1
|
Cambon A, Guervilly C, Delteil C, Potere N, Bachelier R, Tellier E, Abdili E, Leprince M, Giani M, Polidoro I, Albanese V, Ferrante P, Coffin L, Schiffrin M, Arnaud L, Lacroix R, Roque S, Forel JM, Hraiech S, Daniel L, Papazian L, Dignat-George F, Kaplanski G. Caspase-1 activation, IL-1/IL-6 signature and IFNγ-induced chemokines in lungs of COVID-19 patients. Front Immunol 2025; 15:1493306. [PMID: 39882243 PMCID: PMC11774885 DOI: 10.3389/fimmu.2024.1493306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 12/11/2024] [Indexed: 01/31/2025] Open
Abstract
Rationale COVID-19-associated acute-respiratory distress syndrome (C-ARDS) results from a direct viral injury associated with host excessive innate immune response mainly affecting the lungs. However, cytokine profile in the lung compartment of C-ARDS patients has not been widely studied, nor compared to non-COVID related ARDS (NC-ARDS). Objectives To evaluate caspase-1 activation, IL-1 signature, and other inflammatory cytokine pathways associated with tissue damage using post-mortem lung tissues, bronchoalveolar lavage fluids (BALF), and serum across the spectrum of COVID-19 severity. Methods Histological features were described and activated-caspase-1 labeling was performed in 40 post-mortem biopsies. Inflammatory cytokines were quantified in BALF and serum from 19 steroid-treated-C-ARDSand compared to 19 NC-ARDS. Cytokine concentrations were also measured in serum from 128 COVID-19 patients at different severity stages. Measurements and main results Typical "diffuse alveolar damage" in lung biopsies were associated with activated caspase-1 expression and vascular lesions. Soluble Caspase-1p20, IL-1β, IL-1Ra, IL-6 and at lower level IFNγ and CXCL-10, were highly elevated in BALF from steroid-treated-C-ARDS as well as in NC-ARDS. IL-1β appeared concentrated in BALF, whereas circulating IL-6 and IL-1Ra concentrations were comparable to those in BALF and correlated with severity. TNFα, TNFR1 and CXCL8 however, were significantly higher in NC-ARDS compared to C-ARDS, treated by steroid. Conclusions In the lungs of C-ARDS, both caspase-1 activation with a predominant IL-1β/IL-6 signature and IFNγ -associated chemokines are elevated despite steroid treatment. These pathways may be specifically targeted in ARDS to improve response to treatment and to limit alveolar and vascular lung damage.
Collapse
Affiliation(s)
- Audrey Cambon
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Christophe Guervilly
- Centre d’Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, Marseille, France
- Service de Médecine Intensive Réanimation, Hôpital Nord, Assistance Publique- Hôpitaux de Marseille, Chemin des Bourrely, Marseille, France
| | - Clémence Delteil
- Département de Médecine légale, Hôpital de la Timone, Assistance Publique-Hôpitaux de Marseille, Marseille University, Marseille, France
| | - Nicola Potere
- School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | | | - Edwige Tellier
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Evelyne Abdili
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Marine Leprince
- Service de Médecine interne et d’Immunologie clinique, Assistance Publique - Hôpitaux de Marseille, Hôpital La Conception, Marseille, France
| | - Marco Giani
- School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Ildo Polidoro
- Unit of Legal Medicine, “Santo Spirito” Hospital, Local Health Authority of Pescara, Pescara, Italy
| | - Valentina Albanese
- Unit of Legal Medicine, “Santo Spirito” Hospital, Local Health Authority of Pescara, Pescara, Italy
| | - Paolo Ferrante
- Unit of Legal Medicine, “Santo Spirito” Hospital, Local Health Authority of Pescara, Pescara, Italy
| | | | | | - Laurent Arnaud
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Romaric Lacroix
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Sandrine Roque
- Service de Médecine interne et d’Immunologie clinique, Assistance Publique - Hôpitaux de Marseille, Hôpital La Conception, Marseille, France
| | - Jean-Marie Forel
- Centre d’Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, Marseille, France
- Service de Médecine Intensive Réanimation, Hôpital Nord, Assistance Publique- Hôpitaux de Marseille, Chemin des Bourrely, Marseille, France
| | - Sami Hraiech
- Centre d’Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, Marseille, France
- Service de Médecine Intensive Réanimation, Hôpital Nord, Assistance Publique- Hôpitaux de Marseille, Chemin des Bourrely, Marseille, France
| | - Laurent Daniel
- Service d’Anatomopathologie, APHM, Aix Marseille University, Marseille, France
| | - Laurent Papazian
- Service de Réanimation, Centre Hospitalier de Bastia, Bastia, France
| | - Françoise Dignat-George
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Gilles Kaplanski
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service de Médecine interne et d’Immunologie clinique, Assistance Publique - Hôpitaux de Marseille, Hôpital La Conception, Marseille, France
| |
Collapse
|
2
|
Yip JQ, Oo A, Ng YL, Chin KL, Tan KK, Chu JJH, AbuBakar S, Zainal N. The role of inflammatory gene polymorphisms in severe COVID-19: a review. Virol J 2024; 21:327. [PMID: 39707400 DOI: 10.1186/s12985-024-02597-3] [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: 09/03/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
The COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, has profoundly impacted global healthcare systems and spurred extensive research efforts over the past three years. One critical aspect of the disease is the intricate interplay between the virus and the host immune response, particularly the role of inflammatory gene expression in severe COVID-19. While numerous previous studies have explored the role of genetic polymorphisms in COVID-19, research specifically focusing on inflammatory genes and their associations with disease severity remains limited. This review explores the relationship between severe COVID-19 outcomes and genetic polymorphisms within key inflammatory genes. By investigating the impact of genetic variations on immune responses, which include cytokine production and downstream signalling pathways, we aim to provide a comprehensive overview of how genetic polymorphisms contribute to the variability in disease presentation. Through an in-depth analysis of existing literature, we shed light on potential therapeutic targets and personalized approaches that may enhance our understanding of disease pathogenesis and treatment strategies.
Collapse
Affiliation(s)
- Jia Qi Yip
- Tropical Infectious Diseases Research & Education Centre (TIDREC), Universiti Malaya, 50603, Kuala Lumpur, Malaysia
- Institute for Advanced Studies, Advanced Studies Complex, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Adrian Oo
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore
- Infectious Disease Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yan Ling Ng
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore
- Infectious Disease Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Kim Ling Chin
- Institute for Advanced Studies, Advanced Studies Complex, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kim-Kee Tan
- Tropical Infectious Diseases Research & Education Centre (TIDREC), Universiti Malaya, 50603, Kuala Lumpur, Malaysia
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Justin Jang Hann Chu
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore
- Infectious Disease Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore
- NUSMed Biosafety Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Sazaly AbuBakar
- Tropical Infectious Diseases Research & Education Centre (TIDREC), Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Nurhafiza Zainal
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
3
|
Wang P, Wang Z, Zhang J, Lan C, Zhao Y, Chen X, Li Y, Mei Q, Feng H, Wei S, Xue Z, Gao F, Liu X, Liang Y. Effect of Hyperbaric Oxygen Therapy on Patients with SARS-CoV-2 Infection: A Retrospective Cohort Study. J Multidiscip Healthc 2024; 17:5501-5511. [PMID: 39600716 PMCID: PMC11590646 DOI: 10.2147/jmdh.s486170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
Objective The aim of this study was to evaluate the impact of hyperbaric oxygen therapy (HBOT) on patients with SARS-CoV-2 infection and determine its efficacy and safety in reducing treatment failure events. Methods A retrospective cohort study involving patients with COVID-19 was conducted. Inverse probability of treatment weighting (IPTW) was used to balance covariates between the HBOT and non-HBOT groups. The primary endpoint was the occurrence of a clinical treatment failure event, defined as all-cause mortality, abandonment of treatment, or transfer to the Intensive Care Unit due to worsening condition. Results A total of 720 patients with COVID-19 were enrolled in the study, with 27 patients receiving HBOT and 693 patients not receiving HBOT. The occurrence of treatment failure was significantly lower in the HBOT group compared to the non-HBOT group, with no treatment failure events in the HBOT group versus 36 events in the non-HBOT group. The IPTW database analysis results showed that in comparison to the non-HBOT group, the hazard ratio (HR) for treatment failure in the HBOT group was less than 0.001 (95% CI: <0.001 ~ <0.001, p<0.001). Lymphocyte count >0.8×109/L and HBOT was associated with a significantly lower risk of treatment failure. Glucocorticoid use was associated with a higher risk of treatment failure. The incidence of venous thrombosis events was significantly higher in the HBOT group compared to the non-HBOT group. Conclusion This study revealed that adjunctive HBOT significantly reduces the risk of treatment failure in patients with COVID-19 and is associated with satisfactory safety. HBOT shows promise as a beneficial therapy for improving outcomes in COVID-19-infected patients.
Collapse
Affiliation(s)
- Pingzhi Wang
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Zhengtao Wang
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Junyan Zhang
- Department of Clinical Epidemiology and Evidence-Based Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Caiqin Lan
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yani Zhao
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xiaoqing Chen
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Yu Li
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Qi Mei
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Huijing Feng
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Shuang Wei
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Zhifeng Xue
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Fang Gao
- Department of Prevention Care in HealthCare, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Xiaolei Liu
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
- Yunnan Provincial Clinical Research Center for Neurological Diseases, Kunming, People’s Republic of China
| | - Ying Liang
- Department of Rehabilitation Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| |
Collapse
|
4
|
Battista F, Schultz CL. Sampling and collector biases as taphonomic filters: an overview. AN ACAD BRAS CIENC 2024; 96:e20231242. [PMID: 39607126 DOI: 10.1590/0001-3765202420231242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 08/11/2024] [Indexed: 11/29/2024] Open
Abstract
Sampling (or sample) bias is a widespread concern in scientific research, across several disciplines. The concept of sampling bias originated in statistical studies. The consequence of a biased sample is that scientists will conclude about a population different from their target. In paleontology, sampling bias is typically related to fieldwork context. Human factors, known as sullegic (e.g. collection method, historic resampling) and trephic (transport, and curatorial processes) factors can generate bias. Other factor is the ugly fossil syndrome (i.e. choosing based on completeness of the specimens, or according to the researcher interest). Thus, sampling implies information loss. Biased samples add artificial results and can be considered an additional taphonomic filter. Therefore, sampling bias and the collector role and choices are frequently linked and almost indistinguishable. Compared to the treatment of this topic in other research fields, little related discussion has been held in vertebrate paleontology, especially regarding what happens at the interface between the biosphere, lithosphere, and anthroposphere, and during the transition between the anthroposphere and the patrisphere (museums). Numerous questions still arise. As a community, we must pay attention, to minimize the loss of information, from field activities to cataloging.
Collapse
Affiliation(s)
- Francesco Battista
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Geociências, Instituto de Geociências, Av. Bento Gonçalves, 9500, 91501-970 Porto Alegre, RS, Brazil
| | - Cesar L Schultz
- Universidade Federal do Rio Grande do Sul, Programa de Pós-Graduação em Geociências, Instituto de Geociências, Av. Bento Gonçalves, 9500, 91501-970 Porto Alegre, RS, Brazil
| |
Collapse
|
5
|
Da Silva Filho J, Herder V, Gibbins MP, Dos Reis MF, Melo GC, Haley MJ, Judice CC, Val FFA, Borba M, Tavella TA, de Sousa Sampaio V, Attipa C, McMonagle F, Wright D, de Lacerda MVG, Costa FTM, Couper KN, Marcelo Monteiro W, de Lima Ferreira LC, Moxon CA, Palmarini M, Marti M. A spatially resolved single-cell lung atlas integrated with clinical and blood signatures distinguishes COVID-19 disease trajectories. Sci Transl Med 2024; 16:eadk9149. [PMID: 39259811 DOI: 10.1126/scitranslmed.adk9149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/15/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024]
Abstract
COVID-19 is characterized by a broad range of symptoms and disease trajectories. Understanding the correlation between clinical biomarkers and lung pathology during acute COVID-19 is necessary to understand its diverse pathogenesis and inform more effective treatments. Here, we present an integrated analysis of longitudinal clinical parameters, peripheral blood markers, and lung pathology in 142 Brazilian patients hospitalized with COVID-19. We identified core clinical and peripheral blood signatures differentiating disease progression between patients who recovered from severe disease compared with those who succumbed to the disease. Signatures were heterogeneous among fatal cases yet clustered into two patient groups: "early death" (<15 days until death) and "late death" (>15 days). Progression to early death was characterized systemically and in lung histopathological samples by rapid endothelial and myeloid activation and the presence of thrombi associated with SARS-CoV-2+ macrophages. In contrast, progression to late death was associated with fibrosis, apoptosis, and SARS-CoV-2+ epithelial cells in postmortem lung tissue. In late death cases, cytotoxicity, interferon, and T helper 17 (TH17) signatures were only detectable in the peripheral blood after 2 weeks of hospitalization. Progression to recovery was associated with higher lymphocyte counts, TH2 responses, and anti-inflammatory-mediated responses. By integrating antemortem longitudinal blood signatures and spatial single-cell lung signatures from postmortem lung samples, we defined clinical parameters that could be used to help predict COVID-19 outcomes.
Collapse
Affiliation(s)
- João Da Silva Filho
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Vanessa Herder
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Matthew P Gibbins
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Monique Freire Dos Reis
- Department of Education and Research, Oncology Control Centre of Amazonas State (FCECON), Manaus, Brazil
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Federal University of Amazonas, Manaus, Brazil
- Amazonas Oncology Control Center Foundation, Manaus, Brazil
| | | | - Michael J Haley
- Department of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK
| | - Carla Cristina Judice
- Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Fernando Fonseca Almeida Val
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Mayla Borba
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Delphina Rinaldi Abdel Aziz Emergency Hospital (HPSDRA), Manaus, Brazil
| | - Tatyana Almeida Tavella
- Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
- INSERM U1016, CNRS UMR8104, University of Paris Cité, Institut Cochin, Paris, France
| | | | - Charalampos Attipa
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Fiona McMonagle
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Glasgow Imaging Facility/School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Derek Wright
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Marcus Vinicius Guimaraes de Lacerda
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
- Instituto Leônidas e Maria Deane, Fiocruz, Manaus, Brazil
- University of Texas Medical Branch, Galveston, TX, USA
| | | | - Kevin N Couper
- Department of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK
| | - Wuelton Marcelo Monteiro
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Luiz Carlos de Lima Ferreira
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Christopher Alan Moxon
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- (C.A.M.)
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- (M.P.)
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
- (M.M.)
| |
Collapse
|
6
|
Li S, Du Z, Ma H, Cai L, Liu X, He J. Mendelian randomization provides causal association between COVID-19 and thyroid cancer: insights from a multi-cancer analysis. Front Oncol 2024; 14:1419020. [PMID: 39319057 PMCID: PMC11419959 DOI: 10.3389/fonc.2024.1419020] [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: 04/17/2024] [Accepted: 08/19/2024] [Indexed: 09/26/2024] Open
Abstract
Since the onset of the COVID-19 pandemic, the SARS-CoV-2 virus has caused over 600 million confirmed infections and more than 6.8 million deaths worldwide, with ongoing implications for human health. COVID-19 has been extensively documented to have extrapulmonary manifestations due to the widespread expression of necessary ACE2 receptors in the human body. Nevertheless, the association between COVID-19 and cancer risk remains inadequately explored. This study employs Mendelian randomization (MR) methods to examine the causal relationship between genetic variations associated with COVID-19 and the risk of developing cancer. The findings indicate that COVID-19 has negligible impact on most cancer risks. Interestingly, a higher COVID-19 impact is associated with a decreased risk of thyroid cancer. In summary, our findings demonstrate a genetic correlation between COVID-19 and thyroid cancer, contributing to our understanding of the interplay between COVID-19 and cancer risk.
Collapse
Affiliation(s)
- Shuhong Li
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Zedong Du
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Hui Ma
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Liang Cai
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Xiao Liu
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, China
| | - Jie He
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Carstens M, Trujillo J, Dolmus Y, Rivera C, Calderwood S, Lejarza J, López C, Bertram K. Adipose-derived stromal vascular fraction cells to treat long-term pulmonary sequelae of coronavirus disease 2019: 12-month follow-up. Cytotherapy 2024; 26:1076-1083. [PMID: 38639670 DOI: 10.1016/j.jcyt.2024.03.491] [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: 02/06/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND AIMS Long coronavirus disease (COVID) is estimated to occur in up to 20% of patients with coronavirus disease 2019 (COVID-19) infections, with many having persistent pulmonary symptoms. Mesenchymal stromal cells (MSCs) have been shown to have powerful immunomodulatory and anti-fibrotic properties. Autologous adipose-derived (AD) stromal vascular fraction (SVF) contains MSC and other healing cell components and can be obtained by small-volume lipoaspiration and administered on the same day. This study was designed to study the safety of AD SVF infused intravenously to treat the pulmonary symptoms of long COVID. METHODS Five subjects with persistent cough and dyspnea after hospitalization and subsequent discharge for COVID-19 pneumonia were treated with 40 million intravenous autologous AD SVF cells and followed for 12 months, to include with pulmonary function tests and computed tomography scans of the lung. RESULTS SVF infusion was safe, with no significant adverse events related to the infusion out to 12 months. Four subjects had improvements in pulmonary symptoms, pulmonary function tests, and computed tomography scans, with some improvement noted as soon as 1 month after SVF treatment. CONCLUSIONS It is not possible to distinguish between naturally occurring improvement or improvement caused by SVF treatment in this small, uncontrolled study. However, the results support further study of autologous AD SVF as a treatment for long COVID.
Collapse
Affiliation(s)
- Michael Carstens
- Department of Surgery, Hospital Escuela Oscar Danilo Rosale Argüello, León, Nicaragua; Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina, USA.
| | - Jessy Trujillo
- Department of Medicine, Hospital Monte España, Managua, Nicaragua
| | - Yanury Dolmus
- Department of Pediatrics, Hospital Escuela Cesar Amador Molina, Matagalpa, Nicaragua
| | - Carlos Rivera
- Department of Radiology, Hospital Escuela Cesar Amador Molina, Matagalpa, Nicaragua
| | - Santos Calderwood
- Department of Surgery, Hospital Escuela Cesar Amador Molina, Matagalpa, Nicaragua
| | - Judith Lejarza
- Department of Surgery, Hospital Escuela Oscar Danilo Rosale Argüello, León, Nicaragua
| | - Carlos López
- Department of Medicine, Hospital Escuela Oscar Danilo Rosales Argüello, León, Nicaragua
| | - Kenneth Bertram
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
8
|
Meier A, Sakoulas G, Nizet V, Ulloa ER. Neutrophil Extracellular Traps: An Emerging Therapeutic Target to Improve Infectious Disease Outcomes. J Infect Dis 2024; 230:514-521. [PMID: 38728418 PMCID: PMC11326844 DOI: 10.1093/infdis/jiae252] [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: 01/29/2024] [Revised: 04/25/2024] [Accepted: 05/09/2024] [Indexed: 05/12/2024] Open
Abstract
Neutrophils possess a diverse repertoire of pathogen clearance mechanisms, one of which is the formation of neutrophil extracellular traps (NETs). NETs are complexes of histone proteins and DNA coated with proteolytic enzymes that are released extracellularly to entrap pathogens and aid in their clearance, in a process known as NETosis. Intravascular NETosis may drive a massive inflammatory response that has been shown to contribute to morbidity and mortality in many infectious diseases, including malaria, dengue fever, influenza, bacterial sepsis, and severe acute respiratory syndrome coronavirus 2 infection. In this review we seek to (1) summarize the current understanding of NETs, (2) discuss infectious diseases in which NET formation contributes to morbidity and mortality, and (3) explore potential adjunctive therapeutics that may be considered for future study in treating severe infections driven by NET pathophysiology. This includes drugs specifically targeting NET inhibition and US Food and Drug Administration-approved drugs that may be repurposed as NET inhibitors.
Collapse
Affiliation(s)
- Angela Meier
- Department of Anesthesiology, Division of Critical Care, University of California, San Diego School of Medicine, La Jolla
| | - George Sakoulas
- Division of Infectious Diseases, Sharp Rees-Stealy Medical Group, San Diego
- Collaborative to Halt Antibiotic-Resistant Microbes (CHARM)
| | - Victor Nizet
- Collaborative to Halt Antibiotic-Resistant Microbes (CHARM)
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California, San Diego School of Medicine, La Jolla
| | - Erlinda R Ulloa
- Department of Pediatrics, University of California, Irvine School of Medicine
- Division of Infectious Disease, Children's Hospital of Orange County, Orange, California
| |
Collapse
|
9
|
Rahman SM, Buchholz DW, Imbiakha B, Jager MC, Leach J, Osborn RM, Birmingham AO, Dewhurst S, Aguilar HC, Luebke AE. Migraine inhibitor olcegepant reduces weight loss and IL-6 release in SARS-CoV-2-infected older mice with neurological signs. J Virol 2024; 98:e0006624. [PMID: 38814068 PMCID: PMC11265435 DOI: 10.1128/jvi.00066-24] [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: 01/10/2024] [Accepted: 04/15/2024] [Indexed: 05/31/2024] Open
Abstract
COVID-19 can cause neurological symptoms such as fever, dizziness, and nausea. However, such neurological symptoms of SARS-CoV-2 infection have been hardly assessed in mouse models. In this study, we infected two commonly used wild-type mouse lines (C57BL/6J and 129/SvEv) and a 129S calcitonin gene-related peptide (αCGRP) null-line with mouse-adapted SARS-CoV-2 and demonstrated neurological signs including fever, dizziness, and nausea. We then evaluated whether a CGRP receptor antagonist, olcegepant, a "gepant" antagonist used in migraine treatment, could mitigate acute neuroinflammatory and neurological signs of SARS-COV-2 infection. First, we determined whether CGRP receptor antagonism provided protection from permanent weight loss in older (>18 m) C57BL/6J and 129/SvEv mice. We also observed acute fever, dizziness, and nausea in all older mice, regardless of treatment. In both wild-type mouse lines, CGRP antagonism reduced acute interleukin 6 (IL-6) levels with virtually no IL-6 release in mice lacking αCGRP. These findings suggest that migraine inhibitors such as those blocking CGRP receptor signaling protect against acute IL-6 release and subsequent inflammatory events after SARS-CoV-2 infection, which may have repercussions for related pandemic or endemic coronavirus outbreaks.IMPORTANCECoronavirus disease (COVID-19) can cause neurological symptoms such as fever, headache, dizziness, and nausea. However, such neurological symptoms of severe acute respiratory syndrome CoV-2 (SARS-CoV-2) infection have been hardly assessed in mouse models. In this study, we first infected two commonly used wild-type mouse lines (C57BL/6J and 129S) with mouse-adapted SARS-CoV-2 and demonstrated neurological symptoms including fever and nausea. Furthermore, we showed that the migraine treatment drug olcegepant could reduce long-term weight loss and IL-6 release associated with SARS-CoV-2 infection. These findings suggest that a migraine blocker can be protective for at least some acute SARS-CoV-2 infection signs and raise the possibility that it may also impact long-term outcomes.
Collapse
Affiliation(s)
- Shafaqat M. Rahman
- Departments of Biomedical Engineering, Neuroscience, Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - David W. Buchholz
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Brian Imbiakha
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Mason C. Jager
- Department of Population Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Justin Leach
- Departments of Biomedical Engineering, Neuroscience, Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Raven M. Osborn
- Departments of Biomedical Engineering, Neuroscience, Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Ann O. Birmingham
- Departments of Biomedical Engineering, Neuroscience, Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Stephen Dewhurst
- Departments of Biomedical Engineering, Neuroscience, Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Hector C. Aguilar
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Anne E. Luebke
- Departments of Biomedical Engineering, Neuroscience, Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
10
|
Mitri C, Philippart F, Sacco E, Legriel S, Rousselet N, Dupuis G, Colsch B, Corvol H, Touqui L, Tabary O. Multicentric investigations of the role in the disease severity of accelerated phospholipid changes in COVID-19 patient airway. Microbes Infect 2024; 26:105354. [PMID: 38754811 DOI: 10.1016/j.micinf.2024.105354] [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: 05/12/2023] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
CONTEXT The changes in host membrane phospholipids are crucial in airway infection pathogenesis. Phospholipase A2 hydrolyzes host cell membranes, producing lyso-phospholipids and free fatty acids, including arachidonic acid (AA), which contributes significantly to lung inflammation. AIM Follow these changes and their evolution from day 1, day 3 to day 7 in airway aspirates of 89 patients with COVID-19-associated acute respiratory distress syndrome and examine whether they correlate with the severity of the disease. The patients were recruited in three French intensive care units. The analysis was conducted from admission to the intensive care unit until the end of the first week of mechanical ventilation. RESULTS In the airway aspirates, we found significant increases in the levels of host cell phospholipids, including phosphatidyl-serine and phosphatidyl-ethanolamine, and their corresponding lyso-phospholipids. This was accompanied by increased levels of AA and its inflammatory metabolite prostaglandin E2 (PGE2). Additionally, enhanced levels of ceramides, sphingomyelin, and free cholesterol were observed in these aspirates. These lipids are known to be involved in cell death and/or apoptosis, whereas free cholesterol plays a role in virus entry and replication in host cells. However, there were no significant changes in the levels of dipalmitoyl-phosphatidylcholine, the major surfactant phospholipid. A correlation analysis revealed an association between mortality risk and levels of AA and PGE2, as well as host cell phospholipids. CONCLUSION Our findings indicate a correlation between heightened cellular phospholipid modifications and variations in AA and PGE2 with the severity of the disease in patients. Nevertheless, there is no indication of surfactant alteration in the initial phases of the illness.
Collapse
Affiliation(s)
- Christie Mitri
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France
| | - François Philippart
- Endotoxins, Structures and Host Response, Department of Microbiology, Institute for Integrative Biology of the Cell, UMR 9891 CNRS-CEA-Paris Saclay University, 98190 Gif-sur-Yvette, France; Medical-Surgical Intensive Care Unit, Groupe Hospitalier Paris Saint Joseph, Paris, France
| | - Emmanuelle Sacco
- Department of Clinical Research. Groupe Hospitalier Paris Saint Joseph, Paris, France
| | - Stéphane Legriel
- Medical-Surgical Intensive Care Unit, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Nathalie Rousselet
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France
| | - Gabrielle Dupuis
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France
| | - Benoît Colsch
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (MTS), MetaboHUB, F-91191, Gif sur Yvette, France
| | - Harriet Corvol
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France; Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Trousseau, Service de Pneumologie Pédiatrique, 75012, Paris, France
| | - Lhousseine Touqui
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France; Inserm, Institut Pasteur, Mucoviscidose et Bronchopathies Chroniques, Département Santé Globale, Paris, France.
| | - Olivier Tabary
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France.
| |
Collapse
|
11
|
Silva BRDS, Sidarta-Oliveira D, Morari J, Bombassaro B, Jara CP, Simeoni CL, Parise PL, Proenca-Modena JL, Velloso LA, Velander WH, Araújo EP. Protein C Pretreatment Protects Endothelial Cells from SARS-CoV-2-Induced Activation. Viruses 2024; 16:1049. [PMID: 39066212 PMCID: PMC11281670 DOI: 10.3390/v16071049] [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: 05/30/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
SARS-CoV-2 can induce vascular dysfunction and thrombotic events in patients with severe COVID-19; however, the cellular and molecular mechanisms behind these effects remain largely unknown. In this study, we used a combination of experimental and in silico approaches to investigate the role of PC in vascular and thrombotic events in COVID-19. Single-cell RNA-sequencing data from patients with COVID-19 and healthy subjects were obtained from the publicly available Gene Expression Omnibus (GEO) repository. In addition, HUVECs were treated with inactive protein C before exposure to SARS-CoV-2 infection or a severe COVID-19 serum. An RT-qPCR array containing 84 related genes was used, and the candidate genes obtained were evaluated. Activated protein C levels were measured using an ELISA kit. We identified at the single-cell level the expression of several pro-inflammatory and pro-coagulation genes in endothelial cells from the patients with COVID-19. Furthermore, we demonstrated that exposure to SARS-CoV-2 promoted transcriptional changes in HUVECs that were partly reversed by the activated protein C pretreatment. We also observed that the serum of severe COVID-19 had a significant amount of activated protein C that could protect endothelial cells from serum-induced activation. In conclusion, activated protein C protects endothelial cells from pro-inflammatory and pro-coagulant effects during exposure to the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Bruna Rafaela dos Santos Silva
- School of Nursing, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Center (OCRC), Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| | - Davi Sidarta-Oliveira
- Laboratory of Cell Signalling, Obesity and Comorbidities Center (OCRC), Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| | - Joseane Morari
- Laboratory of Cell Signalling, Obesity and Comorbidities Center (OCRC), Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| | - Bruna Bombassaro
- School of Nursing, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
- Laboratory of Cell Signalling, Obesity and Comorbidities Center (OCRC), Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| | | | - Camila Lopes Simeoni
- Laboratory of Emerging Viruses, Institute of Biology, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil; (C.L.S.); (P.L.P.); (J.L.P.-M.)
| | - Pierina Lorencini Parise
- Laboratory of Emerging Viruses, Institute of Biology, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil; (C.L.S.); (P.L.P.); (J.L.P.-M.)
| | - José Luiz Proenca-Modena
- Laboratory of Emerging Viruses, Institute of Biology, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil; (C.L.S.); (P.L.P.); (J.L.P.-M.)
| | - Licio A. Velloso
- Laboratory of Cell Signalling, Obesity and Comorbidities Center (OCRC), Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| | - William H. Velander
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, NE 68588, USA;
| | - Eliana P. Araújo
- School of Nursing, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
- School of Medical Sciences, Universidade Estadual de Campinas, Campinas 13083-970, SP, Brazil
| |
Collapse
|
12
|
Ranjbar M, Cusack RP, Whetstone CE, Brister DL, Wattie J, Wiltshire L, Alsaji N, Le Roux J, Cheng E, Srinathan T, Ho T, Sehmi R, O’Byrne PM, Snow-Smith M, Makiya M, Klion AD, Duong M, Gauvreau GM. Immune Response Dynamics and Biomarkers in COVID-19 Patients. Int J Mol Sci 2024; 25:6427. [PMID: 38928133 PMCID: PMC11204302 DOI: 10.3390/ijms25126427] [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: 05/15/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND The immune response dynamics in COVID-19 patients remain a subject of intense investigation due to their implications for disease severity and treatment outcomes. We examined changes in leukocyte levels, eosinophil activity, and cytokine profiles in patients hospitalized with COVID-19. METHODS Serum samples were collected within the first 10 days of hospitalization/confirmed infection and analyzed for eosinophil granule proteins (EGP) and cytokines. Information from medical records including comorbidities, clinical symptoms, medications, and complete blood counts were collected at the time of admission, during hospitalization and at follow up approximately 3 months later. RESULTS Serum levels of eotaxin, type 1 and type 2 cytokines, and alarmin cytokines were elevated in COVID-19 patients, highlighting the heightened immune response (p < 0.05). However, COVID-19 patients exhibited lower levels of eosinophils and eosinophil degranulation products compared to hospitalized controls (p < 0.05). Leukocyte counts increased consistently from admission to follow-up, indicative of recovery. CONCLUSION Attenuated eosinophil activity alongside elevated chemokine and cytokine levels during active infection, highlights the complex interplay of immune mediators in the pathogenesis COVID-19 and underscores the need for further investigation into immune biomarkers and treatment strategies.
Collapse
Affiliation(s)
- Maral Ranjbar
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | - Ruth P. Cusack
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | - Christiane E. Whetstone
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | - Danica L. Brister
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | - Jennifer Wattie
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | - Lesley Wiltshire
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | - Nadia Alsaji
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| | | | - Eric Cheng
- St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada; (E.C.); (T.S.)
| | - Thivya Srinathan
- St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada; (E.C.); (T.S.)
| | - Terence Ho
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
- The Research Institute of St. Joe’s Hamilton, Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada
| | - Roma Sehmi
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
- The Research Institute of St. Joe’s Hamilton, Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada
| | - Paul M. O’Byrne
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
- Hamilton Health Sciences, Hamilton, ON L8N 3Z5, Canada;
- St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada; (E.C.); (T.S.)
| | - Maryonne Snow-Smith
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (M.S.-S.); (M.M.); (A.D.K.)
| | - Michelle Makiya
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (M.S.-S.); (M.M.); (A.D.K.)
| | - Amy D. Klion
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (M.S.-S.); (M.M.); (A.D.K.)
| | - MyLinh Duong
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
- The Research Institute of St. Joe’s Hamilton, Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada
- Population Health Research Institute, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Gail M. Gauvreau
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (M.R.); (R.P.C.); (C.E.W.); (D.L.B.); (J.W.); (L.W.); (N.A.); (T.H.); (R.S.); (P.M.O.); (M.D.)
| |
Collapse
|
13
|
Rai P, Marano JM, Kang L, Coutermarsh-Ott S, Daamen AR, Lipsky PE, Weger-Lucarelli J. Obesity fosters severe disease outcomes in a mouse model of coronavirus infection associated with transcriptomic abnormalities. J Med Virol 2024; 96:e29587. [PMID: 38587204 DOI: 10.1002/jmv.29587] [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: 02/01/2024] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024]
Abstract
Obesity has been identified as an independent risk factor for severe outcomes in humans with coronavirus disease 2019 (COVID-19) and other infectious diseases. Here, we established a mouse model of COVID-19 using the murine betacoronavirus, mouse hepatitis virus 1 (MHV-1). C57BL/6 and C3H/HeJ mice exposed to MHV-1 developed mild and severe disease, respectively. Obese C57BL/6 mice developed clinical manifestations similar to those of lean controls. In contrast, all obese C3H/HeJ mice succumbed by 8 days postinfection, compared to a 50% mortality rate in lean controls. Notably, both lean and obese C3H/HeJ mice exposed to MHV-1 developed lung lesions consistent with severe human COVID-19, with marked evidence of diffuse alveolar damage (DAD). To identify early predictive biomarkers of worsened disease outcomes in obese C3H/HeJ mice, we sequenced RNA from whole blood 2 days postinfection and assessed changes in gene and pathway expression. Many pathways uniquely altered in obese C3H/HeJ mice postinfection aligned with those found in humans with severe COVID-19. Furthermore, we observed altered gene expression related to the unfolded protein response and lipid metabolism in infected obese mice compared to their lean counterparts, suggesting a role in the severity of disease outcomes. This study presents a novel model for studying COVID-19 and elucidating the mechanisms underlying severe disease outcomes in obese and other hosts.
Collapse
Affiliation(s)
- Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, USA
| | - Jeffrey M Marano
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, USA
- Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Blacksburg, Virginia, USA
| | - Lin Kang
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
- Biomedical Affairs and Research, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, USA
- College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Sheryl Coutermarsh-Ott
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
| | | | | | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
14
|
Harris A, Creecy A, Awosanya OD, McCune T, Ozanne MV, Toepp AJ, Kacena MA, Qiao X. SARS-CoV-2 and its Multifaceted Impact on Bone Health: Mechanisms and Clinical Evidence. Curr Osteoporos Rep 2024; 22:135-145. [PMID: 38236510 PMCID: PMC10912131 DOI: 10.1007/s11914-023-00843-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/15/2023] [Indexed: 01/19/2024]
Abstract
PURPOSE OF REVIEW SARS-CoV-2 infection, the culprit of the COVID-19 pandemic, has been associated with significant long-term effects on various organ systems, including bone health. This review explores the current understanding of the impacts of SARS-CoV-2 infection on bone health and its potential long-term consequences. RECENT FINDINGS As part of the post-acute sequelae of SARS-CoV-2 infection, bone health changes are affected by COVID-19 both directly and indirectly, with multiple potential mechanisms and risk factors involved. In vitro and preclinical studies suggest that SARS-CoV-2 may directly infect bone marrow cells, leading to alterations in bone structure and osteoclast numbers. The virus can also trigger a robust inflammatory response, often referred to as a "cytokine storm", which can stimulate osteoclast activity and contribute to bone loss. Clinical evidence suggests that SARS-CoV-2 may lead to hypocalcemia, altered bone turnover markers, and a high prevalence of vertebral fractures. Furthermore, disease severity has been correlated with a decrease in bone mineral density. Indirect effects of SARS-CoV-2 on bone health, mediated through muscle weakness, mechanical unloading, nutritional deficiencies, and corticosteroid use, also contribute to the long-term consequences. The interplay of concurrent conditions such as diabetes, obesity, and kidney dysfunction with SARS-CoV-2 infection further complicates the disease's impact on bone health. SARS-CoV-2 infection directly and indirectly affects bone health, leading to potential long-term consequences. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.
Collapse
Affiliation(s)
- Alexander Harris
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Amy Creecy
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Olatundun D Awosanya
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Thomas McCune
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
- Division of Nephrology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Marie V Ozanne
- Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA, USA
| | - Angela J Toepp
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
- Enterprise Analytics, Sentara Health, Virginia Beach, VA, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
| | - Xian Qiao
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA.
- SMG Pulmonary, Critical Care, and Sleep Specialists, Norfolk, VA, USA.
- Division of Pulmonary and Critical Care Medicine, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
15
|
Creecy A, Awosanya OD, Harris A, Qiao X, Ozanne M, Toepp AJ, Kacena MA, McCune T. COVID-19 and Bone Loss: A Review of Risk Factors, Mechanisms, and Future Directions. Curr Osteoporos Rep 2024; 22:122-134. [PMID: 38221578 PMCID: PMC10912142 DOI: 10.1007/s11914-023-00842-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/15/2023] [Indexed: 01/16/2024]
Abstract
PURPOSE OF REVIEW SARS-CoV-2 drove the catastrophic global phenomenon of the COVID-19 pandemic resulting in a multitude of systemic health issues, including bone loss. The purpose of this review is to summarize recent findings related to bone loss and potential mechanisms. RECENT FINDINGS The early clinical evidence indicates an increase in vertebral fractures, hypocalcemia, vitamin D deficiencies, and a loss in BMD among COVID-19 patients. Additionally, lower BMD is associated with more severe SARS-CoV-2 infection. Preclinical models have shown bone loss and increased osteoclastogenesis. The bone loss associated with SARS-CoV-2 infection could be the result of many factors that directly affect the bone such as higher inflammation, activation of the NLRP3 inflammasome, recruitment of Th17 cells, the hypoxic environment, and changes in RANKL/OPG signaling. Additionally, SARS-CoV-2 infection can exert indirect effects on the skeleton, as mechanical unloading may occur with severe disease (e.g., bed rest) or with BMI loss and muscle wasting that has also been shown to occur with SARS-CoV-2 infection. Muscle wasting can also cause systemic issues that may influence the bone. Medications used to treat SARS-CoV-2 infection also have a negative effect on the bone. Lastly, SARS-CoV-2 infection may also worsen conditions such as diabetes and negatively affect kidney function, all of which could contribute to bone loss and increased fracture risk. SARS-CoV-2 can negatively affect the bone through multiple direct and indirect mechanisms. Future work will be needed to determine what patient populations are at risk of COVID-19-related increases in fracture risk, the mechanisms behind bone loss, and therapeutic options. This review article is part of a series of multiple manuscripts designed to determine the utility of using artificial intelligence for writing scientific reviews.
Collapse
Affiliation(s)
- Amy Creecy
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Olatundun D Awosanya
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexander Harris
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xian Qiao
- Critical Care, and Sleep Specialists, SMG Pulmonary, Norfolk, VA, USA
- Division of Pulmonary and Critical Care Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Marie Ozanne
- Department of Mathematics and Statistics, Mount Holyoke College, South Hadley, MA, USA
| | - Angela J Toepp
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
- Enterprise Analytics, Sentara Health, Virginia Beach, VA, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
| | - Thomas McCune
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA.
- Division of Nephrology, Eastern Virginia Medical School, Norfolk, VA, USA.
| |
Collapse
|
16
|
Ghosh A, Sanyal A, Mitra P, Dey T, Acharjee A, Pattnaik R, Nesa L. Transmission mechanism and clinical manifestations of SARS-CoV-2. DIAGNOSIS AND ANALYSIS OF COVID-19 USING ARTIFICIAL INTELLIGENCE AND MACHINE LEARNING-BASED TECHNIQUES 2024:65-96. [DOI: 10.1016/b978-0-323-95374-0.00006-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
17
|
Abdel-Salam Elgohary M, Ali A, J Alarfaj S, Shahin H, Ibrahim Zaki A, Medhat Hasan E, Emam Mohamed M, Mahmoud Elkholy A, El-Masry TA, Samir Kamal J, Ali AbdelRahim M, Wageh Saber A, Seadawy MG, Elshishtawy MHM, El-Bouseary MM. Even one dose of tocilizumab could hinder bad prognosis of cytokines storm in COVID-19 patients. Cytokine 2024; 173:156433. [PMID: 37972479 DOI: 10.1016/j.cyto.2023.156433] [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: 02/08/2023] [Revised: 08/04/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Severe COVID-19 pneumonia is a principal cause of death due to cascade of hyper inflammatory condition that leading to lung damage. Therefore, an effective therapy to countercurrent the surge of uncontrolled inflammation is mandatory to propose. Anti-interlukin-6 receptor antagonist monoclonal therapy, tocilizumab (TCZ) showed potential results in COVID-19 patients. This study aimed to emphasize the factors associated with mortality in COVID-19 patients that treated with tocilizumab and may influence the level of serum IL-6. A retrospective cohort study included all patients with clinical parameters that pointed to presence of cytokines storm and treated with one or more doses of TCZ beside the regular protocol of COVID-19 pneumonia. The factors that influence the mortality in addition to the level of serum IL-6 were analyzed. A total of 377 patients were included, 69.5 % of them received only one dose of TCZ which started mainly at the third day of admission. The mortality rate was 29.44 %. Regardless the time of starting TCZ, just one dose was fair enough to prevent bad consequence; OR = 0.04, P = 0.001.However, in spite of protective action of TCZ, older age and female sex were significant risk factors for mortality, P = 0.001 and 0.01 respectively, as well heart disease. Moreover, increasing the level of neutrophil, AST and IL-6 were associated with bad prognosis. In the same line, treatment with ivermectin, chloroquine and remdesivir inversely affect the level of IL-6. Early treatments of COVID-19 pneumonia with at least one dose of tocilizumab minimized the fatality rate.
Collapse
Affiliation(s)
| | - Asmaa Ali
- Department of Pulmonary Medicine, Abbassia Chest Hospital, MOH, Cairo, Egypt; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, PR China.
| | - Sumaiah J Alarfaj
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University. P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Hesham Shahin
- Chest Consultant, Manager of Military Chest Hospital, Cairo, Egypt.
| | | | | | | | | | - Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Egypt.
| | | | | | - Ashgan Wageh Saber
- Chemistry Laboratory Department, Central Military Laboratories, Cairo, Egypt.
| | - Mohamed G Seadawy
- Biological Prevention Department, Ministry of Defense, Cairo, Egypt.
| | | | - Maisra M El-Bouseary
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| |
Collapse
|
18
|
de Maistre E, Savard P, Guinot PG. COVID-19 and the Concept of Thrombo-Inflammation: Review of the Relationship between Immune Response, Endothelium and Coagulation. J Clin Med 2023; 12:7245. [PMID: 38068297 PMCID: PMC10706970 DOI: 10.3390/jcm12237245] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 09/11/2024] Open
Abstract
COVID-19, caused by the SARS-CoV-2 virus, has revealed a complex interplay between inflammation and coagulation, leading to the emergence of the concept of thrombo-inflammation. This concept recognizes that COVID-19 is not solely a respiratory illness, but a systemic disease with significant vascular and hematological components. COVID-19 is associated with an unusual prothrombotic state, with intense endothelial activation leading to vasculopathy, cytokine storm, complement system activation and a hypercoagulability state (the activation of platelets and the coagulation cascade, impaired fibrinolysis). The aim of this review is to discuss the different pathological pathways described in COVID-19 that lead to thromboembolic events. Widespread vaccination and post-COVID-19 immunization allows control over the severity of this pandemic. A better understanding of the pathophysiology of COVID-19 can improve the management of frail patients who are hospitalized in intensive care units.
Collapse
Affiliation(s)
| | - Philippe Savard
- Haemostais Unit, Dijon University Hospital, F-21000 Dijon, France;
| | - Pierre-Gregoire Guinot
- Department of Anesthesiology and Intensive Care, Dijon University Hospital, F-21000 Dijon, France;
| |
Collapse
|
19
|
Wakfie-Corieh CG, Ferrando-Castagnetto F, García-Esquinas M, Cabrera-Martín MN, Rodríguez Rey C, Ortega Candil A, Couto Caro RM, Carreras Delgado JL. Metabolic characterization of structural lung changes in patients with findings suggestive of incidental COVID-19 pneumonia on 18F-FDG PET/CT. Pathophysiological insights from multimodal images obtained during the pandemic. Rev Esp Med Nucl Imagen Mol 2023; 42:380-387. [PMID: 37454730 DOI: 10.1016/j.remnie.2023.07.004] [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: 03/23/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE To evaluate the metabolic uptake of different tomographic signs observed in patients with incidental structural findings suggestive of COVID-19 pneumonia through 18F-FDG PET/CT. MATERIALS AND METHODS We retrospectively analyzed 596 PET/CT studies performed from February 21, 2020 to April 17, 2020. After excluding 37 scans (non-18F-FDG PET tracers and brain studies), we analyzed the metabolic activity of several structural changes integrated in the CO-RADS score using the SUVmax of multimodal studies with 18F-FDG. RESULTS Forty-three patients with 18F-FDG PET/CT findings suggestive of COVID-19 pneumonia were included (mean age: 68±12.3 years, 22 male). SUVmax values were higher in patients with CO-RADS categories 5-6 than in those with lower CO-RADS categories (6.1±3.0 vs. 3.6±2.1, p=0.004). In patients with CO-RADS 5-6, ground-glass opacities, bilaterality and consolidations exhibited higher SUVmax values (p-values of 0.01, 0.02 and 0.01, respectively). Patchy distribution and crazy paving pattern were also associated with higher SUVmax (p-values of 0.002 and 0.01). After multivariate analysis, SUVmax was significantly associated with a positive structural diagnosis of COVID-19 pneumonia (odds ratio=0.63, 95% confidence interval=0.41-0.90; p=0.02). The ROC curve of the regression model intended to confirm or rule out the structural diagnosis of COVID-19 pneumonia showed an AUC of 0.77 (standard error=0.072, p=0.003). CONCLUSIONS In those patients referred for standard oncologic and non-oncologic indications (43/559; 7.7%) during pandemic, imaging with 18F-FDG PET/CT is a useful tool during incidental detection of COVID-19 pneumonia. Several CT findings characteristic of COVID-19 pneumonia, specifically those included in diagnostic CO-RADS scores (5-6), were associated with higher SUVmax values.
Collapse
Affiliation(s)
- C G Wakfie-Corieh
- Department of Nuclear Medicine, Hospital Clínico San Carlos, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain.
| | - F Ferrando-Castagnetto
- Department of Cardiology, Cardiovascular University Center, Hospital de Clínicas Dr. Manuel Quintela, Montevideo, Uruguay
| | - M García-Esquinas
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain; Department of Radiology, Hospital Clínico San Carlos, Madrid, Spain
| | - M N Cabrera-Martín
- Department of Nuclear Medicine, Hospital Clínico San Carlos, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - C Rodríguez Rey
- Department of Nuclear Medicine, Hospital Clínico San Carlos, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - A Ortega Candil
- Department of Nuclear Medicine, Hospital Clínico San Carlos, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - R M Couto Caro
- Department of Nuclear Medicine, Hospital Clínico San Carlos, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - J L Carreras Delgado
- Department of Nuclear Medicine, Hospital Clínico San Carlos, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
20
|
Alba JR, Zapater E, Martin C, Ocete D, Gonzalez-Cruz A, Angel-de-Miguel A, Ferrer C, Oishi N. Mapping of SARS-CoV-2 in Waldeyer's lymphatic ring and visceral biopsies: the age and the illness duration's impact. Braz J Otorhinolaryngol 2023; 89:101317. [PMID: 37696117 PMCID: PMC10498168 DOI: 10.1016/j.bjorl.2023.101317] [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: 05/20/2023] [Accepted: 08/27/2023] [Indexed: 09/13/2023] Open
Abstract
OBJECTIVE To study the impact of age and the interval between disease diagnosis and death on the organotropism of SARS-CoV-2. METHOD Patients underwent post-mortem biopsies from lungs, Waldeyer ring, heart, liver, kidneys and bone marrow between 2020‒2021. SARS-CoV-2 organotropism was mapped by using molecular RT-PCR analyses for SARS-CoV2 targeting the Envelope gene (E), the RNA Polymerase Gene (RdRp), and the Nucleocapsid gene (N). Statistical and linear regression analysis was performed to study the impact of age and illness duration in SARS-CoV-2 organotropism. RESULTS We performed 158 postmortem biopsies in 21 patients, with a mean age of 76 years old. The mean interval between the diagnosis of the infection to the death was 23 days. The RNA of the SARS-CoV-2 was detected in 100% of lung biopsies, 76%‒82% of Waldeyer's ring biopsies, 55% of heart biopsies, 40% of kidney biopsies, 33% of liver and 25% of bone marrow biopsies. Patients who died before the day 9, presented extensive visceral dissemination of SARS-CoV-2 RNA. Most of the patients older than 80 years (90%) presented visceral dissemination of SARS-CoV-2 RNA, while among younger patients, only 3/11 patients presented visceral dissemination of the virus. The relationship between "age" and "illness duration" and multitropism of the virus was statistically significant (p<0.001). CONCLUSION Disease interval and age were factors that were significantly associated with RT-PCR positive results in multiple organs. Critical COVID-19 patients have multiorganic viral dissemination in early stages. In the critical older patients, multiorganic viral dissemination is the rule. LEVEL OF EVIDENCE: 4 CASE SERIES
Collapse
Affiliation(s)
- Jose Ramón Alba
- University of Valencia, Faculty of Medicine and Dentistry, Valencia, Spain
| | - Enrique Zapater
- University of Valencia, Faculty of Medicine and Dentistry, Valencia, Spain.
| | - Cristina Martin
- León University Hospital, ENT Department, Ponferrada, Castilla y Leon, Spain
| | - Dolores Ocete
- Valencia University General Hospital, Microbiology Department, Valencia, Spain
| | | | | | - Carolina Ferrer
- Valencia University General Hospital, Anesthesia Department, Valencia, Spain
| | - Natsuki Oishi
- Valencia University General Hospital, ENT Department, Valencia, Spain
| |
Collapse
|
21
|
Abdullah M, Ali A, Usman M, Naz A, Qureshi JA, Bajaber MA, Zhang X. Post COVID-19 complications and follow up biomarkers. NANOSCALE ADVANCES 2023; 5:5705-5716. [PMID: 37881715 PMCID: PMC10597564 DOI: 10.1039/d3na00342f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/11/2023] [Indexed: 10/27/2023]
Abstract
Millions of people were infected by the coronavirus disease (COVID-19) epidemic, which left a huge burden on the care of post COVID-19 survivors around the globe. The self-reported COVID-19 symptoms were experienced by an estimated 1.3 million people in the United Kingdom (2% of the population), and these symptoms persisted for about 4 weeks from the beginning of the infection. The symptoms most frequently reported were exhaustion, shortness of breath, muscular discomfort, joint pain, headache, cough, chest pain, cognitive impairment, memory loss, anxiety, sleep difficulties, diarrhea, and a decreased sense of smell and taste in post-COVID-19 affected people. The post COVID-19 complications were frequently related to the respiratory, cardiac, nervous, psychological and musculoskeletal systems. The lungs, liver, kidneys, heart, brain and other organs had been impaired by hypoxia and inflammation in post COVID-19 individuals. The upregulation of substance "P" (SP) and various cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin 6 (IL-6), interleukin 10 (IL-10), interleukin 1 beta (IL-1β), angiotensin-converting enzyme 2 (ACE2) and chemokine C-C motif ligand 3 (CCL3) has muddled respiratory, cardiac, neuropsychiatric, dermatological, endocrine, musculoskeletal, gastrointestinal, renal and genitourinary complications in post COVID-19 people. To prevent these complications from worsening, it was therefore important to study how these biomarkers were upregulated and block their receptors.
Collapse
Affiliation(s)
- Muhammad Abdullah
- Institute of Molecular Biology and Biotechnology, University of Lahore Pakistan
| | - Amjed Ali
- University Institute of Physical Therapy, University of Lahore Pakistan
| | - Muhammad Usman
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University Xuzhou China
| | - Anam Naz
- Institute of Molecular Biology and Biotechnology, University of Lahore Pakistan
| | - Javed Anver Qureshi
- Institute of Molecular Biology and Biotechnology, University of Lahore Pakistan
| | - Majed A Bajaber
- Department of Chemistry, Faculty of Science, King Khalid University P.O. Box 9004 Abha 61413 Saudi Arabia
| | - Xiao Zhang
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University Xuzhou China
| |
Collapse
|
22
|
Mikhaleva L, Gioeva Z, Varyasin V, Berezhnaja E, Vandysheva R, Gutyrchik N, Pechnikova V, Kontorshchikov A, Midiber K, Kakturskij L. Pathomorphological Features of the Novel Coronavirus Disease in Patients with Systemic Amyloidosis. Biomedicines 2023; 11:2811. [PMID: 37893183 PMCID: PMC10604009 DOI: 10.3390/biomedicines11102811] [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: 09/12/2023] [Revised: 10/10/2023] [Accepted: 10/14/2023] [Indexed: 10/29/2023] Open
Abstract
Amyloidosis is one of the rare systemic illnesses characterized by the deposition of amyloid fibrils in various organs and tissues. There is a common point between COVID-19 and systemic amyloidosis regarding the multiorgan involvement in the pathological process which leads to a heightened risk for severe morbidity and mortality in amyloidosis patients who contracted COVID-19. We performed a pathomorphological analysis of the autopsy records of 22 patients who had COVID-19 and pre-existing systemic amyloidosis. The premortem diagnosis of systemic amyloidosis was established in 55% of patients, and in other 45% of cases, amyloidosis was found at autopsy. Based on the results of immunohistochemical amyloid typing, amyloid A (AA) amyloidosis was detected in 23%, amyloid light chain (AL) lambda in 32%, AL kappa-in 9%, and transthyretin (ATTR) amyloidosis-in 36% of observations. Immunohistochemical staining with an antibody against SARS-CoV-2 Spike (S) protein revealed positive immune reactions in type II alveolocytes in 59% of deceased persons. The analysis of autopsy findings indicates that patients with systemic amyloidosis are more likely to experience an aggressive clinical course of COVID-19 which leads to a multiorgan failure and a higher risk of fatal outcome.
Collapse
Affiliation(s)
- Liudmila Mikhaleva
- Avtsyn Research Institute of Human Morphology, Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", 117418 Moscow, Russia
| | - Zarina Gioeva
- Avtsyn Research Institute of Human Morphology, Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", 117418 Moscow, Russia
| | | | | | - Rositsa Vandysheva
- Avtsyn Research Institute of Human Morphology, Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", 117418 Moscow, Russia
| | - Nikita Gutyrchik
- Avtsyn Research Institute of Human Morphology, Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", 117418 Moscow, Russia
- Medical Institute, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Valentina Pechnikova
- Avtsyn Research Institute of Human Morphology, Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", 117418 Moscow, Russia
| | - Andrej Kontorshchikov
- Avtsyn Research Institute of Human Morphology, Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", 117418 Moscow, Russia
| | - Konstantin Midiber
- Avtsyn Research Institute of Human Morphology, Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", 117418 Moscow, Russia
| | - Lev Kakturskij
- Avtsyn Research Institute of Human Morphology, Federal State Budgetary Scientific Institution "Petrovsky National Research Centre of Surgery", 117418 Moscow, Russia
| |
Collapse
|
23
|
Lisman D, Zielińska G, Drath J, Łaszczewska A, Savochka I, Parafiniuk M, Ossowski A. Molecular Diagnosis of COVID-19 Sudden and Unexplained Deaths: The Insidious Face of the Pandemic. Diagnostics (Basel) 2023; 13:2980. [PMID: 37761347 PMCID: PMC10529476 DOI: 10.3390/diagnostics13182980] [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: 08/24/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
The COVID-19 epidemic has led to a significant increase in the number of deaths. This has resulted in forensic autopsies focusing on additional diagnostic possibilities. The following article is a summary of 23 autopsies of sudden and unexplained deaths. Particularly noteworthy are the described cases of children whose deaths were originally classified as SIDS (sudden infant death syndrome). All tests were performed at the Department of Forensic Medicine and Forensic Genetics, Pomeranian Medical University in Szczecin. Autopsy analyses were extended to include diagnostics of the SARS-CoV-2 virus using molecular methods and a detailed histopathological analysis of lung tissue. The material for molecular tests consisted of a nasopharyngeal swab taken postmortem and a lung tissue homogenate. In both cases, the RT-PCR method with CT cut-off point analysis was used for diagnosis. In all analyzed cases, the lungs showed massive congestion and increased fragility and cohesion. The tested material showed the presence of the SARS-CoV-2 virus, which indicated various stages of infection. It was observed that the higher the virus expression in the lungs, the lower or undetectable it was in the nasopharyngeal swab. This may explain false negative results during life in swabs. An interesting finding is that child deaths classified as SIDS also showed the presence of the virus. This may constitute a new direction of research.
Collapse
Affiliation(s)
- Dagmara Lisman
- Forensic Genetic Department, Pomeranian Medical University, 70-204 Szczecin, Poland; (G.Z.); (J.D.); (A.Ł.); (A.O.)
| | - Grażyna Zielińska
- Forensic Genetic Department, Pomeranian Medical University, 70-204 Szczecin, Poland; (G.Z.); (J.D.); (A.Ł.); (A.O.)
| | - Joanna Drath
- Forensic Genetic Department, Pomeranian Medical University, 70-204 Szczecin, Poland; (G.Z.); (J.D.); (A.Ł.); (A.O.)
| | - Aleksandra Łaszczewska
- Forensic Genetic Department, Pomeranian Medical University, 70-204 Szczecin, Poland; (G.Z.); (J.D.); (A.Ł.); (A.O.)
| | - Ilona Savochka
- Forensic Medicine Department, Pomeranian Medical University, 70-204 Szczecin, Poland; (I.S.); (M.P.)
| | - Mirosław Parafiniuk
- Forensic Medicine Department, Pomeranian Medical University, 70-204 Szczecin, Poland; (I.S.); (M.P.)
| | - Andrzej Ossowski
- Forensic Genetic Department, Pomeranian Medical University, 70-204 Szczecin, Poland; (G.Z.); (J.D.); (A.Ł.); (A.O.)
| |
Collapse
|
24
|
Bhargavan B, Kanmogne GD. SARS-CoV-2 Spike Proteins and Cell-Cell Communication Induce P-Selectin and Markers of Endothelial Injury, NETosis, and Inflammation in Human Lung Microvascular Endothelial Cells and Neutrophils: Implications for the Pathogenesis of COVID-19 Coagulopathy. Int J Mol Sci 2023; 24:12585. [PMID: 37628764 PMCID: PMC10454213 DOI: 10.3390/ijms241612585] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
COVID-19 progression often involves severe lung injury, inflammation, coagulopathy, and leukocyte infiltration into pulmonary tissues. The pathogenesis of these complications is unknown. Because vascular endothelium and neutrophils express angiotensin-converting enzyme-2 and spike (S)-proteins, which are present in bodily fluids and tissues of SARS-CoV-2-infected patients, we investigated the effect of S-proteins and cell-cell communication on human lung microvascular endothelial cells and neutrophils expression of P-selectin, markers of coagulopathy, NETosis, and inflammation. Exposure of endothelial cells or neutrophils to S-proteins and endothelial-neutrophils co-culture induced P-selectin transcription and expression, significantly increased expression/secretion of IL-6, von Willebrand factor (vWF, pro-coagulant), and citrullinated histone H3 (cit-H3, NETosis marker). Compared to the SARS-CoV-2 Wuhan variant, Delta variant S-proteins induced 1.4-15-fold higher P-selectin and higher IL-6 and vWF. Recombinant tissue factor pathway inhibitor (rTFPI), 5,5'-dithio-bis-(2-nitrobenzoic acid) (thiol blocker), and thrombomodulin (anticoagulant) blocked S-protein-induced vWF, IL-6, and cit-H3. This suggests that following SARS-CoV-2 contact with the pulmonary endothelium or neutrophils and endothelial-neutrophil interactions, S-proteins increase adhesion molecules, induce endothelial injury, inflammation, NETosis and coagulopathy via the tissue factor pathway, mechanisms involving functional thiol groups, and/or the fibrinolysis system. Using rTFPI, effectors of the fibrinolysis system and/or thiol-based drugs could be viable therapeutic strategies against SARS-CoV-2-induced endothelial injury, inflammation, NETosis, and coagulopathy.
Collapse
Affiliation(s)
| | - Georgette D. Kanmogne
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA;
| |
Collapse
|
25
|
Zhang W, Zhao M, Pu Z, Yin Q, Shui Y. Chicoric Acid Presented NLRP3-Mediated Pyroptosis through Mitochondrial Damage by PDPK1 Ubiquitination in an Acute Lung Injury Model. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2023; 51:1431-1457. [PMID: 37530505 DOI: 10.1142/s0192415x23500659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Chicoric acid (CA), a functional food ingredient, is a caffeic acid derivative that is mainly found in lettuce, pulsatilla, and other natural plants. However, the anti-inflammatory effects of CA in acute lung injury (ALI) remain poorly understood. This study was conducted to investigate potential drug usage of CA for ALI and the underlying molecular mechanisms of inflammation. C57BL/6 mice were given injections of liposaccharide (LPS) to establish the in vivo model. Meanwhile, BMDM cells were stimulated with LPS+ATP to build the in vitro model. CA significantly alleviated inflammation and oxidative stress in both the in vivo and in vitro models of ALI through the inhibition of NLR family pyrin domain-containing 3 (NLRP3)-mediated pyroptosis. In addition, CA attenuated mitochondrial damage to suppress NLRP3-mediated pyroptosis in the in vivo and in vitro models of ALI by suppressing the production of reactive oxygen species (ROS) via inhibiting the Akt/nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. CA inhibited the interaction between Akt at T308 and phosphoinositide-dependent kinase-1 (PDPK1) at S549, thus promoting the phosphorylation of the Akt protein. Furthermore, CA directly targeted the PDPK1 protein and accelerated PDPK1 ubiquitination, indicating that 91-GLY, 111-LYS, 126-TYR, 162-ALA, 205-ASP, and 223-ASP might be responsible for the interaction between PDPK1 and CA. In conclusion, CA from Lettuce alleviated NLRP3-mediated pyroptosis in the ALI model through ROS-induced mitochondrial damage by activating Akt/Nrf2 pathway via PDPK1 ubiquitination. The present study suggests that CA might be a potential therapeutic drug to treat or prevent ALI in pneumonia or COVID-19.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Pharmacy, Second Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, P. R. China
- Graduate School, Wannan Medical College, Wuhu 241001, Anhui, P. R. China
| | - Min Zhao
- Department of Pharmacy, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, Fujian, P. R. China
| | - Zhichen Pu
- Drug Clinical Evaluation, Yijishan Hospital of Wannan Medical College, Wuhu 241001, Anhui, P. R. China
| | - Qin Yin
- Department of Pharmacy, Second Affiliated Hospital of Wannan Medical College, Wuhu 241001, Anhui, P. R. China
- Wannan Medical College, Wuhu 241001, Anhui, P. R. China
| | - Yinping Shui
- Graduate School, Wannan Medical College, Wuhu 241001, Anhui, P. R. China
| |
Collapse
|
26
|
Raza SS, Zafar U, Shehwar DE, Amin H. Comparison of Pulmonary Functions After Induction of Stress Between Post-COVID and Healthy Adults. Cureus 2023; 15:e43612. [PMID: 37719508 PMCID: PMC10504465 DOI: 10.7759/cureus.43612] [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] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
Since the emergence of COVID in 2019, it has spread worldwide. COVID has affected all the systems of the human body. The present research aimed to assess the effects of COVID-19 on the pulmonary system after stress induction. Healthy and affected individuals between the age of 18 and 40 years were made to perform the 6-minute walk test and their pulmonary functions were compared before and after the stressor. Individuals who were three months post-COVID-19 infection were included as cases. Healthy individuals with no history of COVID were included as controls. The pulmonary functions were performed and noted both at baseline and after the 6-minute walk test. The forced expiratory flow 25 (FEF 25) and peak expiratory flow (PEF) showed statistical significance between both groups (p=0.033 and p=0.007, respectively). FEF 25, 50, and 75, maximum voluntary ventilation (MVV) index, and PEF were positively correlated with all respiratory parameters. Forced expiratory volume % (FEV%) was negatively correlated with vital capacity (VC) and forced vital capacity (FVC). This research helped us establish that the effect on the lungs due to COVID is not due to airway restriction or obstruction but reduced lung volume.
Collapse
Affiliation(s)
- Syed S Raza
- Department of Physiology, Gajju Khan Medical College, Swabi, PAK
- Robert and Suzanne Tomsich Department of Cardiothoracic Surgery, Cleveland Clinic Florida, Peshawar, PAK
| | - Umema Zafar
- Department of Physiology, Khyber Medical University, Peshawar, PAK
- Department of Physiology, Rehman Medical College, Peshawar, PAK
| | - Dur E Shehwar
- Department of Physiology, Khyber Medical College, Peshawar, PAK
| | - Hunya Amin
- Department of Physiology, Khyber Medical College, Peshawar, PAK
| |
Collapse
|
27
|
Peron JPS. Direct and indirect impact of SARS-CoV-2 on the brain. Hum Genet 2023; 142:1317-1326. [PMID: 37004544 PMCID: PMC10066989 DOI: 10.1007/s00439-023-02549-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/22/2023] [Indexed: 04/04/2023]
Abstract
Although COVID-19 is mostly a pulmonary disease, it is now well accepted that it can cause a much broader spectrum of signs and symptoms and affect many other organs and tissue. From mild anosmia to severe ischemic stroke, the impact of SARS-CoV-2 on the central nervous system is still a great challenge to scientists and health care practitioners. Besides the acute and severe neurological problems described, as encephalopathies, leptomeningitis, and stroke, after 2 years of pandemic, the chronic impact observed during long-COVID or the post-acute sequelae of COVID-19 (PASC) greatly intrigues scientists worldwide. Strikingly, even asymptomatic, and mild diseased patients may evolve with important neurological and psychiatric symptoms, as confusion, memory loss, cognitive decline, chronic fatigue, associated or not with anxiety and depression. Thus, the knowledge on the correlation between COVID-19 and the central nervous system is of great relevance. In this sense, here we discuss some important mechanisms obtained from in vitro and in vivo investigation regarding how SARS-CoV-2 impacts the brain and its cells and function.
Collapse
Affiliation(s)
- J P S Peron
- Neuroimmune Interactions Laboratory, Department of Immunology, University of Sao Paulo, Av. Prof. Lineu Prestes, 1730 Lab 232. Cidade Universitária, São Paulo, SP, CEP 05508-000, Brazil.
| |
Collapse
|
28
|
Guo TJF, Singhera GK, Leung JM, Dorscheid DR. Airway Epithelial-Derived Immune Mediators in COVID-19. Viruses 2023; 15:1655. [PMID: 37631998 PMCID: PMC10458661 DOI: 10.3390/v15081655] [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: 06/13/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
The airway epithelium, which lines the conducting airways, is central to the defense of the lungs against inhaled particulate matter and pathogens such as SARS-CoV-2, the virus that causes COVID-19. Recognition of pathogens results in the activation of an innate and intermediate immune response which involves the release of cytokines and chemokines by the airway epithelium. This response can inhibit further viral invasion and influence adaptive immunity. However, severe COVID-19 is characterized by a hyper-inflammatory response which can give rise to clinical presentations including lung injury and lead to acute respiratory distress syndrome, viral pneumonia, coagulopathy, and multi-system organ failure. In response to SARS-CoV-2 infection, the airway epithelium can mount a maladaptive immune response which can delay viral clearance, perpetuate excessive inflammation, and contribute to the pathogenesis of severe COVID-19. In this article, we will review the barrier and immune functions of the airway epithelium, how SARS-CoV-2 can interact with the epithelium, and epithelial-derived cytokines and chemokines and their roles in COVID-19 and as biomarkers. Finally, we will discuss these immune mediators and their potential as therapeutic targets in COVID-19.
Collapse
Affiliation(s)
- Tony J. F. Guo
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
| | - Gurpreet K. Singhera
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| | - Janice M. Leung
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| | - Delbert R. Dorscheid
- Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul’s Hospital, University of British Columbia, 1081 Burrard St., Vancouver, BC V6Z 1Y6, Canada
- Department of Medicine, University of British Columbia, 2775 Laurel St., Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
29
|
Kalfas T, Kaltsas A, Symeonidis EN, Symeonidis A, Zikopoulos A, Moustakli E, Tsiampali C, Tsampoukas G, Palapela N, Zachariou A, Sofikitis N, Dimitriadis F. COVID-19 and Male Infertility: Is There a Role for Antioxidants? Antioxidants (Basel) 2023; 12:1483. [PMID: 37627478 PMCID: PMC10451649 DOI: 10.3390/antiox12081483] [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: 06/12/2023] [Revised: 07/14/2023] [Accepted: 07/22/2023] [Indexed: 08/27/2023] Open
Abstract
Coronavirus disease 19 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), jeopardizes male fertility because of the vulnerability of the male reproductive system, especially the testes. This study evaluates the effects of the virus on testicular function and examines the potential role of antioxidants in mitigating the damage caused by oxidative stress (OS). A comprehensive PubMed search examined exocrine and endocrine testicular function alteration, the interplay between OS and COVID-19-induced defects, and the potential benefit of antioxidants. Although the virus is rarely directly detectable in sperm and testicular tissue, semen quality and hormonal balance are affected in patients, with some changes persisting throughout a spermatogenesis cycle. Testicular pathology in deceased patients shows defects in spermatogenesis, vascular changes, and inflammation. Acute primary hypogonadism is observed mainly in severely infected cases. Elevated OS and sperm DNA fragmentation markers suggest redox imbalance as a possible mechanism behind the fertility changes. COVID-19 vaccines appear to be safe for male fertility, but the efficacy of antioxidants to improve sperm quality after infection remains unproven due to limited research. Given the limited and inconclusive evidence, careful evaluation of men recovering from COVID-19 seeking fertility improvement is strongly recommended.
Collapse
Affiliation(s)
| | - Aris Kaltsas
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (E.M.); (A.Z.); (N.S.)
| | - Evangelos N. Symeonidis
- Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.N.S.); (A.S.)
| | - Asterios Symeonidis
- Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.N.S.); (A.S.)
| | - Athanasios Zikopoulos
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (E.M.); (A.Z.); (N.S.)
| | - Efthalia Moustakli
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (E.M.); (A.Z.); (N.S.)
| | | | - Georgios Tsampoukas
- Department of Urology, Oxford University Hospital NHS Trust, Oxford OX3 7LE, UK;
| | - Natalia Palapela
- Medical Faculty, Medical University of Sofia, 1431 Sofia, Bulgaria;
| | - Athanasios Zachariou
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (E.M.); (A.Z.); (N.S.)
| | - Nikolaos Sofikitis
- Department of Urology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (A.K.); (A.Z.); (E.M.); (A.Z.); (N.S.)
| | - Fotios Dimitriadis
- Department of Urology, Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.N.S.); (A.S.)
| |
Collapse
|
30
|
Joni SS, Gerami R, Pashaei F, Ebrahiminik H, Karimi M. Quantitative evaluation of CT scan images to determinate the prognosis of COVID-19 patient using deep learning. Eur J Transl Myol 2023; 33:11571. [PMID: 37491956 PMCID: PMC10583151 DOI: 10.4081/ejtm.2023.11571] [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: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 07/27/2023] Open
Abstract
The purpose of this research is to evaluate the accuracy of AI-assisted quantification in comparison to conventional CT parameters reviewed by a radiologist in predicting the severity, progression, and clinical outcome of disease. The current study is a cross-sectional study that was conducted on patients with the diagnosis of COVID-19 and underwent a pulmonary CT scan between August 23th, 2021 to December 21th, 2022. The initial CT scan on admission was used for imaging analysis. The presence of ground glass opacity (GGO), and consolidation were visually evaluated. CT severity score was calculated according to a semi-quantitative method. In addition, AI based quantification of GGO and consolidation volume were also performed. 291 patients (mean age: 64.7 ± 7; 129 males) were included. GGO + consolidation was more frequently revealed in progress-to-severe group whereas pure GGO was more likely to be found in non-severe group. Compared to non-severe group, patients in progress-to-severe group had larger GGO volume percentage (40.6%± 11.9%versus 21.7%± 8.8%, p ˂0.001) as well as consolidation volume percentage (4.8% ± 2% versus 1.9% ± 1%, p < 0.001). Among imaging parameters, consolidation volume percentage and the largest area under curve (AUC) in discriminating non-severe from progress-to-severe group (AUC = 0.91, p < 0.001). According to multivariate regression, consolidation volume was the strongest predictor for disease progression. In conclusion, the consolidation volume measured on the initial chest CT was the most accurate predictor of disease progression, and a larger consolidation volume was associated with a poor clinical outcome. In patients with COVID-19, AI-assisted lesion quantification was useful for risk stratification and prognosis evaluation.
Collapse
Affiliation(s)
- Saeid Sadeghi Joni
- Department of Radiology, Faculty of medicine, Aja University of Medical Sciences, Tehran.
| | - Reza Gerami
- Department of Radiology, Faculty of medicine, Aja University of Medical Sciences, Tehran.
| | - Fakhereh Pashaei
- Radiation Sciences Research Center (RSRC), Aja University of Medical Sciences, Tehran.
| | - Hojat Ebrahiminik
- Department of Interventional Radiology and Radiation Sciences Research Center, Aja University of Medical Sciences, Tehran.
| | - Mahmood Karimi
- Department of Internal Medicine, Faculty of Medicine, AJA University of Medical Sciences, Tehran.
| |
Collapse
|
31
|
Lv K, Li M, Sun C, Miao Y, Zhang Y, Liu Y, Guo J, Meng Q, Yao J, Zhang G, Li J. Jingfang Granule alleviates bleomycin-induced acute lung injury via CD200-CD200R immunoregulatory pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 311:116423. [PMID: 37011735 DOI: 10.1016/j.jep.2023.116423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/01/2023] [Accepted: 03/21/2023] [Indexed: 06/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jingfang granules (JF), one famous traditional Chinese formula in "She Sheng Zhong Miao Fang" written by Shi-Che Zhang during the Ming Dynasty era, has been widely used to prevent epidemic diseases in history and now was recommended for the treatment of coronavirus disease 2019 (COVID-19) in China. However, the roles of JF against acute lung injury and its mechanisms remain unclear. AIM OF THE STUDY Acute lung injury (ALI) and its progressive acute respiratory distress syndrome (ARDS) are a continuum of lung inflammatory disease with high morbidity and mortality in clinic, especially in COVID-19 patients. The present study aims to investigate the effect of JF on ALI and clarify its underlying mechanisms for clinical application in COVID-19 control. METHODS Bleomycin-induced ALI mice were given oral gavage daily for seven days with or without Jingfang granules (2, 4 g/kg). The body weight, lung wet/dry weight ratios, lung appearance and tissue histopathology were evaluated. Quantitative real-time PCR, biochemical bronchoalveolar lavage fluids analysis was used to determine the gene expression of proinflammation factor and infiltrated inflammatory cells in lung. Immunofluorescence image and western blot were used to detect the markers of alveolar macrophages (AMs), endothelial cell apoptosis and changes of CD200-CD200R pathway. RESULTS Firstly, histopathological analysis showed that JF significantly attenuated pulmonary injury and inflammatory response in ALI mice. Then, cytokine detection, inflammatory cells assay, and JNKs and p38 pathway analysis indicated that the recruitment and activation of alveolar macrophages was the main reason to cause ALI and JF could reverse this variation. Next, immunofluorescence staining and TUNEL assay showed that JF upregulated the expression of CD200 and suppressed the apoptosis of alveolar endothelial cells. Finally, double immunofluorescence staining of CD200 and CD11c indicated that the seriously damaged tissue had the lower CD200 while more AMs infiltration, which was confirmed by RT-PCR analysis of CD200/CD200R. CONCLUSIONS Jingfang granules can protect lung from acu te injury and mitigate the recruitment and overactive AMs-induced inflammation via CD200-CD200R immunoregulatory signal axis, which will provide an experimental basis for Jingfang granules clinical applications in COVID-19.
Collapse
Affiliation(s)
- Ke Lv
- The State Key Laboratory of Medicinal Chemical Biology & College of Chemistry, Nankai University, Tianjin, 300071, China.
| | - Mingyue Li
- College of Pharmacy, Nankai University, Tianjin, 300071, China.
| | - Chenghong Sun
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Yu Miao
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Yan Zhang
- The State Key Laboratory of Medicinal Chemical Biology & College of Chemistry, Nankai University, Tianjin, 300071, China.
| | - Yang Liu
- College of Pharmacy, Nankai University, Tianjin, 300071, China.
| | - Jianshuang Guo
- College of Pharmacy, Nankai University, Tianjin, 300071, China.
| | - Qing Meng
- College of Pharmacy, Nankai University, Tianjin, 300071, China.
| | - Jingchun Yao
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Guimin Zhang
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Jing Li
- The State Key Laboratory of Medicinal Chemical Biology & College of Chemistry, Nankai University, Tianjin, 300071, China; College of Pharmacy, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
32
|
Kloping YP, Hidayatullah F, Rahman ZA, Chung E, Hakim L. Male Reproductive Tract Involvement and Sperm Parameters in SARS-CoV-2 Patients: A Systematic Review and Meta-Analysis. World J Mens Health 2023; 41:538-557. [PMID: 36102101 PMCID: PMC10307665 DOI: 10.5534/wjmh.220019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/06/2022] [Accepted: 05/15/2022] [Indexed: 11/26/2023] Open
Abstract
PURPOSE There is a growing concern regarding the impact of SARS-CoV-2 infection on the male reproductive tract due to ACE2 receptor expression, however, its impact remains unclear. We performed this review to evaluate whether SARS-CoV-2 infection affects the male reproductive system. MATERIALS AND METHODS We conducted a search in the Embase, Scopus, and MEDLINE databases, adhering to the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guideline. Eligible studies comprised articles reporting viral RNA presence in semen, sperm parameters, and orchitis or orchiepididymitis occurrence in SARS-CoV-2 patients. Observational studies' quality was determined using the Newcastle-Ottawa Scale (NOS). Case reports were assessed using the Joanna Briggs Institute (JBI)'s checklist. RESULTS A total of 32 relevant articles were included. Viral RNA was found in 7% of infected patients' semen (95% CI, -0.01 to 0.15) from 3 studies. There were also only 7% of patients with orchitis or orchiepididymitis clinical manifestations (95% CI, 0.05-0.10). The semen volume and concentration were 2.34 mL (95% CI, 1.87-2.81) and 51.73 million/mL (95% CI, 31.60-71.85). The progressive and total motility percentages were 36.11% (95% CI, 28.87-43.35) and 43.07% (95% CI, 28.57-57.57), respectively. The morphology was 6.03% (95% CI, -1.05 to 13.10). There is a difference in semen volume between moderate and severe infections (MD, 0.52; 95% CI, 0.27-0.76; p<0.0001) and concentration between mild and moderate (MD, 18.74; 95% CI, 1.02-36.46; p=0.04), mild and severe (MD, 43.50; 95% CI, 13.86-73.14; p=0.004), as well as moderate and severe (MD, 22.25; 95% CI, 9.33-35.17; p=0.0007). CONCLUSIONS SARS-CoV-2 infection may result in decreased sperm concentration in severe cases and the mechanism relates to potential reproductive tract inflammation. The absence of large viral RNA detection in the semen indicates a systemic effect, although this is largely unproven.
Collapse
Affiliation(s)
- Yudhistira Pradnyan Kloping
- Department of Urology, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
- Department of Urology, Dr. Soetomo General-Academic Hospital, Surabaya, East Java, Indonesia
| | - Furqan Hidayatullah
- Department of Urology, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
- Department of Urology, Dr. Soetomo General-Academic Hospital, Surabaya, East Java, Indonesia
| | - Zakaria Aulia Rahman
- Department of Urology, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
- Department of Urology, Dr. Soetomo General-Academic Hospital, Surabaya, East Java, Indonesia
| | - Eric Chung
- Department of Urology, Princess Alexandra Hospital, University of Queensland, Brisbane, QLD, Australia
| | - Lukman Hakim
- Department of Urology, Faculty of Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
- Department of Urology, Universitas Airlangga Teaching Hospital, Surabaya, East Java, Indonesia.
| |
Collapse
|
33
|
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
|
34
|
Sasson J, Moreau GB, Petri WA. The role of interleukin 13 and the type 2 immune pathway in COVID-19: A review. Ann Allergy Asthma Immunol 2023; 130:727-732. [PMID: 36924937 PMCID: PMC10014128 DOI: 10.1016/j.anai.2023.03.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/23/2023] [Accepted: 03/06/2023] [Indexed: 03/16/2023]
Abstract
Although much has been learned about severe acute respiratory syndrome coronavirus 2 since December 2019, uneven global vaccine distribution, rapid viral spread, and variant evasion of preventative measures have led to its persistence in the population for the foreseeable future. Additional therapies are needed to support patients through their acute, immune-mediated disease process that continues to lead to considerable morbidity and mortality. Data revealing the involvement of type 2 immune pathway in acute coronavirus disease 2019 and post-recovery conditions represent a potential additional area for intervention. Herein, we review the current understanding of interleukin 13 in acute severe acute respiratory syndrome coronavirus 2 infection, the clinical outcomes associated with type 2 immune processes, and the impact of type 2 blockade on acute and long-term coronavirus disease 2019 conditions.
Collapse
Affiliation(s)
- Jennifer Sasson
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - G Brett Moreau
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - William A Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia; Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia; Department of Pathology, University of Virginia Health System, Charlottesville, Virginia.
| |
Collapse
|
35
|
Wu K, Zhang Y, Austin SR, Yin-Declue H, Byers DE, Crouch EC, Holtzman MJ. Lung Remodeling Regions in Long-Term Coronavirus Disease 2019 Feature Basal Epithelial Cell Reprogramming. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:680-689. [PMID: 36868468 PMCID: PMC9977469 DOI: 10.1016/j.ajpath.2023.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 03/05/2023]
Abstract
Respiratory viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), can trigger chronic lung disease that persists and even progresses after expected clearance of infectious virus. To gain an understanding of this process, the current study examined a series of consecutive fatal cases of coronavirus disease 2019 (COVID-19) that came to autopsy at 27 to 51 days after hospital admission. In each patient, a stereotyped bronchiolar-alveolar pattern of lung remodeling was identified with basal epithelial cell hyperplasia, immune activation, and mucinous differentiation. Remodeling regions featured macrophage infiltration and apoptosis and a marked depletion of alveolar type 1 and 2 epithelial cells. This pattern closely resembled findings from an experimental model of post-viral lung disease that requires basal-epithelial stem cell growth, immune activation, and differentiation. Together, these results provide evidence of basal epithelial cell reprogramming in long-term COVID-19 and thereby yield a pathway for explaining and correcting lung dysfunction in this type of disease.
Collapse
Affiliation(s)
- Kangyun Wu
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Stephen R Austin
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Huiqing Yin-Declue
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Derek E Byers
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Erika C Crouch
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
36
|
Cong Y, Lee JH, Perry DL, Cooper K, Wang H, Dixit S, Liu DX, Feuerstein IM, Solomon J, Bartos C, Seidel J, Hammoud DA, Adams R, Anthony SM, Liang J, Schuko N, Li R, Liu Y, Wang Z, Tarbet EB, Hischak AMW, Hart R, Isic N, Burdette T, Drawbaugh D, Huzella LM, Byrum R, Ragland D, St Claire MC, Wada J, Kurtz JR, Hensley LE, Schmaljohn CS, Holbrook MR, Johnson RF. Longitudinal analyses using 18F-Fluorodeoxyglucose positron emission tomography with computed tomography as a measure of COVID-19 severity in the aged, young, and humanized ACE2 SARS-CoV-2 hamster models. Antiviral Res 2023; 214:105605. [PMID: 37068595 PMCID: PMC10105383 DOI: 10.1016/j.antiviral.2023.105605] [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: 01/30/2023] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/19/2023]
Abstract
This study compared disease progression of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in three different models of golden hamsters: aged (≈60 weeks old) wild-type (WT), young (6 weeks old) WT, and adult (14-22 weeks old) hamsters expressing the human-angiotensin-converting enzyme 2 (hACE2) receptor. After intranasal (IN) exposure to the SARS-CoV-2 Washington isolate (WA01/2020), 2-deoxy-2-[fluorine-18]fluoro-D-glucose positron emission tomography with computed tomography (18F-FDG PET/CT) was used to monitor disease progression in near real time and animals were euthanized at pre-determined time points to directly compare imaging findings with other disease parameters associated with coronavirus disease 2019 (COVID-19). Consistent with histopathology, 18F-FDG-PET/CT demonstrated that aged WT hamsters exposed to 105 plaque forming units (PFU) developed more severe and protracted pneumonia than young WT hamsters exposed to the same (or lower) dose or hACE2 hamsters exposed to a uniformly lethal dose of virus. Specifically, aged WT hamsters presented with a severe interstitial pneumonia through 8 d post-exposure (PE), while pulmonary regeneration was observed in young WT hamsters at that time. hACE2 hamsters exposed to 100 or 10 PFU virus presented with a minimal to mild hemorrhagic pneumonia but succumbed to SARS-CoV-2-related meningoencephalitis by 6 d PE, suggesting that this model might allow assessment of SARS-CoV-2 infection on the central nervous system (CNS). Our group is the first to use (18F-FDG) PET/CT to differentiate respiratory disease severity ranging from mild to severe in three COVID-19 hamster models. The non-invasive, serial measure of disease progression provided by PET/CT makes it a valuable tool for animal model characterization.
Collapse
Affiliation(s)
- Yu Cong
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Ji Hyun Lee
- Radiology and Imaging Sciences, Clinical Center, National Institute of Health, Bethesda, MD, USA
| | - Donna L Perry
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Kurt Cooper
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Hui Wang
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Saurabh Dixit
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - David X Liu
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Irwin M Feuerstein
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Jeffrey Solomon
- Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Christopher Bartos
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Jurgen Seidel
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Ricky Adams
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Scott M Anthony
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Janie Liang
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Nicolette Schuko
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Rong Li
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, USA.
| | - Yanan Liu
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Zhongde Wang
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - E Bart Tarbet
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Amanda M W Hischak
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Randy Hart
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Nejra Isic
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Tracey Burdette
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA; Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - David Drawbaugh
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Louis M Huzella
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Russell Byrum
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Danny Ragland
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Marisa C St Claire
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Jiro Wada
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Jonathan R Kurtz
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Lisa E Hensley
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Connie S Schmaljohn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Michael R Holbrook
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA.
| | - Reed F Johnson
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA; SARS-CoV-2 Virology Core Laboratory, Division of Intramural Research, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
37
|
Gupta A, Marzook H, Ahmad F. Comorbidities and clinical complications associated with SARS-CoV-2 infection: an overview. Clin Exp Med 2023; 23:313-331. [PMID: 35362771 PMCID: PMC8972750 DOI: 10.1007/s10238-022-00821-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/12/2022] [Indexed: 01/08/2023]
Abstract
The novel severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) causes major challenges to the healthcare system. SARS-CoV-2 infection leads to millions of deaths worldwide and the mortality rate is found to be greatly associated with pre-existing clinical conditions. The existing dataset strongly suggests that cardiometabolic diseases including hypertension, coronary artery disease, diabetes and obesity serve as strong comorbidities in coronavirus disease (COVID-19). Studies have also shown the poor outcome of COVID-19 in patients associated with angiotensin-converting enzyme-2 polymorphism, cancer chemotherapy, chronic kidney disease, thyroid disorder, or coagulation dysfunction. A severe complication of COVID-19 is mostly seen in people with compromised medical history. SARS-CoV-2 appears to attack the respiratory system causing pneumonia, acute respiratory distress syndrome, which lead to induction of severe systemic inflammation, multi-organ dysfunction, and death mostly in the patients who are associated with pre-existing comorbidity factors. In this article, we highlighted the key comorbidities and a variety of clinical complications associated with COVID-19 for a better understanding of the etiopathogenesis of COVID-19.
Collapse
Affiliation(s)
- Anamika Gupta
- Cardiovascular Research Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, UAE
| | - Hezlin Marzook
- Cardiovascular Research Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, UAE
| | - Firdos Ahmad
- Cardiovascular Research Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, UAE.
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, UAE.
| |
Collapse
|
38
|
Awatade NT, Wark PAB, Chan ASL, Mamun SMAA, Mohd Esa NY, Matsunaga K, Rhee CK, Hansbro PM, Sohal SS. The Complex Association between COPD and COVID-19. J Clin Med 2023; 12:jcm12113791. [PMID: 37297985 DOI: 10.3390/jcm12113791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is significant cause of morbidity and mortality worldwide. There is mounting evidence suggesting that COPD patients are at increased risk of severe COVID-19 outcomes; however, it remains unclear whether they are more susceptible to acquiring SARS-CoV-2 infection. In this comprehensive review, we aim to provide an up-to-date perspective of the intricate relationship between COPD and COVID-19. We conducted a thorough review of the literature to examine the evidence regarding the susceptibility of COPD patients to COVID-19 infection and the severity of their disease outcomes. While most studies have found that pre-existing COPD is associated with worse COVID-19 outcomes, some have yielded conflicting results. We also discuss confounding factors such as cigarette smoking, inhaled corticosteroids, and socioeconomic and genetic factors that may influence this association. Furthermore, we review acute COVID-19 management, treatment, rehabilitation, and recovery in COPD patients and how public health measures impact their care. In conclusion, while the association between COPD and COVID-19 is complex and requires further investigation, this review highlights the need for careful management of COPD patients during the pandemic to minimize the risk of severe COVID-19 outcomes.
Collapse
Affiliation(s)
- Nikhil T Awatade
- Immune Health Program, Hunter Medical Research Institute and University of Newcastle, Newcastle 2305, Australia
| | - Peter A B Wark
- Immune Health Program, Hunter Medical Research Institute and University of Newcastle, Newcastle 2305, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle 2305, Australia
| | - Andrew S L Chan
- Department of Respiratory and Sleep Medicine, Royal North Shore Hospital, St. Leonards 2065, Australia
- Northern Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney 2006, Australia
| | - S M Abdullah Al Mamun
- Department of Respiratory Medicine & Sleep Medicine, Evercare Hospitals Dhaka, Dhaka 1229, Bangladesh
| | | | - Kazuto Matsunaga
- Department of Respiratory Medicine and Infectious Disease Graduate School of Medicine, Yamaguchi University, 1-1-1 Minami-kogushi, Ube 755-8505, Japan
| | - Chin Kook Rhee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Philip M Hansbro
- Immune Health Program, Hunter Medical Research Institute and University of Newcastle, Newcastle 2305, Australia
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney 2050, Australia
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston 7248, Australia
| |
Collapse
|
39
|
Friedman A, Ulrich M. Case report: Spontaneous hemothorax following anticoagulation for microthrombi in severe COVID pneumonia. Respir Med Case Rep 2023; 44:101864. [PMID: 37332847 PMCID: PMC10272501 DOI: 10.1016/j.rmcr.2023.101864] [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: 03/02/2023] [Revised: 04/18/2023] [Accepted: 05/10/2023] [Indexed: 06/20/2023] Open
Abstract
Background Anticoagulant therapy has become a hallmark of treatment for critically ill COVID patients. Gastrointestinal and intracranial hemorrhage are known major complications of anticoagulation, but spontaneous hemothorax is a rare event, particularly in the absence of pre-existing structural lung disease, vascular malformations, or genetic bleeding diatheses. Herein is a case of spontaneous hemothorax following anticoagulation for microthrombi in a patient with acute hypoxic respiratory failure due to Covid pneumonia. Case presentation A 49 year old male with hypertension, asthma, and obesity was admitted for acute hypoxic respiratory failure due to Covid pneumonia. He was treated with dexamethasone, baricitinib, and therapeutic enoxaparin as empiric therapy for severe COVID disease. He subsequently developed a massive right hemothorax with associated hemorrhagic shock, which required initiation of massive transfusion protocol, vasopressor support and mechanical ventilation. No clear etiology for the hemothorax was determined upon investigations. The patient eventually improved and was discharged to a skilled nursing facility on chronic oxygen therapy. Conclusions Various mechanisms have been proposed for the development of non-traumatic hemothoraces, including tearing of adhesions and rupture of vascularized bullae. Such explanations find support in radiologic and pathologic studies of pleural changes in Covid pneumonia, and likely played a role in the hemorrhage experienced by our patient.
Collapse
Affiliation(s)
- Alexander Friedman
- Corresponding author. Department of Internal Medicine, Riverside University Health Systems Medical Center, 26520 Cactus Ave, Moreno Valley, CA, 92555, USA.
| | | |
Collapse
|
40
|
Barbalho SM, Minniti G, Miola VFB, Haber JFDS, Bueno PCDS, de Argollo Haber LS, Girio RSJ, Detregiachi CRP, Dall'Antonia CT, Rodrigues VD, Nicolau CCT, Catharin VMCS, Araújo AC, Laurindo LF. Organokines in COVID-19: A Systematic Review. Cells 2023; 12:1349. [PMID: 37408184 DOI: 10.3390/cells12101349] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 07/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a viral infection caused by SARS-CoV-2 that induces a generalized inflammatory state. Organokines (adipokines, osteokines, myokines, hepatokines, and cardiokines) can produce beneficial or harmful effects in this condition. This study aimed to systematically review the role of organokines on COVID-19. PubMed, Embase, Google Scholar, and Cochrane databases were searched, the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed, and 37 studies were selected, comprising more than 2700 individuals infected with the virus. Among COVID-19 patients, organokines have been associated with endothelial dysfunction and multiple organ failure due to augmented cytokines and increased SARS-CoV-2 viremia. Changes in the pattern of organokines secretion can directly or indirectly contribute to aggravating the infection, promoting immune response alterations, and predicting the disease progression. These molecules have the potential to be used as adjuvant biomarkers to predict the severity of the illness and severe outcomes.
Collapse
Affiliation(s)
- Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília 17500-000, SP, Brazil
| | - Giulia Minniti
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Vitor Fernando Bordin Miola
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Jesselina Francisco Dos Santos Haber
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Centro Interdisciplinar em Diabetes (CENID), School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Patrícia Cincotto Dos Santos Bueno
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Department of Animal Sciences, School of Veterinary Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Luiza Santos de Argollo Haber
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Raul S J Girio
- Department of Animal Sciences, School of Veterinary Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Cláudia Rucco Penteado Detregiachi
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Department of Biochemistry and Pharmacology, Faculdade de Medicina de Marília (FAMEMA), School of Medicine, Avenida Monte Carmelo, 800, Marília 17519-030, SP, Brazil
| | - Camila Tiveron Dall'Antonia
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Victória Dogani Rodrigues
- Department of Biochemistry and Pharmacology, Faculdade de Medicina de Marília (FAMEMA), School of Medicine, Avenida Monte Carmelo, 800, Marília 17519-030, SP, Brazil
| | - Claudia C T Nicolau
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília 17500-000, SP, Brazil
| | - Virginia Maria Cavallari Strozze Catharin
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília 17525-902, SP, Brazil
- Department of Biochemistry and Pharmacology, Faculdade de Medicina de Marília (FAMEMA), School of Medicine, Avenida Monte Carmelo, 800, Marília 17519-030, SP, Brazil
| |
Collapse
|
41
|
Çınar C, Balaban Genç ZC, Kesim S, Çağlıyan Şen F, Karakurt S, Erdil TY, Öneş T, Eryuksel E. Effect of Anticoagulants in Pulmonary Thromboembolism in Post-COVID-19 Patients. Cureus 2023; 15:e39382. [PMID: 37362508 PMCID: PMC10286523 DOI: 10.7759/cureus.39382] [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] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Background and objective COVID-19 may predispose to both venous and arterial thromboembolism (VTE and ATE) due to excessive inflammation, immobilization, and hypoxia. The purpose of this study is to evaluate clinical and laboratory risk factors, as well as related medications such as anticoagulants, to predict the risk of thromboembolic disease and/or death in COVID-19 patients. Methods Over a period of 14 months (from August 2020 to September 2021), a total of 145 consecutive patients with signs and symptoms suspicious of pulmonary embolism (PE) were referred for perfusion single-photon emission computed tomography/computed tomography (Q SPECT/CT). All patients had a history of SARS‑CoV‑2 infection, diagnosed with a positive real-time polymerase chain reaction (RT-PCR) test. Results Among the 145 patients included in the study, the risk of PE was found to be greater in elderly patients (odds ratio [OR] [95% CI]: 1.05 [1.02‑1.07]; p<0.001) and in patients with higher maximum d-dimer levels (OR [95% CI]: 1.14 [1.01‑1.3]; p=0.04). We also analyzed the utility of the maximum d-dimer level for predicting acute PE with receiver operating characteristic (ROC) curve analysis. For d‑dimer = 0.5 mg/dL, cut-off sensitivity is 91%, specificity is 23%, and for d-dimer = 1 mg/dL, cut-off sensitivity is 79%, specificity is 43% Conclusion D-dimer titers were higher in the PE group in our study. Another significant finding was that, possibly due to thromboinflammation, anticoagulants did not prevent the development of PE in COVID-19 patients.
Collapse
Affiliation(s)
- Caner Çınar
- Department of Pulmonology, Marmara University, Istanbul, TUR
| | | | - Selin Kesim
- Department of Nuclear Medicine, Marmara University, Istanbul, TUR
| | | | - Sait Karakurt
- Department of Pulmonology, Marmara University, Istanbul, TUR
| | | | - Tunç Öneş
- Department of Nuclear Medicine, Marmara University, Istanbul, TUR
| | - Emel Eryuksel
- Department of Pulmonology, Marmara University, Istanbul, TUR
| |
Collapse
|
42
|
Banko A, Miljanovic D, Cirkovic A. Systematic review with meta-analysis of active herpesvirus infections in patients with COVID-19: Old players on the new field. Int J Infect Dis 2023; 130:108-125. [PMID: 36736577 PMCID: PMC9889115 DOI: 10.1016/j.ijid.2023.01.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVES Herpesviruses are ubiquitous and after primary infection they establish lifelong latency. The impairment of maintaining latency with short-term or long-term consequences could be triggered by other infection. Therefore, reactivation of herpesviruses in COVID-19 patients represents an emerging issue. DESIGN AND METHODS This study provided the first systematic review with meta-analysis of studies that evaluated active human herpesvirus (HHV) infection (defined as the presence of IgM antibodies or HHV-DNA) in COVID-19 patients and included 36 publications collected by searching through PubMed, SCOPUS, and Web of science until November 2022. RESULTS The prevalence of active EBV, HHV6, HSV, CMV, HSV1, and VZV infection in COVID-19 population was 41% (95% CI =27%-57%), 3% (95% CI=17%-54%), 28% (95% CI=1%-85%), 25% (95% CI=1%-63%), 22% (95% CI=10%-35%), and 18% (95% CI=4%-34%), respectively. There was a 6 times higher chance for active EBV infection in patients with severe COVID-19 than in non-COVID-19 controls (OR=6.45, 95% CI=1.09-38.13, p=0.040), although there was no difference in the prevalence of all evaluated active herpesvirus infections between COVID-19 patients and non-COVID-19 controls. CONCLUSIONS Future research of herpesvirus and SARS-CoV-2 coinfections must be prioritized to define: who, when and how to be tested, as well as how to effectively treat HHVs reactivations in acute and long COVID-19 patients.
Collapse
Affiliation(s)
- Ana Banko
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
| | - Danijela Miljanovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
| | - Andja Cirkovic
- Institute for Medical Statistics and Informatics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
43
|
Nappi F, Avtaar Singh SS. SARS-CoV-2-Induced Myocarditis: A State-of-the-Art Review. Viruses 2023; 15:916. [PMID: 37112896 PMCID: PMC10145666 DOI: 10.3390/v15040916] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/25/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
In this review, we investigated whether severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can directly cause myocarditis with severe myocardial damage induced by viral particles. A review of the major data published from 2020 to 2022 was performed by consulting the major databases alongside first-hand experiences that emerged from the cardiac biopsies and autopsy examinations of patients who died of SARS-CoV-2 infections. From this study, a significantly large amount of data suggests that the Dallas criteria were met in a residual percentage of patients, demonstrating that SARS-CoV-2 myocarditis was a rare clinical and pathological entity that occurred in a small percentage of subjects. All cases described here were highly selected and subjected to autopsies or endomyocardial biopsies (EMBs). The most important discovery, through the detection of the SARS-CoV-2 genome using the polymerase chain reaction, consisted in the presence of the viral genome in the lung tissue of most of the patients who died from COVID-19. However, the discovery of the SARS-CoV-2 viral genome was a rare event in cardiac tissue from autopsy findings of patients who died of myocarditis It is important to emphasize that myocardial inflammation alone, as promoted by macrophages and T cell infiltrations, can be observed in noninfectious deaths and COVID-19 cases, but the extent of each cause is varied, and in neither case have such findings been reported to support clinically relevant myocarditis. Therefore, in the different infected vs. non-infected samples examined, none of our findings provide a definitive histochemical assessment for the diagnosis of myocarditis in the majority of cases evaluated. We report evidence suggesting an extremely low frequency of viral myocarditis that has also been associated with unclear therapeutic implications. These two key factors strongly point towards the use of an endomyocardial biopsy to irrefutably reach a diagnosis of viral myocarditis in the context of COVID-19.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| | | |
Collapse
|
44
|
Cuenca-Pardo J, Ramos-Gallardo G, Morales-Olivera M, Contreras-Bulnes L, Bucio-Duarte J, Iribarren-Moreno R, Vélez-Benítez E, Domínguez-Millán R, Salmean-Piña O. HOW TO STRATIFY THE RISK OF THROMBOSIS FOR AESTHETIC PLASTIC SURGERY? THE PROPOSAL OF A NEW SCALE. J Plast Reconstr Aesthet Surg 2023. [PMID: 37302242 DOI: 10.1016/j.bjps.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
Scales to qualify the risk of thrombosis do not include all thrombogenic factors that are generated in esthetic plastic surgery. Methods: We performed a systematic review to assess the risk of thrombosis in plastic surgery. Thrombogenic factors in esthetic surgery were analyzed by a panel of experts. We proposed a scale with 2 versions. In the first version, factors were stratified according to their impact on the possible risk of thrombosis. The second version includes the same factors but in a simplified form. We evaluated the efficacy of the proposed scale by comparing it with the Caprini score; we scored the risk in 124 cases and controls. Results: Using the Caprini score, we found that 81.45% of the patients studied and 62.5% of the cases of thrombosis were observed in the low-risk group. Only 1 case of thrombosis was reported in the high-risk group. Using the stratified version of the scale, we found that the low-risk group comprised 25% of the patients, and there were no cases of thrombosis. The high-risk group included 14.51% of patients; 10 presented thrombosis (62.5%). The proposed scale was very effective in detecting both low-risk and high-risk patients undergoing esthetic surgery procedures.
Collapse
|
45
|
Luo S, Zhang X, Xiao X, Luo W, Yang Z, Tang S, Huang W. Exploring Potential Biomarkers and Molecular Mechanisms of Ischemic Cardiomyopathy and COVID-19 Comorbidity Based on Bioinformatics and Systems Biology. Int J Mol Sci 2023; 24:ijms24076511. [PMID: 37047484 PMCID: PMC10094917 DOI: 10.3390/ijms24076511] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Cardiovascular complications combined with COVID-19 (SARS-CoV-2) lead to a poor prognosis in patients. The common pathogenesis of ischemic cardiomyopathy (ICM) and COVID-19 is still unclear. Here, we explored potential molecular mechanisms and biomarkers for ICM and COVID-19. Common differentially expressed genes (DEGs) of ICM (GSE5406) and COVID-19 (GSE164805) were identified using GEO2R. We performed enrichment and protein–protein interaction analyses and screened key genes. To confirm the diagnostic performance for these hub genes, we used external datasets (GSE116250 and GSE211979) and plotted ROC curves. Transcription factor and microRNA regulatory networks were constructed for the validated hub genes. Finally, drug prediction and molecular docking validation were performed using cMAP. We identified 81 common DEGs, many of which were enriched in terms of their relation to angiogenesis. Three DEGs were identified as key hub genes (HSP90AA1, HSPA9, and SRSF1) in the protein–protein interaction analysis. These hub genes had high diagnostic performance in the four datasets (AUC > 0.7). Mir-16-5p and KLF9 transcription factor co-regulated these hub genes. The drugs vindesine and ON-01910 showed good binding performance to the hub genes. We identified HSP90AA1, HSPA9, and SRSF1 as markers for the co-pathogenesis of ICM and COVID-19, and showed that co-pathogenesis of ICM and COVID-19 may be related to angiogenesis. Vindesine and ON-01910 were predicted as potential therapeutic agents. Our findings will contribute to a deeper understanding of the comorbidity of ICM with COVID-19.
Collapse
|
46
|
Cong Y, Mucker EM, Perry DL, Dixit S, Kollins E, Byrum R, Huzella L, Kim R, Josleyn M, Kwilas S, Stefan C, Shoemaker CJ, Koehler J, Coyne S, Delp K, Liang J, Drawbaugh D, Hischak A, Hart R, Postnikova E, Vaughan N, Asher J, St Claire M, Hanson J, Schmaljohn C, Eakin AE, Hooper JW, Holbrook MR. Evaluation of a panel of therapeutic antibody clinical candidates for efficacy against SARS-CoV-2 in Syrian hamsters. Antiviral Res 2023; 213:105589. [PMID: 37003305 PMCID: PMC10060192 DOI: 10.1016/j.antiviral.2023.105589] [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: 02/12/2023] [Revised: 03/22/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
The COVID-19 pandemic spurred the rapid development of a range of therapeutic antibody treatments. As part of the US government's COVID-19 therapeutic response, a research team was assembled to support assay and animal model development to assess activity for therapeutics candidates against SARS-CoV-2. Candidate treatments included monoclonal antibodies, antibody cocktails, and products derived from blood donated by convalescent patients. Sixteen candidate antibody products were obtained directly from manufacturers and evaluated for neutralization activity against the WA-01 isolate of SARS-CoV-2. Products were further tested in the Syrian hamster model using prophylactic (-24 h) or therapeutic (+8 h) treatment approaches relative to intranasal SARS-CoV-2 exposure. In vivo assessments included daily clinical scores and body weights. Viral RNA and viable virus titers were quantified in serum and lung tissue with histopathology performed at 3d and 7d post-virus-exposure. Sham-treated, virus-exposed hamsters showed consistent clinical signs with concomitant weight loss and had detectable viral RNA and viable virus in lung tissue. Histopathologically, interstitial pneumonia with consolidation was present. Therapeutic efficacy was identified in treated hamsters by the absence or diminution of clinical scores, body weight loss, viral loads, and improved semiquantitative lung histopathology scores. This work serves as a model for the rapid, systematic in vitro and in vivo assessment of the efficacy of candidate therapeutics at various stages of clinical development. These efforts provided preclinical efficacy data for therapeutic candidates. Furthermore, these studies were invaluable for the phenotypic characterization of SARS CoV-2 disease in hamsters and of utility to the broader scientific community.
Collapse
Affiliation(s)
- Yu Cong
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Eric M Mucker
- United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, Frederick, MD, 21702, USA
| | - Donna L Perry
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Saurabh Dixit
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Erin Kollins
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Russ Byrum
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Louis Huzella
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Robert Kim
- United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, Frederick, MD, 21702, USA
| | - Mathew Josleyn
- United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, Frederick, MD, 21702, USA
| | - Steven Kwilas
- United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, Frederick, MD, 21702, USA
| | - Christopher Stefan
- United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, Frederick, MD, 21702, USA
| | - Charles J Shoemaker
- United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, Frederick, MD, 21702, USA
| | - Jeff Koehler
- United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, Frederick, MD, 21702, USA
| | - Susan Coyne
- United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, Frederick, MD, 21702, USA
| | - Korey Delp
- United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, Frederick, MD, 21702, USA
| | - Janie Liang
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - David Drawbaugh
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Amanda Hischak
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Randy Hart
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Elena Postnikova
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Nick Vaughan
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Jason Asher
- Leidos Supporting Department of Health and Human Services, Biomedical Advanced Research and Development Authority, Washington, DC, 20024, USA
| | - Marisa St Claire
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Jarod Hanson
- United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, Frederick, MD, 21702, USA
| | - Connie Schmaljohn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA
| | - Ann E Eakin
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Jay W Hooper
- United States Army Medical Research Institute of Infectious Diseases, Ft. Detrick, Frederick, MD, 21702, USA
| | - Michael R Holbrook
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, MD, 21702, USA.
| |
Collapse
|
47
|
Wang T, Zhai Y, Xue H, Zhou W, Ding Y, Nie H. Regulation of Epithelial Sodium Transport by SARS-CoV-2 Is Closely Related with Fibrinolytic System-Associated Proteins. Biomolecules 2023; 13:biom13040578. [PMID: 37189326 DOI: 10.3390/biom13040578] [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: 02/14/2023] [Revised: 03/08/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023] Open
Abstract
Dyspnea and progressive hypoxemia are the main clinical features of patients with coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Pulmonary pathology shows diffuse alveolar damage with edema, hemorrhage, and the deposition of fibrinogens in the alveolar space, which are consistent with the Berlin Acute Respiratory Distress Syndrome Criteria. The epithelial sodium channel (ENaC) is a key channel protein in alveolar ion transport and the rate-limiting step for pulmonary edema fluid clearance, the dysregulation of which is associated with acute lung injury/acute respiratory distress syndrome. The main protein of the fibrinolysis system, plasmin, can bind to the furin site of γ-ENaC and induce it to an activation state, facilitating pulmonary fluid reabsorption. Intriguingly, the unique feature of SARS-CoV-2 from other β-coronaviruses is that the spike protein of the former has the same furin site (RRAR) with ENaC, suggesting that a potential competition exists between SARS-CoV-2 and ENaC for the cleavage by plasmin. Extensive pulmonary microthrombosis caused by disorders of the coagulation and fibrinolysis system has also been seen in COVID-19 patients. To some extent, high plasmin (ogen) is a common risk factor for SARS-CoV-2 infection since an increased cleavage by plasmin accelerates virus invasion. This review elaborates on the closely related relationship between SARS-CoV-2 and ENaC for fibrinolysis system-related proteins, aiming to clarify the regulation of ENaC under SARS-CoV-2 infection and provide a novel reference for the treatment of COVID-19 from the view of sodium transport regulation in the lung epithelium.
Collapse
Affiliation(s)
- Tingyu Wang
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yiman Zhai
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Hao Xue
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Wei Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang 110122, China
| |
Collapse
|
48
|
Septimiu-Radu S, Gadela T, Gabriela D, Oancea C, Rosca O, Lazureanu VE, Fericean RM, Bratosin F, Dumitrescu A, Stoicescu ER, Bagiu I, Murariu M, Mavrea A. A Systematic Review of Lung Autopsy Findings in Elderly Patients after SARS-CoV-2 Infection. J Clin Med 2023; 12:2070. [PMID: 36902856 PMCID: PMC10004532 DOI: 10.3390/jcm12052070] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/27/2023] [Accepted: 03/05/2023] [Indexed: 03/08/2023] Open
Abstract
Although COVID-19 may cause various and multiorgan diseases, few research studies have examined the postmortem pathological findings of SARS-CoV-2-infected individuals who died. Active autopsy results may be crucial for understanding how COVID-19 infection operates and preventing severe effects. In contrast to younger persons, however, the patient's age, lifestyle, and concomitant comorbidities might alter the morpho-pathological aspects of the damaged lungs. Through a systematic analysis of the available literature until December 2022, we aimed to provide a thorough picture of the histopathological characteristics of the lungs in patients older than 70 years who died of COVID-19. A thorough search was conducted on three electronic databases (PubMed, Scopus, and Web of Science), including 18 studies and a total of 478 autopsies performed. It was observed that the average age of patients was 75.6 years, of which 65.4% were men. COPD was identified in an average of 16.7% of all patients. Autopsy findings indicated significantly heavier lungs, with an average weight of the right lung of 1103 g, while the left lung mass had an average weight of 848 g. Diffuse alveolar damage was a main finding in 67.2% of all autopsies, while pulmonary edema had a prevalence of between 50% and 70%. Thrombosis was also a significant finding, while some studies described focal and extensive pulmonary infarctions in 72.7% of elderly patients. Pneumonia and bronchopneumonia were observed, with a prevalence ranging from 47.6% to 89.5%. Other important findings described in less detail comprise hyaline membranes, the proliferation of pneumocytes and fibroblasts, extensive suppurative bronchopneumonic infiltrates, intra-alveolar edema, thickened alveolar septa, desquamation of pneumocytes, alveolar infiltrates, multinucleated giant cells, and intranuclear inclusion bodies. These findings should be corroborated with children's and adults' autopsies. Postmortem examination as a technique for studying the microscopic and macroscopic features of the lungs might lead to a better knowledge of COVID-19 pathogenesis, diagnosis, and treatment, hence enhancing elderly patient care.
Collapse
Affiliation(s)
- Susa Septimiu-Radu
- Department XIII, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Doctoral School, ‘’Victor Babes’’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Tejaswi Gadela
- School of General Medicine, Bhaskar Medical College, Amdapur Road 156-162, Hyderabad 500075, India
| | - Doros Gabriela
- Department of Pediatrics, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Cristian Oancea
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Ovidiu Rosca
- Department XIII, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Voichita Elena Lazureanu
- Department XIII, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Roxana Manuela Fericean
- Department XIII, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Doctoral School, ‘’Victor Babes’’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Felix Bratosin
- Department XIII, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Doctoral School, ‘’Victor Babes’’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Andreea Dumitrescu
- Cardioprevent Foundation, Calea Dorobantilor 3, Timisoara 300134, Romania
| | - Emil Robert Stoicescu
- Doctoral School, ‘’Victor Babes’’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Department of Radiology and Medical Imaging, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Iulia Bagiu
- Department of Microbiology, Multidisciplinary Research Center on Antimicrobial Resistance, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Mircea Murariu
- Doctoral School, ‘’Victor Babes’’ University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Department of Pediatrics, Discipline of Infectious Disease, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Adelina Mavrea
- Department of Internal Medicine I, Cardiology Clinic, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| |
Collapse
|
49
|
Yang C, Cai L, Xiao SY. Pathologic Characteristics of Digestive Tract and Liver in Patients with Coronavirus Disease 2019. Gastroenterol Clin North Am 2023; 52:201-214. [PMID: 36813426 PMCID: PMC9531645 DOI: 10.1016/j.gtc.2022.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
With the high prevalence of coronavirus disease-2019 (COVID-19), there has been increasing understanding of the pathologic changes associated with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This review summarizes the pathologic changes in the digestive system and liver associated with COVID-19, including the injuries induced by SARS-CoV2 infection of GI epithelial cells and the systemic immune responses. The common digestive manifestations associated with COVID-19 include anorexia, nausea, vomiting, and diarrhea; the clearance of the viruses in COVID-19 patients with digestive symptoms is usually delayed. COVID-19-associated gastrointestinal histopathology is characterized by mucosal damage and lymphocytic infiltration. The most common hepatic changes are steatosis, mild lobular and portal inflammation, congestion/sinusoidal dilatation, lobular necrosis, and cholestasis.
Collapse
Affiliation(s)
- Chunxiu Yang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lijun Cai
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shu-Yuan Xiao
- Department of Pathology, University of Chicago Medicine, University of Chicago Medicine, MC6101, Anatomic Pathology, 5841 South Maryland Avenue, Chicago, IL 60637, USA.
| |
Collapse
|
50
|
Abdaljaleel M, Tawalbeh I, Sallam M, Hani AB, Al-Abdallat IM, Omari BA, Al-Mustafa S, Abder-Rahman H, Abbas AS, Zureigat M, Al-Abbadi MA. Postmortem lung and heart examination of COVID-19 patients in a case series from Jordan. J Pathol Transl Med 2023; 57:102-112. [PMID: 36950812 PMCID: PMC10028009 DOI: 10.4132/jptm.2023.01.30] [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: 11/27/2022] [Accepted: 01/30/2023] [Indexed: 03/24/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has emerged as a pandemic for more than 2 years. Autopsy examination is an invaluable tool to understand the pathogenesis of emerging infections and their consequent mortalities. The aim of the current study was to present the lung and heart pathological findings of COVID-19-positive autopsies performed in Jordan. METHODS The study involved medicolegal cases, where the cause of death was unclear and autopsy examination was mandated by law. We included the clinical and pathologic findings of routine gross and microscopic examination of cases that were positive for COVID-19 at time of death. Testing for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was confirmed through molecular detection by real-time polymerase chain reaction, serologic testing for IgM and electron microscope examination of lung samples. RESULTS Seventeen autopsies were included, with male predominance (76.5%), Jordanians (70.6%), and 50 years as the mean age at time of death. Nine out of 16 cases (56.3%) had co-morbidities, with one case lacking such data. Histologic examination of lung tissue revealed diffuse alveolar damage in 13/17 cases (76.5%), and pulmonary microthrombi in 8/17 cases (47.1%). Microscopic cardiac findings were scarcely detected. Two patients died as a direct result of acute cardiac disease with limited pulmonary findings. CONCLUSIONS The detection of SARS-CoV-2 in postmortem examination can be an incidental or contributory finding which highlights the value of autopsy examination to determine the exact cause of death in controversial cases.
Collapse
Affiliation(s)
- Maram Abdaljaleel
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| | - Isra Tawalbeh
- Department of Forensic Pathology, Ministry of Health, Amman, Jordan
| | - Malik Sallam
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
- Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Amjad Bani Hani
- Department of General Surgery, School of Medicine, The University of Jordan, Amman, Jordan
| | - Imad M Al-Abdallat
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| | - Baheth Al Omari
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| | - Sahar Al-Mustafa
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| | - Hasan Abder-Rahman
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| | - Adnan Said Abbas
- Department of Forensic Pathology, Ministry of Health, Amman, Jordan
| | - Mahmoud Zureigat
- Department of Forensic Pathology, Ministry of Health, Amman, Jordan
| | - Mousa A Al-Abbadi
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| |
Collapse
|