersburg, Russia;
| | - Artem Rubinstein
- Department of immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia;
| | - Igor Kudryavtsev
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia;
- Department of immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia;
| | - Dmitry Kudlay
- Institute of Immunology FMBA of Russia, 115478 Moscow, Russia;
- Department of Pharmacognosy and Industrial Pharmacy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
5
|
Kim M, Hwang J, Grist JT, Abueid G, Yoon SH, Grau V, Fraser E, Gleeson FV. Functional Impairment in Small Airways Associated With the Breathlessness Symptoms in Long-Coronavirus Disease. J Thorac Imaging 2024; 39:79-85. [PMID: 37889567 DOI: 10.1097/rti.0000000000000748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
PURPOSE This study aimed to determine the association between functional impairment in small airways and symptoms of dyspnea in patients with Long-coronavirus disease (COVID), using imaging and computational modeling analysis. PATIENTS AND METHODS Thirty-four patients with Long-COVID underwent thoracic computed tomography and hyperpolarized Xenon-129 magnetic resonance imaging (HP Xe MRI) scans. Twenty-two answered dyspnea-12 questionnaires. We used a computed tomography-based full-scale airway network (FAN) flow model to simulate pulmonary ventilation. The ventilation distribution projected on a coronal plane and the percentage lobar ventilation modeled in the FAN model were compared with the HP Xe MRI data. To assess the ventilation heterogeneity in small airways, we calculated the fractal dimensions of the impaired ventilation regions in the HP Xe MRI and FAN models. RESULTS The ventilation distribution projected on a coronal plane showed an excellent resemblance between HP Xe MRI scans and FAN models (structure similarity index: 0.87 ± 0.04). In both the image and the model, the existence of large clustered ventilation defects was not identifiable regardless of dyspnea severity. The percentage lobar ventilation of the HP Xe MRI and FAN model showed a strong correlation (ρ = 0.63, P < 0.001). The difference in the fractal dimension of impaired ventilation zones between the low and high dyspnea-12 score groups was significant (HP Xe MRI: 1.97 [1.89 to 2.04] and 2.08 [2.06 to 2.14], P = 0.005; FAN: 2.60 [2.59 to 2.64] and 2.64 [2.63 to 2.65], P = 0.056). CONCLUSIONS This study has identified a potential association of small airway functional impairment with breathlessness in Long-COVID, using fractal analysis of HP Xe MRI scans and FAN models.
Collapse
Affiliation(s)
- Minsuok Kim
- School of Mechanical, Electrical, and Manufacturing Engineering, Loughborough University, Loughborough
| | - Jeongeun Hwang
- Department of Engineering Science, Institute of Biomedical Engineering, Oxford e-Research Centre
- Department of Medical IT Engineering, Soonchunhyang University, Chungcheonnam-do
| | - James T Grist
- Department of Physiology, Anatomy, and Genetics
- Department of Radiology
- Oxford Centre for Clinical MR Research, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, UK
| | | | - Soon Ho Yoon
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Vicente Grau
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine
| | - Emily Fraser
- Oxford Interstitial Lung Disease Service, The Churchill Hospital
| | - Fergus V Gleeson
- Department of Oncology, University of Oxford
- Department of Radiology
| |
Collapse
|
6
|
Narasaraju T, Neeli I, Criswell SL, Krishnappa A, Meng W, Silva V, Bila G, Vovk V, Serhiy Z, Bowlin GL, Meyer N, Luning Prak ET, Radic M, Bilyy R. Neutrophil Activity and Extracellular Matrix Degradation: Drivers of Lung Tissue Destruction in Fatal COVID-19 Cases and Implications for Long COVID. Biomolecules 2024; 14:236. [PMID: 38397474 PMCID: PMC10886497 DOI: 10.3390/biom14020236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Pulmonary fibrosis, severe alveolitis, and the inability to restore alveolar epithelial architecture are primary causes of respiratory failure in fatal COVID-19 cases. However, the factors contributing to abnormal fibrosis in critically ill COVID-19 patients remain unclear. This study analyzed the histopathology of lung specimens from eight COVID-19 and six non-COVID-19 postmortems. We assessed the distribution and changes in extracellular matrix (ECM) proteins, including elastin and collagen, in lung alveoli through morphometric analyses. Our findings reveal the significant degradation of elastin fibers along the thin alveolar walls of the lung parenchyma, a process that precedes the onset of interstitial collagen deposition and widespread intra-alveolar fibrosis. Lungs with collapsed alveoli and organized fibrotic regions showed extensive fragmentation of elastin fibers, accompanied by alveolar epithelial cell death. Immunoblotting of lung autopsy tissue extracts confirmed elastin degradation. Importantly, we found that the loss of elastin was strongly correlated with the induction of neutrophil elastase (NE), a potent protease that degrades ECM. This study affirms the critical role of neutrophils and neutrophil enzymes in the pathogenesis of COVID-19. Consistently, we observed increased staining for peptidyl arginine deiminase, a marker for neutrophil extracellular trap release, and myeloperoxidase, an enzyme-generating reactive oxygen radical, indicating active neutrophil involvement in lung pathology. These findings place neutrophils and elastin degradation at the center of impaired alveolar function and argue that elastolysis and alveolitis trigger abnormal ECM repair and fibrosis in fatal COVID-19 cases. Importantly, this study has implications for severe COVID-19 complications, including long COVID and other chronic inflammatory and fibrotic disorders.
Collapse
Affiliation(s)
- Teluguakula Narasaraju
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA; or (T.N.); (I.N.); (V.S.)
- Department of Microbiology, Adichunchanagiri Institute of Medical Sciences, Center for Research and Innovation, Adichunchanagiri University, Mandya 571448, India
| | - Indira Neeli
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA; or (T.N.); (I.N.); (V.S.)
| | - Sheila L. Criswell
- Department of Diagnostic and Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA;
| | - Amita Krishnappa
- Department of Pathology, Adichunchanagiri Institute of Medical Sciences, Adichunchanagiri University, Mandya 571448, India;
| | - Wenzhao Meng
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (W.M.); (E.T.L.P.)
| | - Vasuki Silva
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA; or (T.N.); (I.N.); (V.S.)
| | - Galyna Bila
- Department of Histology, Cytology, Histology & Embryology, Danylo Halytsky Lviv National Medical University, 79010 Lviv, Ukraine; (G.B.); (R.B.)
| | - Volodymyr Vovk
- Department of Pathological Anatomy and Forensic Medicine, Danylo Halytsky Lviv National Medical University, 79010 Lviv, Ukraine;
- Lviv Regional Pathological Anatomy Office, CU ENT (Pulmonology Lviv Regional Diagnostic Center), 79000 Lviv, Ukraine;
| | - Zolotukhin Serhiy
- Lviv Regional Pathological Anatomy Office, CU ENT (Pulmonology Lviv Regional Diagnostic Center), 79000 Lviv, Ukraine;
| | - Gary L. Bowlin
- Department of Biomedical Engineering, University of Memphis, Memphis, TN 38152, USA;
| | - Nuala Meyer
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Pulmonary, Allergy, and Critical Care Medicine and Center for Translational Lung Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eline T. Luning Prak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (W.M.); (E.T.L.P.)
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Marko Radic
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA; or (T.N.); (I.N.); (V.S.)
| | - Rostyslav Bilyy
- Department of Histology, Cytology, Histology & Embryology, Danylo Halytsky Lviv National Medical University, 79010 Lviv, Ukraine; (G.B.); (R.B.)
| |
Collapse
|
7
|
Milross L, Hunter B, McDonald D, Merces G, Thomson A, Hilkens CMU, Wills J, Rees P, Jiwa K, Cooper N, Majo J, Ashwin H, Duncan CJA, Kaye PM, Bayraktar OA, Filby A, Fisher AJ. Distinct lung cell signatures define the temporal evolution of diffuse alveolar damage in fatal COVID-19. EBioMedicine 2024; 99:104945. [PMID: 38142637 PMCID: PMC10788437 DOI: 10.1016/j.ebiom.2023.104945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/26/2023] Open
Abstract
BACKGROUND Lung damage in severe COVID-19 is highly heterogeneous however studies with dedicated spatial distinction of discrete temporal phases of diffuse alveolar damage (DAD) and alternate lung injury patterns are lacking. Existing studies have also not accounted for progressive airspace obliteration in cellularity estimates. We used an imaging mass cytometry (IMC) analysis with an airspace correction step to more accurately identify the cellular immune response that underpins the heterogeneity of severe COVID-19 lung disease. METHODS Lung tissue was obtained at post-mortem from severe COVID-19 deaths. Pathologist-selected regions of interest (ROIs) were chosen by light microscopy representing the patho-evolutionary spectrum of DAD and alternate disease phenotypes were selected for comparison. Architecturally normal SARS-CoV-2-positive lung tissue and tissue from SARS-CoV-2-negative donors served as controls. ROIs were stained for 40 cellular protein markers and ablated using IMC before segmented cells were classified. Cell populations corrected by ROI airspace and their spatial relationships were compared across lung injury patterns. FINDINGS Forty patients (32M:8F, age: 22-98), 345 ROIs and >900k single cells were analysed. DAD progression was marked by airspace obliteration and significant increases in mononuclear phagocytes (MnPs), T and B lymphocytes and significant decreases in alveolar epithelial and endothelial cells. Neutrophil populations proved stable overall although several interferon-responding subsets demonstrated expansion. Spatial analysis revealed immune cell interactions occur prior to microscopically appreciable tissue injury. INTERPRETATION The immunopathogenesis of severe DAD in COVID-19 lung disease is characterised by sustained increases in MnPs and lymphocytes with key interactions occurring even prior to lung injury is established. FUNDING UK Research and Innovation/Medical Research Council through the UK Coronavirus Immunology Consortium, Barbour Foundation, General Sir John Monash Foundation, Newcastle University, JGW Patterson Foundation, Wellcome Trust.
Collapse
Affiliation(s)
- Luke Milross
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Bethany Hunter
- Newcastle University Biosciences Institute, Newcastle upon Tyne, UK; Innovation Methodology and Application Research Theme, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - David McDonald
- Newcastle University Biosciences Institute, Newcastle upon Tyne, UK; Innovation Methodology and Application Research Theme, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - George Merces
- Newcastle University Biosciences Institute, Newcastle upon Tyne, UK; Innovation Methodology and Application Research Theme, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Amanda Thomson
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK; Newcastle University Biosciences Institute, Newcastle upon Tyne, UK; Innovation Methodology and Application Research Theme, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Catharien M U Hilkens
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - John Wills
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Paul Rees
- Department of Biomedical Engineering, Swansea University, Wales, UK; Imaging Platform, Broad Institute of MIT and Harvard, 415 Main Street, Boston, Cambridge, MA, USA
| | - Kasim Jiwa
- Department of Cellular Pathology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Nigel Cooper
- Department of Cellular Pathology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Joaquim Majo
- Department of Cellular Pathology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Helen Ashwin
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | - Christopher J A Duncan
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK; Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Paul M Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | | | - Andrew Filby
- Newcastle University Biosciences Institute, Newcastle upon Tyne, UK; Innovation Methodology and Application Research Theme, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.
| | - Andrew J Fisher
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK; Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
8
|
Cottignies-Calamarte A, He F, Zhu A, Real F, Bomsel M. Protocol to detect infectious SARS-CoV-2 at low levels using in situ hybridization techniques. STAR Protoc 2023; 4:102593. [PMID: 37738115 PMCID: PMC10520661 DOI: 10.1016/j.xpro.2023.102593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/21/2023] [Accepted: 09/05/2023] [Indexed: 09/24/2023] Open
Abstract
Low and persistent levels of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA/protein/virus can be detected in clinical samples months after infection, possibly related to the emergence of SARS-CoV-2 variants or development of long coronavirus disease. Here, we present a protocol to detect low levels of viral RNA together with protein using flow cytometry and microscopy. We describe steps for cell infection with SARS-CoV-2 and quantification by fluorescence in situ hybridization-flow cytometry. We then detail procedures for visualization using immunolabeling and RNAscope. This approach is directly applicable to clinical samples. For complete details on the use and execution of this protocol, please refer to Zhu et al. (2022).1.
Collapse
Affiliation(s)
- Andréa Cottignies-Calamarte
- Laboratory of Mucosal Entry of HIV and Mucosal Immunity, Université de Paris Cité, Paris, France; Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris Cité, Paris, France
| | - Feifan He
- Laboratory of Mucosal Entry of HIV and Mucosal Immunity, Université de Paris Cité, Paris, France; Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris Cité, Paris, France
| | - Aiwei Zhu
- Laboratory of Mucosal Entry of HIV and Mucosal Immunity, Université de Paris Cité, Paris, France; Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris Cité, Paris, France
| | - Fernando Real
- Laboratory of Mucosal Entry of HIV and Mucosal Immunity, Université de Paris Cité, Paris, France; Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris Cité, Paris, France
| | - Morgane Bomsel
- Laboratory of Mucosal Entry of HIV and Mucosal Immunity, Université de Paris Cité, Paris, France; Institut Cochin, INSERM U1016, CNRS UMR8104, Université de Paris Cité, Paris, France.
| |
Collapse
|
9
|
Riegler AN, Benson P, Long K, Leal SM. Differential activation of programmed cell death in patients with severe SARS-CoV-2 infection. Cell Death Discov 2023; 9:420. [PMID: 37985756 PMCID: PMC10662024 DOI: 10.1038/s41420-023-01715-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/26/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes severe lower airway disease and death in a subset of patients. Knowledge on the relative contribution of programmed cell death (PCD) to lung pathology is limited to few human autopsy studies with small sample size/scope, in vitro cell culture, and experimental model systems. In this study, we sought to identify, localize, and quantify activation of apoptosis, ferroptosis, pyroptosis, and necroptosis in FFPE lung tissues from patients that died from severe SARS-CoV-2 infection (n = 28) relative to uninfected controls (n = 13). Immunofluorescence (IF) staining, whole-slide imaging, and Image J software was used to localize and quantify expression of SARS-CoV-2 nucleoprotein and the following PCD protein markers: cleaved Caspase-3, pMLKL, cleaved Gasdermin D, and CD71, respectively. IF showed differential activation of each PCD pathway in infected lungs and dichotomous staining for SARS-CoV-2 nucleoprotein enabling distinction between high (n = 9) vs low viral burden (n = 19). No differences were observed in apoptosis and ferroptosis in SARS-CoV-2 infected lungs relative to uninfected controls. However, both pyroptosis and necroptosis were significantly increased in SARS-CoV-2-infected lungs. Increased pyroptosis was observed in SARS-CoV-2 infected lungs, irrespective of viral burden, suggesting an inflammation-driven mechanism. In contrast, necroptosis exhibited a very strong positive correlation with viral burden (R2 = 0.9925), suggesting a direct SARS-CoV-2 mediated effect. These data indicate a possible novel mechanism for viral-mediated necroptosis and a potential role for both lytic programmed cell death pathways, necroptosis and pyroptosis, in mediating infection outcome.
Collapse
Affiliation(s)
- Ashleigh N Riegler
- Division of Laboratory Medicine, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Paul Benson
- Division of Anatomic Pathology, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth Long
- Division of Infectious Diseases, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sixto M Leal
- Division of Laboratory Medicine, Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
10
|
Ali H, Naseem A, Siddiqui ZI. SARS-CoV-2 Syncytium under the Radar: Molecular Insights of the Spike-Induced Syncytia and Potential Strategies to Limit SARS-CoV-2 Replication. J Clin Med 2023; 12:6079. [PMID: 37763019 PMCID: PMC10531702 DOI: 10.3390/jcm12186079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
SARS-CoV-2 infection induces non-physiological syncytia when its spike fusogenic protein on the surface of the host cells interacts with the ACE2 receptor on adjacent cells. Spike-induced syncytia are beneficial for virus replication, transmission, and immune evasion, and contribute to the progression of COVID-19. In this review, we highlight the properties of viral fusion proteins, mainly the SARS-CoV-2 spike, and the involvement of the host factors in the fusion process. We also highlight the possible use of anti-fusogenic factors as an antiviral for the development of therapeutics against newly emerging SARS-CoV-2 variants and how the fusogenic property of the spike could be exploited for biomedical applications.
Collapse
Affiliation(s)
- Hashim Ali
- Department of Pathology, University of Cambridge, Addenbrookes Hospital, Cambridge CB2 0QQ, UK
| | - Asma Naseem
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
| | - Zaheenul Islam Siddiqui
- Diabetes and Obesity Research Center, NYU Grossman Long Island School of Medicine, New York, NY 11501, USA
| |
Collapse
|
11
|
Swain J, Merida P, Rubio K, Bracquemond D, Neyret A, Aguilar-Ordoñez I, Günther S, Barreto G, Muriaux D. F-actin nanostructures rearrangements and regulation are essential for SARS-CoV-2 particle production in host pulmonary cells. iScience 2023; 26:107384. [PMID: 37564698 PMCID: PMC10410521 DOI: 10.1016/j.isci.2023.107384] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/24/2023] [Accepted: 07/11/2023] [Indexed: 08/12/2023] Open
Abstract
Our study focused on deciphering the role of F-actin and related regulatory factors during SARS-CoV-2 particle production and transmission in human pulmonary cells. Quantitative high-resolution microscopies revealed that the late phases of SARS-CoV-2 infection induce a strong rearrangement of F-actin nanostructures dependent on the viral M, E, and N structural proteins. Intracellular vesicles containing viral components are labeled with Rab7 and Lamp1 and are surrounded by F-actin ring-shaped structures, suggesting their role in viral trafficking toward the cell membrane for virus release. Furthermore, filopodia-like nanostructures were loaded with viruses, potentially facilitating their egress and transmission between lung cells. Gene expression analysis revealed the involvement of alpha-actinins under the regulation of the protein kinase N (PKN). The use of a PKN inhibitor efficiently reduces virus particle production, restoring endoplasmic reticulum and F-actin cellular shape. Our results highlight an important role of F-actin rearrangements during the productive phases of SARS-CoV-2 particles.
Collapse
Affiliation(s)
- Jitendriya Swain
- Institute of Research in Infectiology of Montpellier (IRIM), CNRS, University of Montpellier, UMR9004 CNRS, Montpellier, France
| | - Peggy Merida
- Institute of Research in Infectiology of Montpellier (IRIM), CNRS, University of Montpellier, UMR9004 CNRS, Montpellier, France
| | - Karla Rubio
- Université de Lorraine, CNRS, Laboratoire IMoPA, UMR 7365, 54000 Nancy, France
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Ecocampus, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla 72570, Mexico
| | - David Bracquemond
- Institute of Research in Infectiology of Montpellier (IRIM), CNRS, University of Montpellier, UMR9004 CNRS, Montpellier, France
| | - Aymeric Neyret
- CEMIPAI, CNRS, University of Montpellier, UAR3725 CNRS, Montpellier, France
| | | | - Stefan Günther
- ECCPS Bioinformatics and Deep Sequencing, Max-Planck-Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Guillermo Barreto
- Université de Lorraine, CNRS, Laboratoire IMoPA, UMR 7365, 54000 Nancy, France
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Ecocampus, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla 72570, Mexico
| | - Delphine Muriaux
- Institute of Research in Infectiology of Montpellier (IRIM), CNRS, University of Montpellier, UMR9004 CNRS, Montpellier, France
- CEMIPAI, CNRS, University of Montpellier, UAR3725 CNRS, Montpellier, France
| |
Collapse
|
12
|
Riegler A, Benson P, Long K, Leal S. Differential Activation of Programmed Cell Death in Patients with Severe SARS-CoV-2 Infection. RESEARCH SQUARE 2023:rs.3.rs-3059466. [PMID: 37461686 PMCID: PMC10350212 DOI: 10.21203/rs.3.rs-3059466/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
SARS-CoV-2 (SARS-2) causes severe lower airway disease and death in a subset of patients. Knowledge on the relative contribution of programmed cell death (PCD) to lung pathology is limited to few human autopsy studies with small sample size/scope, in vitro cell culture, and experimental model systems. In this study, we sought to identify, localize, and quantify activation of apoptosis, ferroptosis, pyroptosis, and necroptosis in FFPE lung tissues from patients that died from severe SARS-2 infection (n=28) relative to uninfected controls (n=13). Immunofluorescence (IF) staining, whole-slide imaging, and Image J software was used to localize and quantify expression of SARS-2 nucleoprotein and the following PCD protein markers: cleaved Caspase-3, pMLKL, cleaved Gasdermin D, and CD71, respectively. IF showed differential activation of each PCD pathway in SARS-2 infected lungs and dichotomous staining for SARS-2 nucleoprotein enabling distinction between high (n=9) vs low viral burden (n= 19). No differences were observed in apoptosis and ferroptosis in SARS-2 infected lungs relative to uninfected controls. However, both pyroptosis and necroptosis were significantly increased in SARS-2 infected lungs. Increased pyroptosis was observed in SARS-2 infected lungs, irrespective of viral burden, suggesting an inflammation-driven mechanism. In contrast, necroptosis exhibited a very strong positive correlation with viral burden (R2=0.9925), suggesting a direct SARS-2 mediated effect. These data indicate a possible novel mechanism for viral-mediated necroptosis and a potential role for both lytic programmed cell death pathways, necroptosis and pyroptosis, in mediating infection outcome.
Collapse
|
13
|
Prebensen C, Lefol Y, Myhre PL, Lüders T, Jonassen C, Blomfeldt A, Omland T, Nilsen H, Berdal JE. Longitudinal whole blood transcriptomic analysis characterizes neutrophil activation and interferon signaling in moderate and severe COVID-19. Sci Rep 2023; 13:10368. [PMID: 37365222 PMCID: PMC10293211 DOI: 10.1038/s41598-023-37606-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 06/24/2023] [Indexed: 06/28/2023] Open
Abstract
A maladaptive inflammatory response has been implicated in the pathogenesis of severe COVID-19. This study aimed to characterize the temporal dynamics of this response and investigate whether severe disease is associated with distinct gene expression patterns. We performed microarray analysis of serial whole blood RNA samples from 17 patients with severe COVID-19, 15 patients with moderate disease and 11 healthy controls. All study subjects were unvaccinated. We assessed whole blood gene expression patterns by differential gene expression analysis, gene set enrichment, two clustering methods and estimated relative leukocyte abundance using CIBERSORT. Neutrophils, platelets, cytokine signaling, and the coagulation system were activated in COVID-19, and this broad immune activation was more pronounced in severe vs. moderate disease. We observed two different trajectories of neutrophil-associated genes, indicating the emergence of a more immature neutrophil phenotype over time. Interferon-associated genes were strongly enriched in early COVID-19 before falling markedly, with modest severity-associated differences in trajectory. In conclusion, COVID-19 necessitating hospitalization is associated with a broad inflammatory response, which is more pronounced in severe disease. Our data suggest a progressively more immature circulating neutrophil phenotype over time. Interferon signaling is enriched in COVID-19 but does not seem to drive severe disease.
Collapse
Affiliation(s)
- Christian Prebensen
- Department of Infectious Diseases, Oslo University Hospital, Kirkeveien 166, 0450, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Yohan Lefol
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, University of Oslo, Oslo, Norway
| | - Peder L Myhre
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Torben Lüders
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway
| | | | - Anita Blomfeldt
- Department of Microbiology and Infection Control, Akershus University Hospital, Lørenskog, Norway
| | - Torbjørn Omland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Hilde Nilsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, University of Oslo, Oslo, Norway
| | - Jan-Erik Berdal
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
14
|
Gheban-Roșca IA, Gheban BA, Pop B, Mironescu DC, Siserman VC, Jianu EM, Drugan T, Bolboacă SD. Identification of Histopathological Biomarkers in Fatal Cases of Coronavirus Disease: A Study on Lung Tissue. Diagnostics (Basel) 2023; 13:2039. [PMID: 37370934 DOI: 10.3390/diagnostics13122039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
We aimed to evaluate the primary lung postmortem macro- and microscopic biomarkers and factors associated with diffuse alveolar damage in patients with fatal coronavirus (COVID-19). We retrospectively analyzed lung tissue collected from autopsies performed in Cluj-Napoca, Romania, between April 2020 and April 2021 on patients with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We examined 79 patients with confirmed SARS-CoV-2 infection, ages 34 to 96 years, split into two groups using the cut-off value of 70 years. Arterial hypertension (38%) and type 2 diabetes mellitus (19%) were the most common comorbidities with similar distribution between groups (p-values > 0.14). Macroscopically, bloody exudate was more frequently observed among patients < 70 years (33/36 vs. 29/43, p-value = 0.0091). Diffuse alveolar damage (53.1%) was similarly observed among the evaluated groups (p-value = 0.1354). Histopathological biomarkers of alveolar edema in 83.5% of patients, interstitial pneumonia in 74.7%, and microthrombi in 39.2% of cases were most frequently observed. Half of the evaluated lungs had an Ashcroft score of up to 2 and an alveolar air capacity of up to 12.5%. Bronchopneumonia (11/43 vs. 3/36, p-value = 0.0456) and interstitial edema (9/43 vs. 2/36, p-value = 0.0493) were significantly more frequent in older patients. Age (median: 67.5 vs. 77 years, p-value = 0.023) and infection with the beta variant of the virus (p-value = 0.0071) proved to be significant factors associated with diffuse alveolar damage.
Collapse
Affiliation(s)
- Ioana-Andreea Gheban-Roșca
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
- Clinical Hospital of Infectious Diseases, 400003 Cluj-Napoca, Romania
| | - Bogdan-Alexandru Gheban
- Rouen University Hospital-Charles-Nicolle, 76000 Rouen, France
- Department of Histology, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Bogdan Pop
- The Oncology Institute "Prof. Dr. Ion Chiricuță", 400015 Cluj-Napoca, Romania
- Department of Anatomic Pathology, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Daniela-Cristina Mironescu
- Institute of Legal Medicine, 400006 Cluj-Napoca, Romania
- Department of Forensic Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Vasile Costel Siserman
- Institute of Legal Medicine, 400006 Cluj-Napoca, Romania
- Department of Forensic Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Elena Mihaela Jianu
- Department of Histology, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Tudor Drugan
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Sorana D Bolboacă
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
15
|
Labbé V, Contou D, Heming N, Megarbane B, Razazi K, Boissier F, Ait-Oufella H, Turpin M, Carreira S, Robert A, Monchi M, Souweine B, Preau S, Doyen D, Vivier E, Zucman N, Dres M, Fejjal M, Noel-Savina E, Bachir M, Jaffal K, Timsit JF, Picos SA, Mariotte E, Martis N, Juguet W, Melica G, Rondeau P, Audureau E, Mekontso Dessap A. Effects of Standard-Dose Prophylactic, High-Dose Prophylactic, and Therapeutic Anticoagulation in Patients With Hypoxemic COVID-19 Pneumonia: The ANTICOVID Randomized Clinical Trial. JAMA Intern Med 2023:2802821. [PMID: 36946232 PMCID: PMC10034664 DOI: 10.1001/jamainternmed.2023.0456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Importance Given the high risk of thrombosis and anticoagulation-related bleeding in patients with hypoxemic COVID-19 pneumonia, identifying the lowest effective dose of anticoagulation therapy for these patients is imperative. Objectives To determine whether therapeutic anticoagulation (TA) or high-dose prophylactic anticoagulation (HD-PA) decreases mortality and/or disease duration compared with standard-dose prophylactic anticoagulation (SD-PA), and whether TA outperforms HD-PA; and to compare the net clinical outcomes among the 3 strategies. Design, Settings, and Participants The ANTICOVID randomized clinical open-label trial included patients with hypoxemic COVID-19 pneumonia requiring supplemental oxygen and having no initial thrombosis on chest computer tomography with pulmonary angiogram at 23 health centers in France from April 14 to December 13, 2021. Of 339 patients randomized, 334 were included in the primary analysis-114 patients in the SD-PA group, 110 in the HD-PA, and 110 in the TA. At randomization, 90% of the patients were in the intensive care unit. Data analyses were performed from April 13, 2022, to January 3, 2023. Interventions Patients were randomly assigned (1:1:1) to receive either SD-PA, HD-PA, or TA with low-molecular-weight or unfractionated heparin for 14 days. Main Outcomes and Measures A hierarchical criterion of all-cause mortality followed by time to clinical improvement at day 28. Main secondary outcome was net clinical outcome at day 28 (composite of thrombosis, major bleeding, and all-cause death). Results Among the study population of 334 individuals (mean [SD] age, 58.3 [13.0] years; 226 [67.7%] men and 108 [32.3%] women), use of HD-PA and SD-PA had similar probabilities of favorable outcome (47.3% [95% CI, 39.9% to 54.8%] vs 52.7% [95% CI, 45.2% to 60.1%]; P = .48), as did TA compared with SD-PA (50.9% [95% CI, 43.4% to 58.3%] vs 49.1% [95% CI, 41.7% to 56.6%]; P = .82) and TA compared with HD-PA (53.5% [95% CI 45.8% to 60.9%] vs 46.5% [95% CI, 39.1% to 54.2%]; P = .37). Net clinical outcome was met in 29.8% of patients receiving SD-PA (20.2% thrombosis, 2.6% bleeding, 14.0% death), 16.4% receiving HD-PA (5.5% thrombosis, 3.6% bleeding, 11.8% death), and 20.0% receiving TA (5.5% thrombosis, 3.6% bleeding, 12.7% death). Moreover, HD-PA and TA use significantly reduced thrombosis compared with SD-PA (absolute difference, -14.7 [95% CI -6.2 to -23.2] and -14.7 [95% CI -6.2 to -23.2], respectively). Use of HD-PA significantly reduced net clinical outcome compared with SD-PA (absolute difference, -13.5; 95% CI -2.6 to -24.3). Conclusions and Relevance This randomized clinical trial found that compared with SD-PA, neither HD-PA nor TA use improved the primary hierarchical outcome of all-cause mortality or time to clinical improvement in patients with hypoxemic COVID-19 pneumonia; however, HD-PA resulted in significantly better net clinical outcome by decreasing the risk of de novo thrombosis. Trial Registration ClinicalTrials.gov Identifier: NCT04808882.
Collapse
Affiliation(s)
- Vincent Labbé
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire Tenon, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris, France
- Service des Soins Intensifs, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Bruxelles, Belgium
- Université Paris Est Créteil, Institut Mondor de Recherche Biomédicale, Groupe de Recherche Clinique CARMAS (Cardiovascular and Respiratory Manifestations of Acute lung injury and Sepsis), Créteil, France
| | - Damien Contou
- Service de Réanimation Polyvalente, Centre Hospitalier Victor Dupouy, Argenteuil, France
| | - Nicholas Heming
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire Raymond Poincaré, Assistance Publique-Hôpitaux de Paris, Université Versailles Saint Quentin-Université Paris Saclay, Garches, France
- Laboratoire d'infection et inflammation, Unité 1173, Faculté de Médecine Simone Veil, Institut national de la santé et de la recherche médicale, Université Versailles Saint Quentin - Université Paris Saclay, Garches, France
| | - Bruno Megarbane
- Service de Réanimation Médicale et Toxicologique, Centre Hospitalier Universitaire Lariboisière, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
- Unité Mixte de Recherche en Santé 1144, Institut national de la santé et de la recherche médicale, Université Paris Cité, Paris, France
| | - Keyvan Razazi
- Université Paris Est Créteil, Institut Mondor de Recherche Biomédicale, Groupe de Recherche Clinique CARMAS (Cardiovascular and Respiratory Manifestations of Acute lung injury and Sepsis), Créteil, France
- Service de Médecine Intensive Réanimation, Hôpitaux Universitaires Henri Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est Créteil, Créteil, France
| | - Florence Boissier
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire de Poitiers, Université de Poitiers, Poitiers, France
- Centre d'Investigation Clinique 1402 (Investigations of Sleep, Acute Lung Injury, & Ventilation group), Institut national de la santé et de la recherche médicale, Université de Poitiers, Poitiers, France
| | - Hafid Ait-Oufella
- Service de Médecine Intensive Réanimation, Hôpital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Matthieu Turpin
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire Tenon, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Serge Carreira
- Service d'Anesthésie-Réanimation polyvalente, Hôpital Saint Camille, Bry-sur-Marne, France
| | - Alexandre Robert
- Service de Médecine Intensive Réanimation, Hôpital Simone Veil, Centre Hospitalier de Cannes, Cannes, France
- Centre Méditerranéen de Médecine Moléculaire, Institut national de la santé et de la recherche médicale, Université Côte d'Azur, Nice, France
| | - Mehran Monchi
- Service de Médecine Intensive Réanimation, Groupe Hospitalier Sud Ile de France, Melun, France
| | - Bertrand Souweine
- Service de Médecine Intensive Réanimation, Hôpital Universitaire Gabriel-Montpied, Clermont-Ferrand, France
| | - Sebastien Preau
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire Lille, Université de Lille, Lille, France
- Unité 1167, Institut Pasteur de Lille, Institut national de la santé et de la recherche médicale, Université de Lille, Lille, France
| | - Denis Doyen
- Service de Médecine Intensive Réanimation, Hôpital l'Archet 1, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
- Unité de Recherche Clinique Côte d'Azur, Université Côte d'Azur, Nice, France
| | - Emmanuel Vivier
- Service de Réanimation Polyvalente, Centre Hospitalier Saint Joseph-Saint Luc, Lyon, France
| | - Noémie Zucman
- Service de Médecine Intensive Réanimation, Departement Médico-Universitaire ESPRIT, Centre Hospitalier Universitaire Louis Mourier, Assistance Publique-Hôpitaux de Paris, Colombes, France
- Unité de Formation et de Recherche de Médecine, Université Paris Cité, Paris, France
| | - Martin Dres
- Service de Médecine Intensive Réanimation, Centre Hospitalier Universitaire Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Mohamed Fejjal
- Service de Médecine Intensive Réanimation, Centre Hospitalier Léon Binet, Provins, France
| | - Elise Noel-Savina
- Service de Pneumologie et de Soins Intensifs Respiratoires, Hôpital Larrey, Toulouse, France
| | - Marwa Bachir
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire Tenon, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Karim Jaffal
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Universitaire Raymond Poincaré, Assistance Publique-Hôpitaux de Paris, Garches, France
| | - Jean-François Timsit
- Service de Médecine Intensive et Réanimation Infectieuse, Centre Hospitalier Universitaire Bichat, Assistance Publique-Hôpitaux de Paris, Université de Paris-Cité, Paris, France
- Infection, Anti-microbien, Modélisation, Evolution, Institut National de la Santé et de la Recherche Médicale, Unité 1137, Université de Paris-Cité, Paris, France
| | - Santiago Alberto Picos
- Service de Médecine Intensive Réanimation, Centre Hospitalier La Dracénie De Draguignan, Draguignan, France
| | - Eric Mariotte
- Service de Médecine Intensive-Réanimation, Centre Hospitalier Universitaire Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nihal Martis
- Service de Médecine Interne, Hôpital l'Archet 1, Centre Hospitalier Universitaire de Nice, Nice, France
| | - William Juguet
- Service de Réanimation Médico-Chirurgicale, Centre Hospitalier Universitaire Avicenne, Assistance Publique-Hôpitaux de Paris, Université Sorbonne Paris Nord, Bobigny, France
| | - Giovanna Melica
- Service des Maladies Infectieuses et Tropicales, Hôpitaux Universitaires Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est Créteil, Créteil, France
| | - Paul Rondeau
- Service de Médecine Interne, Hôpital Saint Camille, Bry-sur-Marne, France
| | - Etienne Audureau
- Unité de Recherche Clinique Henri Mondor, Hôpitaux Universitaires Henri Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est Créteil, Créteil, France
- Institut Mondor de Recherche Biomédicale, Unité 955, Institut National de la Santé et de la Recherche Médicale, Université Paris Est Créteil, Créteil, France
| | - Armand Mekontso Dessap
- Université Paris Est Créteil, Institut Mondor de Recherche Biomédicale, Groupe de Recherche Clinique CARMAS (Cardiovascular and Respiratory Manifestations of Acute lung injury and Sepsis), Créteil, France
- Service de Médecine Intensive Réanimation, Hôpitaux Universitaires Henri Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est Créteil, Créteil, France
- Université Paris Est Créteil, Institut Mondor de recherche biomédicale, Institut national de la santé et de la recherche médicale, Créteil, France
| | | |
Collapse
|
16
|
Falcón-Cama V, Montero-González T, Acosta-Medina EF, Guillen-Nieto G, Berlanga-Acosta J, Fernández-Ortega C, Alfonso-Falcón A, Gilva-Rodríguez N, López-Nocedo L, Cremata-García D, Matos-Terrero M, Pentón-Rol G, Valdés I, Oramas-Díaz L, Suarez-Batista A, Noa-Romero E, Cruz-Sui O, Sánchez D, Borrego-Díaz AI, Valdés-Carreras JE, Vizcaino A, Suárez-Alba J, Valdés-Véliz R, Bergado G, González MA, Hernandez T, Alvarez-Arzola R, Ramírez-Suárez AC, Casillas-Casanova D, Lemos-Pérez G, Blanco-Águila OR, Díaz A, González Y, Bequet-Romero M, Marín-Prida J, Hernández-Perera JC, Del Rosario-Cruz L, Marin-Díaz AP, González-Bravo M, Borrajero I, Acosta-Rivero N. Evidence of SARS-CoV-2 infection in postmortem lung, kidney, and liver samples, revealing cellular targets involved in COVID-19 pathogenesis. Arch Virol 2023; 168:96. [PMID: 36842152 PMCID: PMC9968404 DOI: 10.1007/s00705-023-05711-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/29/2022] [Indexed: 02/27/2023]
Abstract
There is an urgent need to understand severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-host interactions involved in virus spread and pathogenesis, which might contribute to the identification of new therapeutic targets. In this study, we investigated the presence of SARS-CoV-2 in postmortem lung, kidney, and liver samples of patients who died with coronavirus disease (COVID-19) and its relationship with host factors involved in virus spread and pathogenesis, using microscopy-based methods. The cases analyzed showed advanced stages of diffuse acute alveolar damage and fibrosis. We identified the SARS-CoV-2 nucleocapsid (NC) in a variety of cells, colocalizing with mitochondrial proteins, lipid droplets (LDs), and key host proteins that have been implicated in inflammation, tissue repair, and the SARS-CoV-2 life cycle (vimentin, NLRP3, fibronectin, LC3B, DDX3X, and PPARγ), pointing to vimentin and LDs as platforms involved not only in the viral life cycle but also in inflammation and pathogenesis. SARS-CoV-2 isolated from a patient´s nasal swab was grown in cell culture and used to infect hamsters. Target cells identified in human tissue samples included lung epithelial and endothelial cells; lipogenic fibroblast-like cells (FLCs) showing features of lipofibroblasts such as activated PPARγ signaling and LDs; lung FLCs expressing fibronectin and vimentin and macrophages, both with evidence of NLRP3- and IL1β-induced responses; regulatory cells expressing immune-checkpoint proteins involved in lung repair responses and contributing to inflammatory responses in the lung; CD34+ liver endothelial cells and hepatocytes expressing vimentin; renal interstitial cells; and the juxtaglomerular apparatus. This suggests that SARS-CoV-2 may directly interfere with critical lung, renal, and liver functions involved in COVID-19-pathogenesis.
Collapse
Affiliation(s)
- Viviana Falcón-Cama
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba. .,Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba.
| | | | - Emilio F Acosta-Medina
- Center for Advanced Studies of Cuba, Havana, Cuba. .,Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba.
| | - Gerardo Guillen-Nieto
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba.,Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba
| | - Jorge Berlanga-Acosta
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba.,Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba
| | - Celia Fernández-Ortega
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba.,Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba
| | | | - Nathalie Gilva-Rodríguez
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Lilianne López-Nocedo
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Daina Cremata-García
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Mariuska Matos-Terrero
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Giselle Pentón-Rol
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba.,Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba
| | - Iris Valdés
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Leonardo Oramas-Díaz
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Anamarys Suarez-Batista
- Department of Virology, Civilian Defense Scientific Research Center (CICDC), Havana, Mayabeque, Cuba
| | - Enrique Noa-Romero
- Department of Virology, Civilian Defense Scientific Research Center (CICDC), Havana, Mayabeque, Cuba
| | - Otto Cruz-Sui
- Department of Virology, Civilian Defense Scientific Research Center (CICDC), Havana, Mayabeque, Cuba
| | | | | | | | | | - José Suárez-Alba
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Rodolfo Valdés-Véliz
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Gretchen Bergado
- Direction of Immunology and Immunotherapy, Center of Molecular Immunology, Havana, Cuba
| | - Miguel A González
- Direction of Immunology and Immunotherapy, Center of Molecular Immunology, Havana, Cuba
| | - Tays Hernandez
- Direction of Immunology and Immunotherapy, Center of Molecular Immunology, Havana, Cuba
| | - Rydell Alvarez-Arzola
- Direction of Immunology and Immunotherapy, Center of Molecular Immunology, Havana, Cuba
| | - Anna C Ramírez-Suárez
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Dionne Casillas-Casanova
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Gilda Lemos-Pérez
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | | | | | | | - Mónica Bequet-Romero
- Center for Genetic Engineering and Biotechnology (CIGB), Ave 31 be/ 158 and 190, Cubanacán, Playa, PO Box 6162, 10699, Havana, Cuba
| | - Javier Marín-Prida
- Center for Research and Biological Evaluations, Institute of Pharmacy and Food, University of Havana, Havana, Cuba
| | | | | | - Alina P Marin-Díaz
- International Orthopedic Scientific Complex 'Frank Pais Garcia', Havana, Cuba
| | - Maritza González-Bravo
- Latin American School of Medicine, Calle Panamericana Km 3 1/2, Playa, 11600, Havana, Cuba
| | | | - Nelson Acosta-Rivero
- Center for Protein Studies, Department of Biochemistry, Faculty of Biology, University of Habana, Calle 25 entre J e I, #455, Plaza de la Revolucion, 10400, Havana, Cuba. .,Department of Infectious Diseases, Centre for Integrative Infectious Disease Research (CIID), Molecular Virology, University of Heidelberg, Medical Faculty Heidelberg, INF 344, GO.1, 69120, Heidelberg, Germany.
| |
Collapse
|
17
|
Iwabuchi S, Tsukahara T, Okayama T, Kitabatake M, Motobayashi H, Shichino S, Imafuku T, Yamaji K, Miyamoto K, Tamura S, Ueha S, Ito T, Murata SI, Kondo T, Ikeo K, Suzuki Y, Matsushima K, Kohara M, Torigoe T, Yamaue H, Hashimoto S. B cell receptor repertoire analysis from autopsy samples of COVID-19 patients. Front Immunol 2023; 14:1034978. [PMID: 36911681 PMCID: PMC9996338 DOI: 10.3389/fimmu.2023.1034978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
Neutralizing antibodies against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are being developed world over. We investigated the possibility of producing artificial antibodies from the formalin fixation and paraffin-embedding (FFPE) lung lobes of a patient who died by coronavirus disease 2019 (COVID-19). The B-cell receptors repertoire in the lung tissue where SARS-CoV-2 was detected were considered to have highly sensitive virus-neutralizing activity, and artificial antibodies were produced by combining the most frequently detected heavy and light chains. Some neutralizing effects against the SARS-CoV-2 were observed, and mixing two different artificial antibodies had a higher tendency to suppress the virus. The neutralizing effects were similar to the immunoglobulin G obtained from healthy donors who had received a COVID-19 mRNA vaccine. Therefore, the use of FFPE lung tissue, which preserves the condition of direct virus sensitization, to generate artificial antibodies may be useful against future unknown infectious diseases.
Collapse
Affiliation(s)
- Sadahiro Iwabuchi
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tomohide Tsukahara
- Department of Pathology, Sapporo Medical University School of Medicine, Hokkaido, Japan
| | - Toshitugu Okayama
- Laboratory of DNA Data Analysis, National Institute of Genetics, Shizuoka, Japan
| | | | - Hideki Motobayashi
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Disease, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Tadashi Imafuku
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kenzaburo Yamaji
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kyohei Miyamoto
- Department of Emergency and Critical Care Medicine, Wakayama Medical University, Wakayama, Japan
| | - Shinobu Tamura
- Department of Emergency and Critical Care Medicine, Wakayama Medical University, Wakayama, Japan
| | - Satoshi Ueha
- Division of Molecular Regulation of Inflammatory and Immune Disease, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Nara, Japan
| | - Shin-Ichi Murata
- Departments of Human Pathology, Wakayama Medical University, Wakayama, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kazuho Ikeo
- Laboratory of DNA Data Analysis, National Institute of Genetics, Shizuoka, Japan
| | - Yutaka Suzuki
- Department of Computational Biology, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Kouji Matsushima
- Division of Molecular Regulation of Inflammatory and Immune Disease, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Michinori Kohara
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, Hokkaido, Japan
| | - Hiroki Yamaue
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan.,Departments of Cancer Immunology, Wakayama Medical University, Wakayama, Japan
| | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
18
|
Arish M, Qian W, Narasimhan H, Sun J. COVID-19 immunopathology: From acute diseases to chronic sequelae. J Med Virol 2023; 95:e28122. [PMID: 36056655 PMCID: PMC9537925 DOI: 10.1002/jmv.28122] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/27/2022] [Accepted: 08/29/2022] [Indexed: 01/17/2023]
Abstract
The clinical manifestation of coronavirus disease 2019 (COVID-19) mainly targets the lung as a primary affected organ, which is also a critical site of immune cell activation by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, recent reports also suggest the involvement of extrapulmonary tissues in COVID-19 pathology. The interplay of both innate and adaptive immune responses is key to COVID-19 management. As a result, a robust innate immune response provides the first line of defense, concomitantly, adaptive immunity neutralizes the infection and builds memory for long-term protection. However, dysregulated immunity, both innate and adaptive, can skew towards immunopathology both in acute and chronic cases. Here we have summarized some of the recent findings that provide critical insight into the immunopathology caused by SARS-CoV-2, in acute and post-acute cases. Finally, we further discuss some of the immunomodulatory drugs in preclinical and clinical trials for dampening the immunopathology caused by COVID-19.
Collapse
Affiliation(s)
- Mohd Arish
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Wei Qian
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Harish Narasimhan
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Jie Sun
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
19
|
A Pleomorphic Puzzle: Heterogeneous Pulmonary Vascular Occlusions in Patients with COVID-19. Int J Mol Sci 2022; 23:ijms232315126. [PMID: 36499449 PMCID: PMC9739020 DOI: 10.3390/ijms232315126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Vascular occlusions in patients with coronavirus diseases 2019 (COVID-19) have been frequently reported in severe outcomes mainly due to a dysregulation of neutrophils mediating neutrophil extracellular trap (NET) formation. Lung specimens from patients with COVID-19 have previously shown a dynamic morphology, categorized into three types of pleomorphic occurrence based on histological findings in this study. These vascular occlusions in lung specimens were also detected using native endogenous fluorescence or NEF in a label-free method. The three types of vascular occlusions exhibit morphology of DNA rich neutrophil elastase (NE) poor (type I), NE rich DNA poor (type II), and DNA and NE rich (type III) cohort of eleven patients with six males and five females. Age and gender have been presented in this study as influencing variables linking the occurrence of several occlusions with pleomorphic contents within a patient specimen and amongst them. This study reports the categorization of pleomorphic occlusions in patients with COVID-19 and the detection of these occlusions in a label-free method utilizing NEF.
Collapse
|
20
|
Bhattacharya M. Insights from Transcriptomics: CD163 + Profibrotic Lung Macrophages in COVID-19. Am J Respir Cell Mol Biol 2022; 67:520-527. [PMID: 35675555 PMCID: PMC9651197 DOI: 10.1165/rcmb.2022-0107tr] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 06/08/2022] [Indexed: 11/24/2022] Open
Abstract
Coronavirus disease (COVID-19) begins with upper airway symptoms but proceeds in a significant proportion of patients to life-threatening infection of the lower respiratory tract, where an exuberant inflammatory response, edema, and adverse parenchymal remodeling impair gas exchange. Respiratory failure is caused initially by flooding of the airspaces with plasma exudate, sloughed epithelium, and inflammatory cells. For many patients with COVID-19, this acute phase has been observed to give way to a prolonged course of acute respiratory distress syndrome, and a significant proportion of patients go on to develop fibroproliferative remodeling of the lung parenchyma, which lengthens the duration of respiratory impairment and mechanical ventilation. Monocyte-derived macrophages have previously been implicated in the fibrotic phase of lung injury in multiple models. From several recent studies that used single-cell genomic techniques, a profile of the transcriptomic state of COVID-19 lung macrophages has emerged. Linkages have been made between these macrophages, which are monocyte-derived and CD163+, and profibrotic macrophages found in other contexts, including animal models of fibrosis and idiopathic pulmonary fibrosis. Here, emerging concepts of macrophage profibrotic function in COVID-19 are highlighted with a focus on gaps in knowledge to be addressed by future research.
Collapse
Affiliation(s)
- Mallar Bhattacharya
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| |
Collapse
|
21
|
Gál P, Brábek J, Holub M, Jakubek M, Šedo A, Lacina L, Strnadová K, Dubový P, Hornychová H, Ryška A, Smetana K. Autoimmunity, cancer and COVID-19 abnormally activate wound healing pathways: critical role of inflammation. Histochem Cell Biol 2022; 158:415-434. [PMID: 35867145 PMCID: PMC9305064 DOI: 10.1007/s00418-022-02140-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2022] [Indexed: 11/30/2022]
Abstract
Recent evidence indicates that targeting IL-6 provides broad therapeutic approaches to several diseases. In patients with cancer, autoimmune diseases, severe respiratory infections [e.g. coronavirus disease 2019 (COVID-19)] and wound healing, IL-6 plays a critical role in modulating the systemic and local microenvironment. Elevated serum levels of IL-6 interfere with the systemic immune response and are associated with disease progression and prognosis. As already noted, monoclonal antibodies blocking either IL-6 or binding of IL-6 to receptors have been used/tested successfully in the treatment of rheumatoid arthritis, many cancer types, and COVID-19. Therefore, in the present review, we compare the impact of IL-6 and anti-IL-6 therapy to demonstrate common (pathological) features of the studied diseases such as formation of granulation tissue with the presence of myofibroblasts and deposition of new extracellular matrix. We also discuss abnormal activation of other wound-healing-related pathways that have been implicated in autoimmune disorders, cancer or COVID-19.
Collapse
Affiliation(s)
- Peter Gál
- Department of Pharmacology, Pavol Jozef Šafárik University, Košice, Slovak Republic
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases, Košice, Slovak Republic
- Prague Burn Centre, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Faculty of Science, Charles University, 120 00 Prague 2, Czech Republic
- BIOCEV, Faculty of Science, Charles University, 252 50 Vestec, Czech Republic
| | - Michal Holub
- Department of Infectious Diseases, First Faculty of Medicine, Military University Hospital Prague and Charles University, 160 00 Prague, Czech Republic
| | - Milan Jakubek
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, 166 28 Prague 6, Czech Republic
| | - Aleksi Šedo
- Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University, 120 00 Praha 2, Czech Republic
| | - Lukáš Lacina
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
- Department of Dermatovenereology, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
| | - Karolína Strnadová
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
| | - Petr Dubový
- Institute of Anatomy, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Helena Hornychová
- The Fingerland Department of Pathology, Faculty of Medicine Hradec Králové, Charles University, 500 05 Hradec Králové, Czech Republic
| | - Aleš Ryška
- The Fingerland Department of Pathology, Faculty of Medicine Hradec Králové, Charles University, 500 05 Hradec Králové, Czech Republic
| | - Karel Smetana
- BIOCEV, First Faculty of Medicine, Charles University, 252 50 Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00 Prague 2, Czech Republic
| |
Collapse
|
22
|
Chavda VP, Patel AB, Pandya A, Vora LK, Patravale V, Tambuwala ZM, Aljabali AAA, Serrano-Aroca Á, Mishra V, Tambuwala MM. Co-infection associated with SARS-CoV-2 and their management. Future Sci OA 2022; 8:FSO819. [PMID: 36788985 PMCID: PMC9912272 DOI: 10.2144/fsoa-2022-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 10/18/2022] [Indexed: 02/05/2023] Open
Abstract
SARS-CoV-2 was discovered in Wuhan, China and quickly spread throughout the world. This deadly virus moved from person to person, resulting in severe pneumonia, fever, chills and hypoxia. Patients are still experiencing problems after recovering from COVID-19. This review covers COVID-19 and associated issues following recovery from COVID-19, as well as multiorgan damage risk factors and treatment techniques. Several unusual illnesses, including mucormycosis, white fungus infection, happy hypoxia and other systemic abnormalities, have been reported in recovered individuals. In children, multisystem inflammatory syndrome with COVID-19 (MIS-C) is identified. The reasons for this might include uncontrollable steroid usage, reduced immunity, uncontrollable diabetes mellitus and inadequate care following COVID-19 recovery.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics & Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad, Gujarat, 380009, India
| | - Aayushi B Patel
- Pharmacy Section, LM College of Pharmacy, Ahmedabad, Gujarat, 380058, India
| | - Anjali Pandya
- Department of Pharmaceutical Sciences & Technology, Institute of Chemical Technology, Mumbai, 400 019, India
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Vandana Patravale
- Department of Pharmaceutical Sciences & Technology, Institute of Chemical Technology, Mumbai, 400 019, India
| | - Zara M Tambuwala
- College of Science, University of Lincoln, Brayford Campus, Lincoln, LN6 7TS, UK
| | - Alaa AA Aljabali
- Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Faculty of Pharmacy, Irbid, 566, Jordan
| | - Ángel Serrano-Aroca
- Biomaterials & Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, Valencia, 46001, Spain
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Murtaza M Tambuwala
- Lincoln Medical School University of Lincoln, Brayford Campus, Lincoln, LN6 7TS, UK
| |
Collapse
|
23
|
Siserman CV, Jeican II, Gheban D, Anton V, Mironescu D, Șușman S, Vică ML, Lazăr M, Aluaș M, Toader C, Albu S. Fatal Form of COVID-19 in a Young Male Bodybuilder Anabolic Steroid Using: The First Autopsied Case. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1373. [PMID: 36295534 PMCID: PMC9611349 DOI: 10.3390/medicina58101373] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022]
Abstract
We report the case of a 34-year-old male patient, a bodybuilding trainer and user of anabolic androgenic steroids (AASs) for 16 years. He was found in cardio-respiratory arrest in his home. By performing a medico-legal autopsy, a severe form of COVID-19, aortic atherosclerotic plaques, and an old myocardial infarction was found. The SARS-CoV-2 RT-PCR test on necroptic lung fragments was positive, with a B.1.258 genetic line. The histopathological examinations showed microthrombi with endothelitis in the cerebral tissue, massive pulmonary edema, diffuse alveolar damage grade 1, pulmonary thromboembolism, hepatic peliosis, and severe nesidioblastosis. The immunohistochemical examinations showed SARS-CoV-2 positive in the myocardium, lung, kidneys, and pancreas. ACE-2 receptor was positive in the same organs, but also in the spleen and liver. HLA alleles A*03, A*25, B*18, B*35, C*04, C*12, DRB1*04, DRB1*15, DQB1*03, DQB1*06 were also identified. In conclusion, death was due to a genetic predisposition, a long-term abuse of AASs that favored the development of a pluriorganic pathological tissue terrain, and recent consumption of AASs, which influenced the immune system at the time of infection.
Collapse
Affiliation(s)
- Costel Vasile Siserman
- Institute of Legal Medicine, 400006 Cluj-Napoca, Romania
- Department of Legal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Ionuț Isaia Jeican
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
- Infectious Disease Clinical Hospital, 400000 Cluj-Napoca, Romania
| | - Dan Gheban
- Department of Pathology, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
- Department of Pathology, Emergency Clinical Hospital for Children, 400370 Cluj-Napoca, Romania
| | - Vlad Anton
- Department of Medical Biochemistry, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | | | - Sergiu Șușman
- Imogen Medical Research Institute, County Clinical Emergency Hospital, 400000 Cluj-Napoca, Romania
- Department of Histology, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Mihaela Laura Vică
- Institute of Legal Medicine, 400006 Cluj-Napoca, Romania
- Department of Cell and Molecular Biology, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Mihaela Lazăr
- Viral Respiratory Infections Laboratory, Cantacuzino National Military-Medical Institute for Research and Development, 050096 Bucharest, Romania
| | - Maria Aluaș
- Department of Oral Health, Iuliu Hatieganu University of Medicine and Pharmacy, Victor Babeș Str., No. 15, 400012 Cluj-Napoca, Romania
| | - Corneliu Toader
- Clinic of Neurosurgery, National Institute of Neurology and Neurovascular Diseases, 041914 Bucharest, Romania
| | - Silviu Albu
- Department of Head and Neck Surgery and Otorhinolaryngology, University Clinical Hospital of Railway Company, Iuliu Hatieganu University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania
| |
Collapse
|
24
|
Xiang M, Wu X, Jing H, Liu L, Wang C, Wang Y, Novakovic VA, Shi J. The impact of platelets on pulmonary microcirculation throughout COVID-19 and its persistent activating factors. Front Immunol 2022; 13:955654. [PMID: 36248790 PMCID: PMC9559186 DOI: 10.3389/fimmu.2022.955654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 08/15/2022] [Indexed: 12/05/2022] Open
Abstract
Patients with COVID-19 often have hypoxemia, impaired lung function, and abnormal imaging manifestations in acute and convalescent stages. Alveolar inflammation, pulmonary vasculitis, and thromboembolism synergistically damage the blood-air barrier, resulting in increased pulmonary permeability and gas exchange disorders. The incidence of low platelet counts correlates with disease severity. Platelets are also involved in the impairment of pulmonary microcirculation leading to abnormal lung function at different phases of COVID-19. Activated platelets lose the ability to protect the integrity of blood vessel walls, increasing the permeability of pulmonary microvasculature. High levels of platelet activation markers are observed in both mild and severe cases, short and long term. Therefore, the risk of thrombotic events may always be present. Vascular endothelial injury, immune cells, inflammatory mediators, and hypoxia participate in the high reactivity and aggregation of platelets in various ways. Microvesicles, phosphatidylserine (PS), platelets, and coagulation factors are closely related. The release of various cell-derived microvesicles can be detected in COVID-19 patients. In addition to providing a phospholipid surface for the synthesis of intrinsic factor Xase complex and prothrombinase complex, exposed PS also promotes the decryption of tissue factor (TF) which then promotes coagulant activity by complexing with factor VIIa to activate factor X. The treatment of COVID-19 hypercoagulability and thrombosis still focuses on early intervention. Antiplatelet therapy plays a role in relieving the disease, inhibiting the formation of the hypercoagulable state, reducing thrombotic events and mortality, and improving sequelae. PS can be another potential target for the inhibition of hypercoagulable states.
Collapse
Affiliation(s)
- Mengqi Xiang
- Department of Hematology, the First Hospital, Harbin Medical University, Harbin, China
| | - Xiaoming Wu
- Department of Hematology, the First Hospital, Harbin Medical University, Harbin, China
| | - Haijiao Jing
- Department of Hematology, the First Hospital, Harbin Medical University, Harbin, China
| | - Langjiao Liu
- Department of Hematology, the First Hospital, Harbin Medical University, Harbin, China
| | - Chunxu Wang
- Department of Hematology, the First Hospital, Harbin Medical University, Harbin, China
| | - Yufeng Wang
- Department of Hematology, the First Hospital, Harbin Medical University, Harbin, China
| | - Valerie A. Novakovic
- Department of Research, Veterans Affairs (VA) Boston Healthcare System, Harvard Medical School, Boston, MA, United States
| | - Jialan Shi
- Department of Hematology, the First Hospital, Harbin Medical University, Harbin, China
- Department of Research, Veterans Affairs (VA) Boston Healthcare System, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
- *Correspondence: Jialan Shi, ;
| |
Collapse
|
25
|
Roy B, Runa SA. SARS-CoV-2 infection and diabetes: Pathophysiological mechanism of multi-system organ failure. World J Virol 2022; 11:252-274. [PMID: 36188734 PMCID: PMC9523319 DOI: 10.5501/wjv.v11.i5.252] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/25/2022] [Accepted: 08/01/2022] [Indexed: 02/05/2023] Open
Abstract
Since the discovery of the coronavirus disease 2019 outbreak, a vast majority of studies have been carried out that confirmed the worst outcome of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in people with preexisting health conditions, including diabetes, obesity, hypertension, cancer, and cardiovascular diseases. Likewise, diabetes itself is one of the leading causes of global public health concerns that impose a heavy global burden on public health as well as socio-economic development. Both diabetes and SARS-CoV-2 infection have their independent ability to induce the pathogenesis and severity of multi-system organ failure, while the co-existence of these two culprits can accelerate the rate of disease progression and magnify the severity of the disease. However, the exact pathophysiology of multi-system organ failure in diabetic patients after SARS-CoV-2 infection is still obscure. This review summarized the organ-specific possible molecular mechanisms of SARS-CoV-2 and diabetes-induced pathophysiology of several diseases of multiple organs, including the lungs, heart, kidneys, brain, eyes, gastrointestinal system, and bones, and sub-sequent manifestation of multi-system organ failure.
Collapse
Affiliation(s)
- Bipradas Roy
- Department of Physiology, Wayne State University, Detroit, MI 48201, United States
- Division of Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, United States
| | - Sadia Afrin Runa
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| |
Collapse
|
26
|
Daniell H, Nair SK, Shi Y, Wang P, Montone KT, Shaw PA, Choi GH, Ghani D, Weaver J, Rader DJ, Margulies KB, Collman RG, Laudanski K, Bar KJ. Decrease in Angiotensin-Converting Enzyme activity but not concentration in plasma/lungs in COVID-19 patients offers clues for diagnosis/treatment. Mol Ther Methods Clin Dev 2022; 26:266-278. [PMID: 35818571 PMCID: PMC9258412 DOI: 10.1016/j.omtm.2022.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 07/03/2022] [Indexed: 12/12/2022]
Abstract
Although several therapeutics are used to treat coronavirus disease 2019 (COVID-19) patients, there is still no definitive metabolic marker to evaluate disease severity and recovery or a quantitative test to end quarantine. Because severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infects human cells via the angiotensin-converting-enzyme 2 (ACE2) receptor and COVID-19 is associated with renin-angiotensin system dysregulation, we evaluated soluble ACE2 (sACE2) activity in the plasma/saliva of 80 hospitalized COVID-19 patients and 27 non-COVID-19 volunteers, and levels of ACE2/Ang (1-7) in plasma or membrane (mACE2) in lung autopsy samples. sACE2 activity was markedly reduced (p < 0.0001) in COVID-19 plasma (n = 59) compared with controls (n = 27). Nadir sACE2 activity in early hospitalization was restored during disease recovery, irrespective of patient age, demographic variations, or comorbidity; in convalescent plasma-administered patients (n = 45), restoration was statistically higher than matched controls (n = 22, p = 0.0021). ACE2 activity was also substantially reduced in the saliva of COVID-19 patients compared with controls (p = 0.0065). There is a strong inverse correlation between sACE2 concentration and sACE2 activity and Ang (1-7) levels in participant plasmas. However, there were no difference in membrane ACE2 levels in lungs of autopsy tissues of COVID-19 (n = 800) versus other conditions (n = 300). These clinical observations suggest sACE2 activity as a potential biomarker and therapeutic target for COVID-19.
Collapse
Affiliation(s)
- Henry Daniell
- W. D. Miller Professor & Director of Translational Research, Vice Chair, Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, 547 Levy Building, Philadelphia, PA 19104-6030, USA
| | - Smruti K. Nair
- W. D. Miller Professor & Director of Translational Research, Vice Chair, Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, 547 Levy Building, Philadelphia, PA 19104-6030, USA
| | - Yao Shi
- W. D. Miller Professor & Director of Translational Research, Vice Chair, Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, 547 Levy Building, Philadelphia, PA 19104-6030, USA
| | - Ping Wang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathleen T. Montone
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pamela A. Shaw
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Kaiser Permanente Washington Health Research Group, Seattle, WA, USA
| | - Grace H. Choi
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Danyal Ghani
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - JoEllen Weaver
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel J. Rader
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kenneth B. Margulies
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ronald G. Collman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Krzysztof Laudanski
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Katharine J. Bar
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
27
|
Gonzalez JC, Chakraborty S, Thulin NK, Wang TT. Heterogeneity in IgG-CD16 signaling in infectious disease outcomes. Immunol Rev 2022; 309:64-74. [PMID: 35781671 PMCID: PMC9539944 DOI: 10.1111/imr.13109] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In this review, we discuss how IgG antibodies can modulate inflammatory signaling during viral infections with a focus on CD16a-mediated functions. We describe the structural heterogeneity of IgG antibody ligands, including subclass and glycosylation that impact binding by and downstream activity of CD16a, as well as the heterogeneity of CD16a itself, including allele and expression density. While inflammation is a mechanism required for immune homeostasis and resolution of acute infections, we focus here on two infectious diseases that are driven by pathogenic inflammatory responses during infection. Specifically, we review and discuss the evolving body of literature showing that afucosylated IgG immune complex signaling through CD16a contributes to the overwhelming inflammatory response that is central to the pathogenesis of severe forms of dengue disease and coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Joseph C. Gonzalez
- Department of Medicine, Division of Infectious DiseasesStanford University School of MedicineStanfordCaliforniaUSA,Program in ImmunologyStanford University School of MedicineStanfordCaliforniaUSA
| | - Saborni Chakraborty
- Department of Medicine, Division of Infectious DiseasesStanford University School of MedicineStanfordCaliforniaUSA
| | - Natalie K. Thulin
- Department of ImmunologyUniversity of WashingtonSeattleWashingtonUSA
| | - Taia T. Wang
- Department of Medicine, Division of Infectious DiseasesStanford University School of MedicineStanfordCaliforniaUSA,Program in ImmunologyStanford University School of MedicineStanfordCaliforniaUSA,Department of Microbiology and ImmunologyStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
28
|
Infection of lung megakaryocytes and platelets by SARS-CoV-2 anticipate fatal COVID-19. Cell Mol Life Sci 2022; 79:365. [PMID: 35708858 PMCID: PMC9201269 DOI: 10.1007/s00018-022-04318-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 04/01/2022] [Accepted: 04/19/2022] [Indexed: 12/11/2022]
Abstract
SARS-CoV-2, although not being a circulatory virus, spread from the respiratory tract resulting in multiorgan failures and thrombotic complications, the hallmarks of fatal COVID-19. A convergent contributor could be platelets that beyond hemostatic functions can carry infectious viruses. Here, we profiled 52 patients with severe COVID-19 and demonstrated that circulating platelets of 19 out 20 non-survivor patients contain SARS-CoV-2 in robust correlation with fatal outcome. Platelets containing SARS-CoV-2 might originate from bone marrow and lung megakaryocytes (MKs), the platelet precursors, which were found infected by SARS-CoV-2 in COVID-19 autopsies. Accordingly, MKs undergoing shortened differentiation and expressing anti-viral IFITM1 and IFITM3 RNA as a sign of viral sensing were enriched in the circulation of deadly COVID-19. Infected MKs reach the lung concomitant with a specific MK-related cytokine storm rich in VEGF, PDGF and inflammatory molecules, anticipating fatal outcome. Lung macrophages capture SARS-CoV-2-containing platelets in vivo. The virus contained by platelets is infectious as capture of platelets carrying SARS-CoV-2 propagates infection to macrophages in vitro, in a process blocked by an anti-GPIIbIIIa drug. Altogether, platelets containing infectious SARS-CoV-2 alter COVID-19 pathogenesis and provide a powerful fatality marker. Clinical targeting of platelets might prevent viral spread, thrombus formation and exacerbated inflammation at once and increase survival in COVID-19.
Collapse
|
29
|
Ponce-Campos SD, Díaz JM, Moreno-Agundis D, González-Delgado AL, Andrade-Lozano P, Avelar-González FJ, Hernández-Cuellar E, Torres-Flores F. A Physiotherapy Treatment Plan for Post-COVID-19 Patients That Improves the FEV1, FVC, and 6-Min Walk Values, and Reduces the Sequelae in 12 Sessions. FRONTIERS IN REHABILITATION SCIENCES 2022; 3:907603. [PMID: 36188947 PMCID: PMC9397810 DOI: 10.3389/fresc.2022.907603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causal agent of Coronavirus disease 2019 (COVID-19), a pandemic disease declared in 2020. The clinical manifestations of this pathology are heterogeneous including fever, cough, dyspnea, anosmia, headache, fatigue, taste dysfunction, among others. Survivors of COVID-19 have demonstrated several persistent symptoms derived from its multisystemic physiopathology. These symptoms can be fatigue, dyspnea, chest pain, dry and productive cough, respiratory insufficiency, and psychoemotional disturbance. To reduce and recover from the post-COVID-19 sequelae is fundamental an early and multifactorial medical treatment. Integral post-COVID-19 physiotherapy is a tool to reduce dyspnea, improve lung capacity, decrease psychoemotional alterations, as well as increase the muscle strength affected by this disease. Thus, the aim of this study was to establish a novel physiotherapeutic plan for post-COVID-19 patients, evaluating the effect of this treatment in the reduction of the sequelae in terms of lung capacity, cardio-respiratory, and muscular strength improvements. This was a cross-sectional study in which a protocol of 12 sessions in 4 weeks of physiotherapy was implemented in the patients enrolled. We conducted a medical assessment, an interview, a DASS-21 test, a spirometry, a 6-min walk test, and a hand dynamometer test to evaluate the post-COVID condition of patients before and after the sessions. A total of 42 patients participated in the program. Results of this work showed a decrease of around 50% of post-COVID-19 sequelae and an improvement in the psychoemotional status of patients. Also, we observed an increase of 7.16% in the FEV1 value and 7.56% for FVC. In addition, the maximal functional capacity increased by 0.577 METs, the 6-min walk test performance increased by 13%, and the SpO2 improved by 1.40%. Finally, the handgrip strength test showed an improvement in the left hand and right hand of 2.90 and 2.24 Kg, respectively. We developed this study to propose a novel methodology to provide information for a better treatment and management of post-COVID-19 patients.
Collapse
Affiliation(s)
- Silvia Denise Ponce-Campos
- Unidad Medico Didáctica, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
- Unidad de Medicina Interna, Adscripción al Servicio de Neumología, Hospital General ISSSTE Aguascalientes, Aguascalientes, Mexico
| | - Juan Manuel Díaz
- Unidad Medico Didáctica, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
- *Correspondence: Juan Manuel Díaz
| | | | | | - Paulina Andrade-Lozano
- Departamento de Medicina, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | | | | | | |
Collapse
|
30
|
Wang Z, Zhang Y, Yang R, Wang Y, Guo J, Sun R, Zhou Y, Su L, Ge Q, Feng Y. Landscape of Peripheral Blood Mononuclear Cells and Soluble Factors in Severe COVID-19 Patients With Pulmonary Fibrosis Development. Front Immunol 2022; 13:831194. [PMID: 35558069 PMCID: PMC9088015 DOI: 10.3389/fimmu.2022.831194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/28/2022] [Indexed: 01/08/2023] Open
Abstract
Resulting from severe inflammation and cell destruction, COVID-19 patients could develop pulmonary fibrosis (PF), which remains in the convalescent stage. Nevertheless, how immune response participates in the pathogenesis of PF progression is not well defined. To investigate that question, 12 patients with severe COVID-19 were included in the study. Peripheral mononuclear cell (PBMC) samples were collected shortly after their admission and proceeded for single-cell RNA sequencing (scRNA-seq). After 14 days of discharge, the patients were revisited for chest CT scan. PF index (FI) was computed by AI-assisted CT images. Patients were categorized into FIhi and FIlo based on median of FI. By scRNA-seq analysis, our data demonstrated that frequency of CD4+ activated T cells and Treg cells were approximately 3-fold higher in FIhi patients compared with FIlo ones (p < 0.034 for all). By dissecting the differentially expressed genes, we found an overall downregulation of IFN-responsive genes (STAT1, IRF7, ISG15, ISG20, IFIs, and IFITMs) and S100s alarmins (S100A8, S100A9, S100A12, etc.) in all T-cell clusters, and cytotoxicity-related genes (GZMB, PRF1, and GNLY) in CTLs and γδ T cells in the FIhi cohort, compared with FIlo subjects. The GSEA analysis illustrated decreased expression of genes enriched in IFN signaling, innate immune response, adaptive immune response in T cells, NK cells, and monocytes in FIhi patients compared with FIlo ones. In conclusion, these data indicated that the attenuated IFN-responsive genes and their related signaling pathways could be critical for PF progression in COVID-19 patients.
Collapse
Affiliation(s)
- Zhuolin Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University. National Health Commission (NHC) Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Yang Zhang
- Beijing Youan Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Rirong Yang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, China.,Department of Immunology, School of Preclinical Medicine, Guangxi Medical University, Guangxi, China
| | - Yujia Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University. National Health Commission (NHC) Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Jiapei Guo
- Department of Immunology, School of Basic Medical Sciences, Peking University. National Health Commission (NHC) Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Ruya Sun
- Department of Biomedical Informatics, Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Ministry of Education (MOE) Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yuan Zhou
- Department of Biomedical Informatics, Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, Ministry of Education (MOE) Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Li Su
- Neuroscience Research Institute, Peking University Center of Medical and Health Analysis, Peking University, Beijing, China
| | - Qing Ge
- Department of Immunology, School of Basic Medical Sciences, Peking University. National Health Commission (NHC) Key Laboratory of Medical Immunology (Peking University), Beijing, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yingmei Feng
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
31
|
Gavriilidis E, Antoniadou C, Chrysanthopoulou A, Ntinopoulou M, Smyrlis A, Fotiadou I, Zioga N, Kogias D, Natsi AM, Pelekoudas C, Satiridou E, Bakola SA, Papagoras C, Mitroulis I, Peichamperis P, Mikroulis D, Papadopoulos V, Skendros P, Ritis K. Combined administration of inhaled DNase, baricitinib and tocilizumab as rescue treatment in COVID-19 patients with severe respiratory failure. Clin Immunol 2022; 238:109016. [PMID: 35447311 PMCID: PMC9014660 DOI: 10.1016/j.clim.2022.109016] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/09/2022] [Accepted: 04/13/2022] [Indexed: 02/07/2023]
Abstract
Aiming to reduce mortality in COVID-19 with severe respiratory failure we administered a combined rescue treatment (COMBI) on top of standard-of-care (SOC: dexamethasone/heparin) consisted of inhaled DNase to dissolve thrombogenic neutrophil extracellular traps, plus agents against cytokine-mediated hyperinflammation, namely anti-IL-6-receptor tocilizumab and JAK1/2 inhibitor baricitinib. Patients with PaO2/FiO2 < 100 mmHg were analysed. COMBI group (n = 22) was compared with similar groups that had received SOC alone (n = 26) or SOC plus monotherapy with either IL-1-receptor antagonist anakinra (n = 19) or tocilizumab (n = 11). COMBI was significantly associated with lower in-hospital mortality and intubation rate, shorter duration of hospitalization, and prolonged overall survival after a median follow-up of 110 days. In vitro, COVID-19 plasma induced tissue factor/thrombin pathway in primary lung fibroblasts. This effect was inhibited by the immunomodulatory agents of COMBI providing a mechanistic explanation for the clinical observations. These results support the conduct of randomized trials using combined immunomodulation in COVID-19 to target multiple interconnected pathways of immunothrombosis.
Collapse
Affiliation(s)
- Efstratios Gavriilidis
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Christina Antoniadou
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Akrivi Chrysanthopoulou
- Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Maria Ntinopoulou
- Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Andreas Smyrlis
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Iliana Fotiadou
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Nikoleta Zioga
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Dionysios Kogias
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Anastasia-Maria Natsi
- Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Christos Pelekoudas
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Evangelia Satiridou
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Stefania-Aspasia Bakola
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Charalampos Papagoras
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece; Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Ioannis Mitroulis
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece; Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Paschalis Peichamperis
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Dimitrios Mikroulis
- Department of Cardiovascular Surgery, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | - Vasileios Papadopoulos
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece; Department of Internal Medicine, Xanthi General Hospital, Xanthi, Greece
| | - Panagiotis Skendros
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece; Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece.
| | - Konstantinos Ritis
- First Department of Internal Medicine, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece; Laboratory of Molecular Hematology, Democritus University of Thrace, University Hospital of Alexandroupolis, Alexandroupolis, Greece.
| |
Collapse
|
32
|
Schwab N, Nienhold R, Henkel M, Baschong A, Graber A, Frank A, Mensah N, Koike J, Hernach C, Sachs M, Daun T, Zsikla V, Willi N, Junt T, Mertz KD. COVID-19 Autopsies Reveal Underreporting of SARS-CoV-2 Infection and Scarcity of Co-infections. Front Med (Lausanne) 2022; 9:868954. [PMID: 35492342 PMCID: PMC9046787 DOI: 10.3389/fmed.2022.868954] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) mortality can be estimated based on reliable mortality data. Variable testing procedures and heterogeneous disease course suggest that a substantial number of COVID-19 deaths is undetected. To address this question, we screened an unselected autopsy cohort for the presence of SARS-CoV-2 and a panel of common respiratory pathogens. Lung tissues from 62 consecutive autopsies, conducted during the first and second COVID-19 pandemic waves in Switzerland, were analyzed for bacterial, viral and fungal respiratory pathogens including SARS-CoV-2. SARS-CoV-2 was detected in 28 lungs of 62 deceased patients (45%), although only 18 patients (29%) were reported to have COVID-19 at the time of death. In 23 patients (37% of all), the clinical cause of death and/or autopsy findings together with the presence of SARS-CoV-2 suggested death due to COVID-19. Our autopsy results reveal a 16% higher SARS-CoV-2 infection rate and an 8% higher SARS-CoV-2 related mortality rate than reported by clinicians before death. The majority of SARS-CoV-2 infected patients (75%) did not suffer from respiratory co-infections, as long as they were treated with antibiotics. In the lungs of 5 patients (8% of all), SARS-CoV-2 was found, yet without typical clinical and/or autopsy findings. Our findings suggest that underreporting of COVID-19 contributes substantially to excess mortality. The small percentage of co-infections in SARS-CoV-2 positive patients who died with typical COVID-19 symptoms strongly suggests that the majority of SARS-CoV-2 infected patients died from and not with the virus.
Collapse
Affiliation(s)
- Nathalie Schwab
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Ronny Nienhold
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Maurice Henkel
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
- Department of Radiology, University Hospital Basel, Basel, Switzerland
| | - Albert Baschong
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Anne Graber
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Angela Frank
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Nadine Mensah
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Jacqueline Koike
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Claudia Hernach
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Melanie Sachs
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Till Daun
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Veronika Zsikla
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Niels Willi
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Tobias Junt
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Kirsten D. Mertz
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
33
|
Dhawan S. Therapeutic Potential of Inducible Endogenous Cytoprotective Heme Oxygenase-1 in Mitigating SARS-CoV-2 Infection and Associated Inflammation. Antioxidants (Basel) 2022; 11:662. [PMID: 35453347 PMCID: PMC9028590 DOI: 10.3390/antiox11040662] [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: 02/25/2022] [Revised: 03/23/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
The inducible cytoprotective enzyme heme oxygenase-1 (HO-1) has gained significant recognition in recent years for mediating strong cellular resistance to a broad range of viral infections, regardless of the type of viruses, viral strains, or mutants. HO-1 is not a typical antiviral agent that targets any particular pathogen. It is a "viral tropism independent" endogenous host defense factor that upon induction provides general cellular protection against pathogens. By virtue of HO-1 being widely distributed intracellular enzyme in virtually every cell, this unique host factor presents a novel class of generic host defense system against a variety of viral infections. This Viewpoint proposes pharmacological evaluation of the HO-1-dependent cellular resistance for its potential in mitigating infections by deadly viruses, including the current severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), its variants, and mutants. HO-1-dependent cellular resistance against SARS-CoV-2 can complement current medical modalities for much effective control of the COVID-19 pandemic, especially with constantly emerging new viral variants and limited therapeutic options to treat SARS-CoV-2 infection and associated severe health consequences.
Collapse
Affiliation(s)
- Subhash Dhawan
- Retired Senior FDA Research & Regulatory Scientist, 9890 Washingtonian Blvd., #703, Gaithersburg, MD 20878, USA
| |
Collapse
|
34
|
Nunes MC, Hale MJ, Mahtab S, Mabena FC, Dludlu N, Baillie VL, Thwala BN, Els T, du Plessis J, Laubscher M, Mckenzie S, Mtshali S, Menezes C, Serafin N, van Blydenstein S, Tsitsi M, Dulisse B, Madhi SA. Clinical characteristics and histopathology of COVID-19 related deaths in South African adults. PLoS One 2022; 17:e0262179. [PMID: 35051205 PMCID: PMC8775212 DOI: 10.1371/journal.pone.0262179] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/17/2021] [Indexed: 12/22/2022] Open
Abstract
Comparisons of histopathological features and microbiological findings between decedents with respiratory symptoms due to SARS-CoV-2 infection or other causes, in settings with high prevalence of HIV and Mycobacterium tuberculosis (MTB) infections have not been reported. Deaths associated with a positive ante-mortem SARS-CoV-2 PCR test and/or respiratory disease symptoms at Chris Hani Baragwanath Academic Hospital in Soweto, South Africa from 15th April to 2nd November 2020, during the first wave of the South African COVID-19 epidemic, were investigated. Deceased adult patients had post-mortem minimally-invasive tissue sampling (MITS) performed to investigate for SARS-CoV-2 infection and molecular detection of putative pathogens on blood and lung samples, and histopathology examination of lung, liver and heart tissue. During the study period MITS were done in patients displaying symptoms of respiratory disease including 75 COVID-19-related deaths (COVID+) and 42 non-COVID-19-related deaths (COVID-). The prevalence of HIV-infection was lower in COVID+ (27%) than in the COVID- (64%), MTB detection was also less common among COVID+ (3% vs 13%). Lung histopathology findings showed differences between COVID+ and COVID- in the severity of the morphological appearance of Type-II pneumocytes, alveolar injury and repair initiated by SARS-CoV-2 infection. In the liver necrotising granulomatous inflammation was more common among COVID+. No differences were found in heart analyses. The prevalence of bacterial co-infections was higher in COVID+. Most indicators of respiratory distress syndrome were undifferentiated between COVID+ and COVID- except for Type-II pneumocytes. HIV or MTB infection does not appear in these data to have a meaningful correspondence with COVID-related deaths.
Collapse
Affiliation(s)
- Marta C. Nunes
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Martin J. Hale
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Division of Anatomical Pathology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sana Mahtab
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Fikile C. Mabena
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Paediatrics and Child Health, Chris Hani Baragwanath Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Noluthando Dludlu
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Vicky L. Baillie
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Bukiwe N. Thwala
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Toyah Els
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jeanine du Plessis
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Marius Laubscher
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shakeel Mckenzie
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sihle Mtshali
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Colin Menezes
- Department of Internal Medicine, Chris Hani Baragwanath Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Natali Serafin
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sarah van Blydenstein
- Department of Internal Medicine, Chris Hani Baragwanath Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Merika Tsitsi
- Department of Internal Medicine, Chris Hani Baragwanath Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Brian Dulisse
- BDII Analytics, Atlanta, GA, United States of America
| | - Shabir A. Madhi
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
35
|
Cavallieri F, Sellner J, Zedde M, Moro E. Neurologic complications of coronavirus and other respiratory viral infections. HANDBOOK OF CLINICAL NEUROLOGY 2022; 189:331-358. [PMID: 36031313 PMCID: PMC9418023 DOI: 10.1016/b978-0-323-91532-8.00004-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In humans, several respiratory viruses can have neurologic implications affecting both central and peripheral nervous system. Neurologic manifestations can be linked to viral neurotropism and/or indirect effects of the infection due to endothelitis with vascular damage and ischemia, hypercoagulation state with thrombosis and hemorrhages, systemic inflammatory response, autoimmune reactions, and other damages. Among these respiratory viruses, recent and huge attention has been given to the coronaviruses, especially the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic started in 2020. Besides the common respiratory symptoms and the lung tropism of SARS-CoV-2 (COVID-19), neurologic manifestations are not rare and often present in the severe forms of the infection. The most common acute and subacute symptoms and signs include headache, fatigue, myalgia, anosmia, ageusia, sleep disturbances, whereas clinical syndromes include mainly encephalopathy, ischemic stroke, seizures, and autoimmune peripheral neuropathies. Although the pathogenetic mechanisms of COVID-19 in the various acute neurologic manifestations are partially understood, little is known about long-term consequences of the infection. These consequences concern both the so-called long-COVID (characterized by the persistence of neurological manifestations after the resolution of the acute viral phase), and the onset of new neurological symptoms that may be linked to the previous infection.
Collapse
Affiliation(s)
- Francesco Cavallieri
- Neurology Unit, Neuromotor and Rehabilitation Department, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Johann Sellner
- Department of Neurology, Landesklinikum Mistelbach-Gänserndorf, Mistelbach, Austria,Department of Neurology, Christian Doppler Medical Center, Paracelsus Medical University, Salzburg, Austria
| | - Marialuisa Zedde
- Neurology Unit, Neuromotor and Rehabilitation Department, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Elena Moro
- Division of Neurology, CHU of Grenoble, Grenoble Alpes University, Grenoble Institute of Neurosciences, Grenoble, France,Correspondence to: Elena Moro, Service de neurologie, CHU de Grenoble (Hôpital Nord), Boulevard de la Chantourne, 38043 La Tronche, France. Tel: + 33-4-76-76-94-52, Fax: +33-4-76-76-56-31
| |
Collapse
|
36
|
Abstract
Pulmonary fibrosis is the end stage of a broad range of heterogeneous interstitial lung diseases and more than 200 factors contribute to it. In recent years, the relationship between virus infection and pulmonary fibrosis is getting more and more attention, especially after the outbreak of SARS-CoV-2 in 2019, however, the mechanisms underlying the virus-induced pulmonary fibrosis are not fully understood. Here, we review the relationship between pulmonary fibrosis and several viruses such as Human T-cell leukemia virus (HTLV), Human immunodeficiency virus (HIV), Cytomegalovirus (CMV), Epstein–Barr virus (EBV), Murine γ-herpesvirus 68 (MHV-68), Influenza virus, Avian influenza virus, Middle East Respiratory Syndrome (MERS)-CoV, Severe acute respiratory syndrome (SARS)-CoV and SARS-CoV-2 as well as the mechanisms underlying the virus infection induced pulmonary fibrosis. This may shed new light on the potential targets for anti-fibrotic therapy to treat pulmonary fibrosis induced by viruses including SARS-CoV-2.
Collapse
Affiliation(s)
- Wei Jie Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiao Xiao Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China. .,Guangzhou Laboratory, Bio-island, Guangzhou, China.
| |
Collapse
|