1
|
Sakai A, Singh G, Khoshbakht M, Bittner S, Löhr CV, Diaz-Tapia R, Warang P, White K, Luo LL, Tolbert B, Blanco M, Chow A, Guttman M, Li C, Bao Y, Ho J, Maurer-Stroh S, Chatterjee A, Chanda S, García-Sastre A, Schotsaert M, Teijaro JR, Moulton HM, Stein DA. Inhibition of SARS-CoV-2 growth in the lungs of mice by a peptide-conjugated morpholino oligomer targeting viral RNA. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102331. [PMID: 39376996 PMCID: PMC11456799 DOI: 10.1016/j.omtn.2024.102331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024]
Abstract
Further development of direct-acting antiviral agents against human SARS-CoV-2 infections remains a public health priority. Here, we report that an antisense peptide-conjugated morpholino oligomer (PPMO) named 5'END-2, targeting a highly conserved sequence in the 5' UTR of SARS-CoV-2 genomic RNA, potently suppressed SARS-CoV-2 growth in vitro and in vivo. In HeLa-ACE 2 cells, 5'END-2 produced IC50 values of between 40 nM and 1.15 μM in challenges using six genetically disparate strains of SARS-CoV-2, including JN.1. In vivo, using K18-hACE2 mice and the WA-1/2020 virus isolate, two doses of 5'END-2 at 10 mg/kg, administered intranasally on the day before and the day after infection, produced approximately 1.4 log10 virus titer reduction in lung tissue at 3 days post-infection. Under a similar dosing schedule, intratracheal administration of 1.0-2.0 mg/kg 5'END-2 produced over 3.5 log10 virus growth suppression in mouse lungs. Electrophoretic mobility shift assays characterized specific binding of 5'END-2 to its complementary target RNA. Furthermore, using reporter constructs containing SARS-CoV-2 5' UTR leader sequence, in an in-cell system, we observed that 5'END-2 could interfere with translation in a sequence-specific manner. The results demonstrate that direct pulmonary delivery of 5'END-2 PPMO is a promising antiviral strategy against SARS-CoV-2 infections and warrants further development.
Collapse
Affiliation(s)
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mahsa Khoshbakht
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Scott Bittner
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Christiane V. Löhr
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - Randy Diaz-Tapia
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kris White
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Luke Le Luo
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Blanton Tolbert
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mario Blanco
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | - Amy Chow
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | - Mitchell Guttman
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA
| | - Cuiping Li
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
| | - Yiming Bao
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Joses Ho
- GISAID @ A∗STAR Bioinformatics Institute, Singapore 138632, Singapore
| | | | | | - Sumit Chanda
- Scripps Research Institute, La Jolla, CA 92037, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Molecular, and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Hong M. Moulton
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| | - David A. Stein
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
2
|
Kirk NM, Liang Y, Ly H. Pathogenesis and virulence of coronavirus disease: Comparative pathology of animal models for COVID-19. Virulence 2024; 15:2316438. [PMID: 38362881 PMCID: PMC10878030 DOI: 10.1080/21505594.2024.2316438] [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: 10/20/2023] [Accepted: 02/04/2024] [Indexed: 02/17/2024] Open
Abstract
Animal models that can replicate clinical and pathologic features of severe human coronavirus infections have been instrumental in the development of novel vaccines and therapeutics. The goal of this review is to summarize our current understanding of the pathogenesis of coronavirus disease 2019 (COVID-19) and the pathologic features that can be observed in several currently available animal models. Knowledge gained from studying these animal models of SARS-CoV-2 infection can help inform appropriate model selection for disease modelling as well as for vaccine and therapeutic developments.
Collapse
Affiliation(s)
- Natalie M. Kirk
- Department of Veterinary & Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, MN, USA
| | - Yuying Liang
- Department of Veterinary & Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, MN, USA
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Twin Cities, MN, USA
| |
Collapse
|
3
|
Lu J, Zuo X, Cai A, Xiao F, Xu Z, Wang R, Miao C, Yang C, Zheng X, Wang J, Ding X, Xiong W. Cerebral small vessel injury in mice with damage to ACE2-expressing cerebral vascular endothelial cells and post COVID-19 patients. Alzheimers Dement 2024. [PMID: 39352003 DOI: 10.1002/alz.14279] [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: 02/26/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 10/03/2024]
Abstract
INTRODUCTION The angiotensin-converting enzyme 2 (ACE2), which is expressed in cerebral vascular endothelial cells (CVECs), has been currently identified as a functional receptor for SARS-CoV-2. METHODS We specifically induced injury to ACE2-expressing CVECs in mice and evaluated the effects of such targeted damage through magnetic resonance imaging (MRI) and cognitive behavioral tests. In parallel, we recruited a single-center cohort of COVID-19 survivors and further assessed their brain microvascular injury based on cognition and emotional scales, cranial MRI scans, and blood proteomic measurements. RESULTS Here, we show an array of pathological and behavioral alterations characteristic of cerebral small vessel disease (CSVD) in mice that targeted damage to ACE2-expressing CVECs, and COVID-19 survivors. These CSVD-like manifestations persist for at least 7 months post-recovery from COVID-19. DISCUSSION Our findings suggest that SARS-CoV-2 may induce cerebral small vessel damage with persistent sequelae, underscoring the imperative for heightened clinical vigilance in mitigating or treating SARS-CoV-2-mediated cerebral endothelial injury throughout infection and convalescence. HIGHLIGHTS Cerebral small vessel disease-associated changes were observed after targeted damage to angiotensin-converting enzyme 2-expressing cerebral vascular endothelial cells. SARS-CoV-2 may induce cerebral small vessel damage with persistent sequelae. Clinical vigilance is needed in preventing SARS-CoV-2-induced cerebral endothelial damage during infection and recovery.
Collapse
Affiliation(s)
- Jieping Lu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xin Zuo
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
| | - Aoling Cai
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, China
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Fang Xiao
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhenyu Xu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Rui Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chenjian Miao
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chen Yang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xingxing Zheng
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoling Ding
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wei Xiong
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, China
- CAS Key Laboratory of Brain Function and Disease, Hefei, China
| |
Collapse
|
4
|
Jia Y, Tian W, Li Y, Teng Y, Liu X, Li Z, Zhao M. Chloroquine: Rapidly withdrawing from first-line treatment of COVID-19. Heliyon 2024; 10:e37098. [PMID: 39281655 PMCID: PMC11402237 DOI: 10.1016/j.heliyon.2024.e37098] [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: 08/16/2023] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/18/2024] Open
Abstract
The COVID-19 outbreak has garnered significant global attention due to its impact on human health. Despite its relatively low fatality rate, the virus affects multiple organ systems, resulting in various symptoms such as palpitations, headaches, muscle pain, and hearing loss among COVID-19 patients and those recovering from the disease. These symptoms impose a substantial physical, psychological, and social burden on affected individuals. On February 15, 2020, the Chinese government advised incorporating antimalarial drugs into the guidelines issued by the National Health Commission of China for preventing, diagnosing, and treating COVID-19 pneumonia. We examine the adverse effects of Chloroquine (CQ) in treating COVID-19 complications to understand why it is no longer the primary treatment for the disease.
Collapse
Affiliation(s)
- Yunlong Jia
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Wenjie Tian
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Yuyao Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Yuyan Teng
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Xiaolin Liu
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Zhengyu Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Mingsheng Zhao
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| |
Collapse
|
5
|
Evering TH, Moser C, Jilg N, Ritz J, Wohl DA, Li JZ, Margolis D, Javan AC, Eron JJ, Currier JS, Daar ES, Smith DM, Hughes MD, Chew KW. Post-acute COVID-19 outcomes including participant-reported long COVID: amubarvimab/romlusevimab versus placebo in the ACTIV-2 trial. EClinicalMedicine 2024; 75:102787. [PMID: 39252866 PMCID: PMC11381616 DOI: 10.1016/j.eclinm.2024.102787] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/11/2024] Open
Abstract
Background It is unknown if early COVID-19 monoclonal antibody (mAb) therapy can reduce risk of Long COVID. The mAbs amubarvimab/romlusevimab were previously demonstrated to reduce risk of hospitalization/death by 79%. This study assessed the impact of amubarvimab/romlusevimab on late outcomes, including Long COVID. Methods Non-hospitalized high-risk adults within 10 days of COVID-19 symptom onset enrolled in a randomized, double-blind, placebo-controlled phase 2/3 trial of amubarvimab/romlusevimab for COVID-19 treatment. Late symptoms, assessed using a participant-completed symptom diary, were a pre-specified exploratory endpoint. The primary outcome for this analysis was the composite of Long COVID by participant self-report (presence of COVID-19 symptoms as recorded in the diary at week 36) or hospitalization or death by week 36. Inverse probability weighting (IPW) was used to address incomplete outcome ascertainment, giving weighted risk ratios (wRR) comparing amubarvimab/romlusevimab to placebo. Findings Participants received amubarvimab/romlusevimab (n = 390) or placebo (n = 390) between January and July 2021. Median age was 49 years, 52% were female, 18% Black/African American, 49% Hispanic/Latino, and 9% COVID-19-vaccinated at entry. At week 36, 103 (13%) had incomplete outcome ascertainment, and 66 (17%) on amubarvimab/romlusevimab and 92 (24%) on placebo met the primary outcome (wRR = 0.70, 95% confidence interval (CI) 0.53-0.93). The difference was driven by fewer hospitalizations/deaths with amubarvimab/romlusevimab (4%) than placebo (13%). Among 652 participants with available diary responses, 53 (16%) on amubarvimab/romlusevimab and 44 (14%) on placebo reported presence of Long COVID. Interpretation Amubarvimab/romlusevimab treatment, while highly effective in preventing hospitalizations/deaths, did not reduce risk of Long COVID. Additional interventions are needed to prevent Long COVID. Funding National Institute of Allergy and Infectious Diseases of the National Institutes of Health. Amubarvimab and romlusevimab supplied by Brii Biosciences.
Collapse
Affiliation(s)
| | - Carlee Moser
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Nikolaus Jilg
- Massachusetts General Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Justin Ritz
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Jonathan Z. Li
- Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - Eric S. Daar
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | | | | | - Kara W. Chew
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
6
|
Subramanian G, Hage A, Feldmann F, Chiramel AI, McNally KL, Sturdevant GL, Beare PA, Best SM. AP3B1 Has Type I Interferon-Independent Antiviral Function against SARS-CoV-2. Viruses 2024; 16:1377. [PMID: 39339853 PMCID: PMC11437497 DOI: 10.3390/v16091377] [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: 06/01/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
The unprecedented research effort associated with the emergence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) included several extensive proteomic studies that identified host proteins that interact with individual viral gene products. However, in most cases, the consequences of those virus-host interactions for virus replication were not experimentally pursued, which is a necessary step in determining whether the interactions represent pro- or anti-viral events. One putative interaction commonly identified in multiple studies was between the host adaptor protein complex 3 (AP-3) subunit B1 (AP3B1) and the SARS-CoV-2 envelope protein (E). AP3B1 is one subunit of AP-3 required for the biogenesis of lysosomal-related organelles (LROs), and its function impacts important disease processes including inflammation and vascular health. Thus, interactions between AP3B1 and SARS-CoV-2 might influence the clinical outcomes of infection. To determine if AP3B1 has a role in the SARS-CoV-2 replication cycle, we first confirmed the interaction in virus-infected cells using immunoprecipitation (IP) and immunofluorescence assays (IFA). AP3B1 is required by multiple viruses to aid in the replication cycle and therefore may be a therapeutic target. However, we found that the overexpression of AP3B1 suppressed SARS-CoV-2 replication, whereas the siRNA-mediated depletion of AP3B1 increased the release of infectious virus, suggesting an antiviral role for AP3B1. Together, our findings suggest that AP3B1 is an intrinsic barrier to SARS-CoV-2 replication through interactions with the viral E protein. Our work justifies further investigations of LRO trafficking in SARS-CoV-2 target cells and their role in viral pathogenesis.
Collapse
Affiliation(s)
- Gayatri Subramanian
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Adam Hage
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Abhilash I Chiramel
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Kristin L McNally
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Gail L Sturdevant
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Paul A Beare
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Sonja M Best
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| |
Collapse
|
7
|
Wang C, Wang S, Ma X, Yao X, Zhan K, Wang Z, He D, Zuo W, Han S, Zhao G, Cao B, Zhao J, Bian X, Wang J. P-selectin Facilitates SARS-CoV-2 Spike 1 Subunit Attachment to Vesicular Endothelium and Platelets. ACS Infect Dis 2024; 10:2656-2667. [PMID: 38912949 DOI: 10.1021/acsinfecdis.3c00728] [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] [Indexed: 06/25/2024]
Abstract
SARS-CoV-2 infection starts from the association of its spike 1 (S1) subunit with sensitive cells. Vesicular endothelial cells and platelets are among the cell types that bind SARS-CoV-2, but the effectors that mediate viral attachment on the cell membrane have not been fully elucidated. Herein, we show that P-selectin (SELP), a biomarker for endothelial dysfunction and platelet activation, can facilitate the attachment of SARS-CoV-2 S1. Since we observe colocalization of SELP with S1 in the lung tissues of COVID-19 patients, we perform molecular biology experiments on human umbilical vein endothelial cells (HUVECs) to confirm the intermolecular interaction between SELP and S1. SELP overexpression increases S1 recruitment to HUVECs and enhances SARS-CoV-2 spike pseudovirion infection. The opposite results are determined after SELP downregulation. As S1 causes endothelial inflammatory responses in a dose-dependent manner, by activating the interleukin (IL)-17 signaling pathway, SELP-induced S1 recruitment may contribute to the development of a "cytokine storm" after viral infection. Furthermore, SELP also promotes the attachment of S1 to the platelet membrane. Employment of PSI-697, a small inhibitor of SELP, markedly decreases S1 adhesion to both HUVECs and platelets. In addition to the role of membrane SELP in facilitating S1 attachment, we also discover that soluble SELP is a prognostic factor for severe COVID-19 through a meta-analysis. In this study, we identify SELP as an adhesive site for the SARS-CoV-2 S1, thus providing a potential drug target for COVID-19 treatment.
Collapse
Affiliation(s)
- Cheng Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shaobo Wang
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Xiangyu Ma
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaohong Yao
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kegang Zhan
- Department of Epidemiology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, China
| | - Di He
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing 100069, China
| | - Wenting Zuo
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Songling Han
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Gaomei Zhao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bin Cao
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, Capital Medical University, Beijing 100069, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing 100084, China
- Changping Laboratory, Beijing 102206, China
- New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing 100029, China
| | - Jinghong Zhao
- Department of Nephrology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Xiuwu Bian
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
8
|
McClelland AC, Benitez SJ, Burns J. COVID-19 Neuroimaging Update: Pathophysiology, Acute Findings, and Post-Acute Developments. Semin Ultrasound CT MR 2024; 45:318-331. [PMID: 38518814 DOI: 10.1053/j.sult.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
COVID-19 has prominent effects on the nervous system with important manifestations on neuroimaging. In this review, we discuss the neuroimaging appearance of acute COVID-19 that became evident during the early stages of the pandemic. We highlight the underlying pathophysiology mediating nervous system effects and neuroimaging appearances including systemic inflammatory response such as cytokine storm, coagulopathy, and para/post-infections immune mediated phenomena. We also discuss the nervous system manifestations of COVID-19 and the role of imaging as the pandemic has evolved over time, including related to the development of vaccines and the emergence of post-acute sequalae such as long COVID.
Collapse
Affiliation(s)
| | - Steven J Benitez
- Department of Radiology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY
| | - Judah Burns
- Department of Radiology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
9
|
Marcos-Neira P, Morales-Indiano C, Fernández-Caballero M, Tomasa-Irriguible T, Bordejé-Laguna L, Ruíz-Artola V. Anti-Xa activity below range is related to thrombosis in patients with severe COVID-19. Med Intensiva 2024:S2173-5727(24)00191-7. [PMID: 39054216 DOI: 10.1016/j.medine.2024.07.003] [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: 04/16/2024] [Revised: 06/05/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE We aimed to anlayse the relationship between anti-Xa activity below range and thomboembolic events. DESIGN Single center prospective observational longitudinal cohort study (February-November 2021). SETTING Patients admitted to the ICU of a University Hospital. PARTICIPANTS Patients with severe COVID-19 pneumoniae. INTERVENTIONS Enoxaparin was used for prophylactic and therapeutic anticoagulation. Enoxaparin dosing and dose adjustment were based on anti-Xa activity according to the hospital protocol. MAIN VARIABLES OF INTEREST Target: thomboembolic events. PREDICTORS demographics, pharmacotherapy, anti-Xa measurements, clinical data, and laboratory results. Logistic regression was used to identify independent risk factors for thomboembolic events. RESULTS Data were available for 896 serum anti-Xa measurements from 228 subjects. Overall, 71.9% were male, with a median age of 62. Most patients needed invasive mechanical ventilation (87.7%) and mortality was 24.1%. A total of 28.9% new thomboembolic events were diagnosed. There were 27.1% anti-Xa measesurements below range. When multivariable logistic regression analysis was performed anti-Xa activity below range (RR, 4.2; p = 0.000), C-reactive protein (25 mg/L increase) (RR, 1.14; p = 0.005) and D-dimer (1000 ng/L increase) (RR, 1.06; p = 0.002) were the independent factors related to new thomboembolic events in patients with severe COVID-19. CONCLUSIONS Anti-Xa activity below range, C-reactive protein and D-dimer were the independent factors related to thomboembolic events in patients with severe COVID-19. Purposely designed clinical trials should be carried out to confirm the benefit of an anti-Xa monitoring.
Collapse
Affiliation(s)
- Pilar Marcos-Neira
- Intensive Care Unit, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain.
| | - Cristian Morales-Indiano
- Department of Clinical Analysis and Biochemistry, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain.
| | | | | | - Luisa Bordejé-Laguna
- Intensive Care Unit, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain.
| | - Víctor Ruíz-Artola
- Intensive Care Unit, Germans Trias i Pujol University Hospital, Badalona, Barcelona, Spain.
| |
Collapse
|
10
|
Sherman JD, Karmali V, Kumar B, Simon TW, Bechnak S, Panjwani A, Ciric CR, Wang D, Huerta C, Johnson B, Anderson EJ, Rouphael N, Collins MH, Rostad CA, Azadi P, Scherer EM. Altered spike IgG Fc N-linked glycans are associated with hyperinflammatory state in adult COVID and Multisystem Inflammatory Syndrome in Children. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.14.24310381. [PMID: 39040211 PMCID: PMC11261911 DOI: 10.1101/2024.07.14.24310381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Background Severe COVID and multisystem inflammatory syndrome (MIS-C) are characterized by excessive inflammatory cytokines/chemokines. In adults, disease severity is associated with SARS-CoV-2-specific IgG Fc afucosylation, which induces pro-inflammatory cytokine secretion from innate immune cells. This study aimed to define spike IgG Fc glycosylation following SARS-CoV-2 infection in adults and children and following SARS-CoV-2 vaccination in adults and the relationships between glycan modifications and cytokine/chemokine levels. Methods We analyzed longitudinal (n=146) and cross-sectional (n=49) serum/plasma samples from adult and pediatric COVID patients, MIS-C patients, adult vaccinees, and adult and pediatric healthy controls. We developed methods for characterizing bulk and spike IgG Fc glycosylation by capillary electrophoresis (CE) and measured levels of ten inflammatory cytokines/chemokines by multiplexed ELISA. Results Spike IgG were more afucosylated than bulk IgG during acute adult COVID and MIS-C. We observed an opposite trend following vaccination, but it was not significant. Spike IgG were more galactosylated and sialylated and less bisected than bulk IgG during adult COVID, with similar trends observed during pediatric COVID/MIS-C and following SARS-CoV-2 vaccination. Spike IgG glycosylation changed with time following adult COVID or vaccination. Afucosylated spike IgG exhibited inverse and positive correlations with inflammatory markers in MIS-C and following vaccination, respectively; galactosylated and sialylated spike IgG inversely correlated with pro-inflammatory cytokines in adult COVID and MIS-C; and bisected spike IgG positively correlated with inflammatory cytokines/chemokines in multiple groups. Conclusions We identified previously undescribed relationships between spike IgG glycan modifications and inflammatory cytokines/chemokines that expand our understanding of IgG glycosylation changes that may impact COVID and MIS-C immunopathology.
Collapse
Affiliation(s)
- Jacob D. Sherman
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Vinit Karmali
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Bhoj Kumar
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - Trevor W. Simon
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Sarah Bechnak
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Anusha Panjwani
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Caroline R. Ciric
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Dongli Wang
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Chris Huerta
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Brandi Johnson
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Evan J. Anderson
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Nadine Rouphael
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Matthew H. Collins
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Christina A. Rostad
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, 30602, USA
| | - Erin M. Scherer
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| |
Collapse
|
11
|
Gururangan K, Peschansky VJ, Van Hyfte G, Agarwal P, Blank LJ, Mathew B, Goldstein J, Kwon CS, McCarthy L, Cohen A, Chan AHW, Deng P, Dhamoon M, Gutzwiller E, Hao Q, He C, Klenofsky B, Lemus HN, Marcuse L, Navis A, Heredia Nunez WD, Luckey MN, Schorr EM, Singh A, Tantillo GB, Ufongene C, Young JJ, Balchandani P, Festa JR, Naasan G, Charney AW, Nadkarni GN, Jetté N. Neuropsychiatric complications of coronavirus disease 2019: Mount Sinai Health System cohort study. J Neurol 2024; 271:3991-4007. [PMID: 38656620 DOI: 10.1007/s00415-024-12370-9] [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: 01/06/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/26/2024]
Abstract
OBJECTIVE To describe the frequency of neuropsychiatric complications among hospitalized patients with coronavirus disease 2019 (COVID-19) and their association with pre-existing comorbidities and clinical outcomes. METHODS We retrospectively identified all patients hospitalized with COVID-19 within a large multicenter New York City health system between March 15, 2020 and May 17, 2021 and randomly selected a representative cohort for detailed chart review. Clinical data, including the occurrence of neuropsychiatric complications (categorized as either altered mental status [AMS] or other neuropsychiatric complications) and in-hospital mortality, were extracted using an electronic medical record database and individual chart review. Associations between neuropsychiatric complications, comorbidities, laboratory findings, and in-hospital mortality were assessed using multivariate logistic regression. RESULTS Our study cohort consisted of 974 patients, the majority were admitted during the first wave of the pandemic. Patients were treated with anticoagulation (88.4%), glucocorticoids (24.8%), and remdesivir (10.5%); 18.6% experienced severe COVID-19 pneumonia (evidenced by ventilator requirement). Neuropsychiatric complications occurred in 58.8% of patients; 39.8% experienced AMS; and 19.0% experienced at least one other complication (seizures in 1.4%, ischemic stroke in 1.6%, hemorrhagic stroke in 1.0%) or symptom (headache in 11.4%, anxiety in 6.8%, ataxia in 6.3%). Higher odds of mortality, which occurred in 22.0%, were associated with AMS, ventilator support, increasing age, and higher serum inflammatory marker levels. Anticoagulant therapy was associated with lower odds of mortality and AMS. CONCLUSION Neuropsychiatric complications of COVID-19, especially AMS, were common, varied, and associated with in-hospital mortality in a diverse multicenter cohort at an epicenter of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Kapil Gururangan
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Veronica J Peschansky
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Grace Van Hyfte
- Department of Population Health Science and Policy, Institute of Health Care Delivery Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Parul Agarwal
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Population Health Science and Policy, Institute of Health Care Delivery Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leah J Blank
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Population Health Science and Policy, Institute of Health Care Delivery Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian Mathew
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jonathan Goldstein
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Churl-Su Kwon
- Departments of Neurosurgery, Neurology, Epidemiology, and the Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
| | - Louise McCarthy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ariella Cohen
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andy Ho Wing Chan
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pojen Deng
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mandip Dhamoon
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eveline Gutzwiller
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qing Hao
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Celestine He
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Britany Klenofsky
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hernan Nicolas Lemus
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lara Marcuse
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Allison Navis
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Mallory N Luckey
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily M Schorr
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anuradha Singh
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriela B Tantillo
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Claire Ufongene
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James J Young
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Priti Balchandani
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joanne R Festa
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Barbara and Maurice Deane Center for Wellness and Cognitive Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georges Naasan
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Barbara and Maurice Deane Center for Wellness and Cognitive Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander W Charney
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish N Nadkarni
- Division of Data Driven and Digital Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nathalie Jetté
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Population Health Science and Policy, Institute of Health Care Delivery Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, 1403 29 Street NW, Calgary, AB, T2N 2T9, Canada.
| |
Collapse
|
12
|
Miksová L, Dytrych V, Ptáčník V, Balík M, Linhart A, Bělohlávek J, Jansa P. Pulmonary perfusion in long-term survivors of COVID-19-related severe acute respiratory distress syndrome treated by extracorporeal membrane oxygenation. Pulm Circ 2024; 14:e12431. [PMID: 39188535 PMCID: PMC11345203 DOI: 10.1002/pul2.12431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024] Open
Abstract
COVID-19 associates with a hypercoagulant state and an increased risk for venous thromboembolic events (VTEs). Whether severe COVID-19 infection requiring extracorporeal membrane oxygenation (ECMO) support might lead to chronic pulmonary perfusion abnormalities and chronic thromboembolic pulmonary disease/hypertension remains unclear. The purpose of this study was to evaluate chronic pulmonary perfusion abnormalities in long-term survivors of COVID-19-related severe acute respiratory distress syndrome (ARDS) treated by ECMO at our institution. Pulmonary perfusion was examined by ventilation/perfusion (V/Q) single-photon emission computed tomography or V/Q planar scintigraphy at least 3 months after ECMO explantation, comorbidities and incidence of thromboembolic events were recorded as well. Of 172 COVID-19 patients treated by ECMO for severe COVID-19 pneumonia between March 2020 and November 2021, only 80 were successfully weaned from ECMO. Of those, 37 patients were enrolled into the present analysis (27% female, mean age 52 years). Median duration of ECMO support was 12 days. In 24 (65%) patients VTE was recorded in the acute phase (23 patients developed ECMO cannula-related deep vein thrombosis, 5 of them had also a pulmonary embolism, and one thrombus was associated with a central catheter). The median duration between ECMO explantation and assessment of pulmonary perfusion was 420 days. No segmental or larger mismatched perfusion defects were then detected in any patient. In conclusion, in long-term survivors of COVID-19-related ARDS treated by ECMO, no persistent pulmonary perfusion abnormalities were detected although VTE was common.
Collapse
Affiliation(s)
- Lucie Miksová
- Clinical Department of Cardiology and Angiology of the 2nd Department of MedicineGeneral University Hospital in PraguePragueCzech Republic
| | - Vladimír Dytrych
- Clinical Department of Cardiology and Angiology of the 2nd Department of MedicineGeneral University Hospital in PraguePragueCzech Republic
| | - Václav Ptáčník
- Institute of Nuclear Medicine of the 1st Faculty of Medicine and General University Hospital in PraguePragueCzech Republic
| | - Martin Balík
- Department of Anesthesiology and Intensive Care of the 1st Faculty of Medicine and General University Hospital in PraguePragueCzech Republic
| | - Aleš Linhart
- Clinical Department of Cardiology and Angiology of the 2nd Department of MedicineGeneral University Hospital in PraguePragueCzech Republic
| | - Jan Bělohlávek
- Clinical Department of Cardiology and Angiology of the 2nd Department of MedicineGeneral University Hospital in PraguePragueCzech Republic
| | - Pavel Jansa
- Clinical Department of Cardiology and Angiology of the 2nd Department of MedicineGeneral University Hospital in PraguePragueCzech Republic
| |
Collapse
|
13
|
Sideratou CM, Papaneophytou C. Persistent Vascular Complications in Long COVID: The Role of ACE2 Deactivation, Microclots, and Uniform Fibrosis. Infect Dis Rep 2024; 16:561-571. [PMID: 39051242 PMCID: PMC11270324 DOI: 10.3390/idr16040042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2), a key regulator in vasoregulation and the renin-angiotensin system, is hypothesized to be downregulated in patients with COVID-19, leading to a cascade of cardiovascular complications. This deactivation potentially results in increased blood pressure and vessel injury, contributing to the formation and persistence of microclots in the circulation. Herein, we propose a hypothesis regarding the prolonged vascular complications observed in long COVID, focusing on the role of ACE2 deactivation and/or shedding, the persistence of microclots, and the unique pattern of fibrosis induced by severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). Furthermore, we propose that the distinctive, uniform fibrosis associated with COVID-19, which is challenging to detect through conventional X-ray imaging, exacerbates vascular injury and impairs oxygenation. The persistence of these microclots and the unique fibrosis pattern are suggested as key factors in the extended duration of vascular complications post-COVID-19 infection, regardless of the initial disease severity. Moreover, plasma ACE2 activity has the potential to serve as prognostic or diagnostic biomarkers for monitoring disease severity and managing long COVID symptoms. Elucidating the role of ACE2 deactivation and the consequent events is vital for understanding the long-term effects of COVID-19. The experimental verification of this hypothesis through in vitro studies, clinical longitudinal studies, and advanced imaging techniques could yield significant insights into the pathophysiological mechanisms underlying long COVID, thereby improving the management of patients, particularly those with cardiovascular complications.
Collapse
Affiliation(s)
| | - Christos Papaneophytou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus;
| |
Collapse
|
14
|
Anzalone N, Gerevini S, Del Poggio A, Gaudino S, Causin F, Politi LS, Triulzi FM, Pero G, Pichiecchio A, Bastianello S, Baruzzi FM, Bianchini E, Foti G, Ricciardi GK, Sponza M, Menozzi R, Cosottini M, Chirico PD, Saba L, Gasparotti R. Neuroradiological manifestations in hospitalized patients with COVID-19: An Italian national multicenter study on behalf of AINR (Associazione Italiana di Neuroradiologia) and SIRM (Società Italiana di Radiologia Medica). Neuroradiol J 2024:19714009241240312. [PMID: 38897216 DOI: 10.1177/19714009241240312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024] Open
Abstract
PURPOSE This multicentric study aims to characterize and assess the occurrence of neuroradiological findings among patients with SARS-CoV-2 infection during the first Italian wave of the pandemic outbreak. MATERIALS AND METHODS Patients' data were collected between May 2020 and June 2020. Clinical and laboratory data, chest imaging, brain CT, and MRI imaging were included. Acquired data were centralized and analyzed in two hospitals: ASST Spedali Civili, Brescia, and IRRCS San Raffaele Research Hospital, Milan, Italy. COVID-19 patients were classified into two different subgroups, vascular and nonvascular. The vascular pattern was further divided into ischemic and hemorrhagic stroke groups. RESULTS Four hundred and fifteen patients from 20 different Italian Centers were enrolled in the study. The most frequent symptom was focal neurological deficit, found in 143 patients (34.5%). The most frequent neuroradiological finding was ischemic stroke in 122 (29.4%) patients. Forty-four (10.6%) patients presented a cerebral hemorrhage. Forty-seven patients had non-stroke neuroimaging lesions (11.3%). The most common was PRES-like syndrome (28%), SWI hypointensities (22%), and encephalitis (19%). The stroke group had higher CAD risk (37.5% vs 20%, p = .016) and higher D-dimer levels (1875 ng/mL vs 451 ng/mL, p < .001) compared to the negative group. CONCLUSION Our study describes the biggest cohort study in Italy on brain imaging of COVID-19 patients and confirms that COVID-19 patients are at risk of strokes, possibly due to a pro-thrombotic microenvironment. Moreover, apart from stroke, the other neuroradiological patterns described align with the ones reported worldwide.
Collapse
Affiliation(s)
- Nicoletta Anzalone
- Neuroradiology Department, IRCCS San Raffaele Scientific Institute, Italy
- Vita-Salute San Raffaele University, Italy
| | | | - Anna Del Poggio
- Neuroradiology Department, IRCCS San Raffaele Scientific Institute, Italy
| | - Simona Gaudino
- Radiology Department, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Italy
| | | | | | - Fabio Maria Triulzi
- Neuroradiology Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Italy
| | - Guglielmo Pero
- Neuroradiology Department, ASST Grande Ospedale Metropolitano Niguarda, Italy
| | | | | | | | - Elena Bianchini
- Neuroradiology Unit, Radiology Department, Ospedale Legnano, Italy
| | - Giovanni Foti
- Radiology Department, Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| | | | - Massimo Sponza
- Neuroradiology Department, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Roberto Menozzi
- Neuroradiology Department, Azienda Ospedaliera Universitaria, Parma, Italy
| | - Mirco Cosottini
- Neuroradiology Department, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | | | - Luca Saba
- Radiology Department, Azienda Ospedaliero Universitaria, Cagliari, Italy
| | | |
Collapse
|
15
|
Nyvlt P, Schuster FS, Ihlow J, Heeren P, Spies C, Hiesgen J, Schenk T, von Brünneck AC, Westermann J, Brunkhorst FM, La Rosée P, Janka G, Lachmann C, Lachmann G. Value of hemophagocytosis in the diagnosis of hemophagocytic lymphohistiocytosis in critically ill patients. Eur J Haematol 2024; 112:917-926. [PMID: 38368850 DOI: 10.1111/ejh.14185] [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: 12/13/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Ferritin is an established biomarker in the diagnosis of secondary hemophagocytic lymphohistiocytosis (HLH), which is diagnosed by the HLH-2004 criteria. Among these criteria, detection of hemophagocytosis through invasive procedures may delay early life saving treatment. Our aim was to investigate the value of hemophagocytosis in diagnosing HLH in critically ill patients. METHODS In this secondary analysis of a retrospective observational study, we included all patients aged ≥18 years and admitted to any adult ICU at Charité-Universitätsmedizin Berlin between January 2006 and August 2018, who had hyperferritinemia (≥500 μg/L) and underwent bone marrow biopsy during their ICU course. RESULTS Two hundred fifty-two patients were included, of whom 31 (12.3%) showed hemophagocytosis. In multivariable logistic regression analysis, maximum ferritin was independently associated with hemophagocytosis. By removing hemophagocytosis from HLH-2004 criteria and HScore, prediction accuracy for HLH diagnosis was only marginally decreased compared to the original scores. CONCLUSIONS Our results strengthen the diagnostic value of ferritin and underline the importance of considering HLH diagnosis in patients with high ferritin but only four fulfilled HLH-2004 criteria, when hemophagocytosis was not assessed or not detectable. Proof of hemophagocytosis is not required for a reliable HLH diagnosis.
Collapse
Affiliation(s)
- Peter Nyvlt
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Anesthesiology and Intensive Care Medicine (CCM, CVK), Berlin, Germany
| | - Friederike S Schuster
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Anesthesiology and Intensive Care Medicine (CCM, CVK), Berlin, Germany
| | - Jana Ihlow
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Patrick Heeren
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Anesthesiology and Intensive Care Medicine (CCM, CVK), Berlin, Germany
| | - Claudia Spies
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Anesthesiology and Intensive Care Medicine (CCM, CVK), Berlin, Germany
| | - Josephine Hiesgen
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Anesthesiology and Intensive Care Medicine (CCM, CVK), Berlin, Germany
| | - Thomas Schenk
- Department of Hematology and Oncology, Universitätsklinikum Jena, Jena, Germany
| | - Ann-Christin von Brünneck
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany
| | - Jörg Westermann
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hematology, Oncology and Tumorimmunology, Berlin, Germany
| | - Frank M Brunkhorst
- Center for Clinical Studies, Department of Anesthesiology and Intensive Care Medicine, Universitätsklinikum Jena, Jena, Germany
| | - Paul La Rosée
- Klinik für Innere Medizin II, Schwarzwald-Baar-Klinikum, Villingen-Schwenningen, Germany
| | - Gritta Janka
- Clinic of Pediatric Hematology and Oncology, University Medical Center Eppendorf, Hamburg, Germany
| | - Cornelia Lachmann
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Anesthesiology and Intensive Care Medicine (CCM, CVK), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gunnar Lachmann
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Anesthesiology and Intensive Care Medicine (CCM, CVK), Berlin, Germany
- Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
16
|
Alfieri L, Franceschetti L, Frisoni P, Bonato O, Radaelli D, Bonuccelli D, D’Errico S, Neri M. Cardiac SARS-CoV-2 Infection, Involvement of Cytokines in Postmortem Immunohistochemical Study. Diagnostics (Basel) 2024; 14:787. [PMID: 38667433 PMCID: PMC11049034 DOI: 10.3390/diagnostics14080787] [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: 02/29/2024] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
In the context of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, significant attention was given to pulmonary manifestations. However, cardiac involvement is increasingly recognized as a critical factor influencing the prognosis, leading to myocardial damage, heart failure, acute coronary syndromes, potentially lethal arrhythmic events, and sudden cardiac death. Despite these findings, there is a lack of studies detailing the necroscopic, macroscopic, and microscopic cardiac changes associated with SARS-CoV-2. This study aimed to investigate the presence of SARS-CoV-2 viral proteins in cardiac tissue using immunohistochemical techniques to assess viral tropism. The analysis of cardiac tissue samples from deceased subjects, in different stages of conservation, confirmed to be positive for SARS-CoV-2 via reverse transcriptase-polymerase chain reaction (RT-PCR), showed immunopositivity for the SARS-CoV-2-NP viral antigen in 33% of cases. Notably, the presence of leukocyte infiltrates sufficient for diagnosing lymphocytic myocarditis was not observed. The central proinflammatory cytokines involved in the pathogenetic mechanism of coronavirus disease 19 (COVID-19) were researched using the immunohistochemical method. A significant increase in cytokine expression was detected, indicating myocardial involvement and dysfunction during SARS-CoV-2 infection. These findings suggest that the immunohistochemical detection of SARS-CoV-2 viral antigens and inflammatory cytokine expression in cardiac tissue could be crucial for a proper forensic assessment of the cause of death, even in sudden cardiac death.
Collapse
Affiliation(s)
- Letizia Alfieri
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Lorenzo Franceschetti
- Institute of Legal Medicine, Department of Biomedical Sciences for Health, University of Milan, 20133 Milano, Italy
| | - Paolo Frisoni
- Unit of Legal Medicine, AUSL Romagna, G.B. Morgagni-L. Pierantoni Hospital, 47100 Forlì, Italy;
| | - Omar Bonato
- Unit of Legal Medicine, AULSS 5 Polesana, 45100 Rovigo, Italy;
| | - Davide Radaelli
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (D.R.); (S.D.)
| | - Diana Bonuccelli
- Department of Legal Medicine, Territorial Unit USL Toscana Nord-Ovest, 55100 Lucca, Italy;
| | - Stefano D’Errico
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (D.R.); (S.D.)
| | - Margherita Neri
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy;
| |
Collapse
|
17
|
Conte C, Cipponeri E, Roden M. Diabetes Mellitus, Energy Metabolism, and COVID-19. Endocr Rev 2024; 45:281-308. [PMID: 37934800 PMCID: PMC10911957 DOI: 10.1210/endrev/bnad032] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/30/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023]
Abstract
Obesity, diabetes mellitus (mostly type 2), and COVID-19 show mutual interactions because they are not only risk factors for both acute and chronic COVID-19 manifestations, but also because COVID-19 alters energy metabolism. Such metabolic alterations can lead to dysglycemia and long-lasting effects. Thus, the COVID-19 pandemic has the potential for a further rise of the diabetes pandemic. This review outlines how preexisting metabolic alterations spanning from excess visceral adipose tissue to hyperglycemia and overt diabetes may exacerbate COVID-19 severity. We also summarize the different effects of SARS-CoV-2 infection on the key organs and tissues orchestrating energy metabolism, including adipose tissue, liver, skeletal muscle, and pancreas. Last, we provide an integrative view of the metabolic derangements that occur during COVID-19. Altogether, this review allows for better understanding of the metabolic derangements occurring when a fire starts from a small flame, and thereby help reducing the impact of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Caterina Conte
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome 00166, Italy
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan 20099, Italy
| | - Elisa Cipponeri
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milan 20099, Italy
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
- German Center for Diabetes Research, Partner Düsseldorf, Neuherberg 85764, Germany
| |
Collapse
|
18
|
Bradshaw CM, Georgieva T, Tankersley TN, Taylor-Doyle T, Johnson L, Uhrlaub JL, Besselsen D, Nikolich JŽ. Cutting Edge: Characterization of Low Copy Number Human Angiotensin-Converting Enzyme 2-Transgenic Mice as an Improved Model of SARS-CoV-2 Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:523-528. [PMID: 38197714 DOI: 10.4049/jimmunol.2300591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024]
Abstract
A popular mouse model of COVID-19, the K18-hACE2 mouse, expresses the SARS-coronavirus entry receptor, human angiotensin-converting enzyme 2 (hACE2) driven by the keratin-18 promoter. SARS-CoV-2-infected K18-hACE2 mice exhibit neuropathology not representative of human infection. They contain eight transgene (Tg) copies, leading to excess hACE2 expression and rampant viral replication. We generated two new lines of K18-hACE2 mice encoding one and two copies of hACE2 (1-hACE2-Tg and 2-hACE2-Tg, respectively). Relative to the original strain (called 8-hACE2-Tg in this study), 2-hACE2-Tg mice exhibited lower mortality, with less viral replication in the lung and brain. Furthermore, 1-hACE2-Tg mice exhibited no mortality and had no detectable virus in the brain; yet, they exhibited clear viral replication in the lung. All three strains showed SARS-CoV-2-related weight loss commensurate with the mortality rates. 1-hACE2-Tg mice mounted detectable primary and memory T effector cell and Ab responses. We conclude that these strains provide improved models to study hACE2-mediated viral infections.
Collapse
Affiliation(s)
- Christine M Bradshaw
- Department of Immunobiology, University of Arizona College of Medicine - Tucson, Tucson, AZ
- Arizona Center on Aging, University of Arizona College of Medicine - Tucson, Tucson, AZ
| | - Teodora Georgieva
- Genetically Engineered Mouse Model (GEMM) Core, University of Arizona, Tucson, AZ
- BIO5 Institute, University of Arizona, Tucson, AZ
| | - Trevor N Tankersley
- Department of Immunobiology, University of Arizona College of Medicine - Tucson, Tucson, AZ
- Arizona Center on Aging, University of Arizona College of Medicine - Tucson, Tucson, AZ
| | - Tama Taylor-Doyle
- Genetically Engineered Mouse Model (GEMM) Core, University of Arizona, Tucson, AZ
- BIO5 Institute, University of Arizona, Tucson, AZ
| | - Larry Johnson
- Genetically Engineered Mouse Model (GEMM) Core, University of Arizona, Tucson, AZ
- BIO5 Institute, University of Arizona, Tucson, AZ
| | - Jennifer L Uhrlaub
- Department of Immunobiology, University of Arizona College of Medicine - Tucson, Tucson, AZ
- Arizona Center on Aging, University of Arizona College of Medicine - Tucson, Tucson, AZ
| | - David Besselsen
- BIO5 Institute, University of Arizona, Tucson, AZ
- Arizona Animal Care, University of Arizona, Tucson, AZ
| | - Janko Ž Nikolich
- Department of Immunobiology, University of Arizona College of Medicine - Tucson, Tucson, AZ
- Arizona Center on Aging, University of Arizona College of Medicine - Tucson, Tucson, AZ
- BIO5 Institute, University of Arizona, Tucson, AZ
- Aegis Consortium for Pandemic-Free Future, University of Arizona Health Sciences, Tucson, AZ
| |
Collapse
|
19
|
Borczuk AC. Pathogenesis of Pulmonary Long COVID-19. Mod Pathol 2024; 37:100378. [PMID: 37931841 DOI: 10.1016/j.modpat.2023.100378] [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: 10/23/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
COVID-19 is characterized by an acute respiratory illness that, in some patients, progresses to respiratory failure, largely demonstrating a pattern of acute respiratory distress syndrome. Excluding fatal cases, the outcome of this severe illness ranges from complete resolution to persistent respiratory dysfunction. This subacute-to-chronic respiratory illness has different manifestations and is collectively termed as "long COVID." The pathogenesis of organ dysfunction in acute injury stems from exaggerated innate immune response, complement activation, and monocyte influx, with a shift toward an organ injury state with abnormalities in cellular maturation. Although the increased rate of thrombosis observed in acute COVID-19 does not appear to persist, interestingly, ongoing symptomatic COVID-19 and post-COVID pathogeneses appear to reflect the persistence of immune and cellular disturbances triggered by the acute and subacute periods.
Collapse
|
20
|
Aziz MF, Schenning K, Koike S, O'Glasser A, O'Reilly-Shah VN, Sera V, Mathis M. Perioperative Mortality of the COVID-19 Recovered Patient Compared to a Matched Control: A Multicenter Retrospective Cohort Study. Anesthesiology 2024; 140:195-206. [PMID: 37844271 DOI: 10.1097/aln.0000000000004809] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
BACKGROUND Surgical procedures performed on patients with recent exposure to COVID-19 infection have been associated with increased mortality risk in previous studies. Accordingly, elective surgery is often delayed after infection. The study aimed to compare 30-day hospital mortality and postoperative complications (acute kidney injury, pulmonary complications) of surgical patients with a previous COVID-19 infection to a matched cohort of patients without known previous COVID-19. The authors hypothesized that COVID-19 exposure would be associated with an increased mortality risk. METHODS In this retrospective observational cohort study, patients presenting for elective inpatient surgery across a multicenter cohort of academic and community hospitals from April 2020 to April 2021 who had previously tested positive for COVID-19 were compared to controls who had received at least one previous COVID-19 test but without a known previous COVID-19-positive test. The cases were matched based on anthropometric data, institution, and comorbidities. Further, the outcomes were analyzed stratified by timing of a positive test result in relation to surgery. RESULTS Thirty-day mortality occurred in 229 of 4,951 (4.6%) COVID-19-exposed patients and 122 of 4,951 (2.5%) controls. Acute kidney injury was observed in 172 of 1,814 (9.5%) exposed patients and 156 of 1,814 (8.6%) controls. Pulmonary complications were observed in 237 of 1,637 (14%) exposed patients and 164 of 1,637 (10%) controls. COVID-19 exposure was associated with an increased 30-day mortality risk (adjusted odds ratio, 1.63; 95% CI, 1.38 to 1.91) and an increased risk of pulmonary complications (1.60; 1.36 to 1.88), but was not associated with an increased risk of acute kidney injury (1.03; 0.87 to 1.22). Surgery within 2 weeks of infection was associated with a significantly increased risk of mortality and pulmonary complications, but that effect was nonsignificant after 2 weeks. CONCLUSIONS Patients with a positive test for COVID-19 before elective surgery early in the pandemic have an elevated risk of perioperative mortality and pulmonary complications but not acute kidney injury as compared to matched controls. The span of time from positive test to time of surgery affected the mortality and pulmonary risk, which subsided after 2 weeks. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Michael F Aziz
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - Katie Schenning
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - Seiji Koike
- Biostatistics and Design Program, Oregon Health & Science University, Portland, Oregon
| | - Avital O'Glasser
- Departments of Medicine and Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | | | - Valerie Sera
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon
| | - Michael Mathis
- Department of Anesthesiology, University of Michigan Medicine, Ann Arbor, Michigan
| |
Collapse
|
21
|
Angiola F, Franchetti G, Cestonaro C, Agnolucci J, Giordano R, Viel G. Dying at home during the SARS-CoV-2 endemic: The importance of defining the exact mechanism of death. Leg Med (Tokyo) 2024; 66:102361. [PMID: 38039658 DOI: 10.1016/j.legalmed.2023.102361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 11/08/2023] [Accepted: 11/22/2023] [Indexed: 12/03/2023]
Abstract
INTRODUCTION Coronavirus Disease 2019 (COVID-19) has become endemic in Europe thanks to the presence of less deadly and more infectious variants and to the existence of a significant portion of unvaccinated people among the general population. SARS-Cov-2 related deaths are probably going to fade in the next years, but Covid-19 should still be considered a potential cause of death in the out-of-hospital setting in the next future. MATERIAL AND METHODS Three (3) cases of unexpected death at home are here presented. Each case has been investigated with the same methodological approach: death scene investigation (DSI), complete autopsy with histology, immunohistochemistry, RNA in situ hybridization for SARS-CoV-2 spike protein in lung tissue, toxicology and microbiology. RESULTS AND DISCUSSION All three cases had a COVID + post-mortem nasopharyngeal swab. Histology and immunohistochemistry revealed a SARS-CoV-2 lung involvement in only two of the cases (Cases 2 and 3), while a septic bacterial pneumonia was found in Case 1, where RNA-in situ hybridization for viral spike protein showed no reactivity in pneumocytes. The integration of all postmortem evidence allowed to attribute a different role of SARS-Cov-2 in the determinism of the death. CONCLUSION In the current post-pandemic context, SARS-CoV-2 remains a possible cause of death when investigating out-of-hospital unexpected deaths. Since a positive post-mortem swab does not automatically imply a COVID-19-related death, histology and immunohistochemistry are helpful for identifying SARS-CoV-2 lung involvement and, therefore, its potential active role in the determinism of death.
Collapse
Affiliation(s)
- Francesco Angiola
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Falloppio 50, 35121 Padova, Italy
| | - Giorgia Franchetti
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Falloppio 50, 35121 Padova, Italy
| | - Clara Cestonaro
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Falloppio 50, 35121 Padova, Italy
| | - Jacopo Agnolucci
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Falloppio 50, 35121 Padova, Italy
| | - Renzo Giordano
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Falloppio 50, 35121 Padova, Italy
| | - Guido Viel
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Via Falloppio 50, 35121 Padova, Italy.
| |
Collapse
|
22
|
Troxel AB, Bind MAC, Flotte TJ, Cordon-Cardo C, Decker LA, Finn AV, Padera RF, Reichard RR, Stone JR, Adolphi NL, Casimero FVC, Crary JF, Elifritz J, Faustin A, Ghosh SKB, Krausert A, Martinez-Lage M, Melamed J, Mitchell RA, Sampson BA, Seifert AC, Simsir A, Adams C, Haasnoot S, Hafner S, Siciliano MA, Vallejos BB, Del Boccio P, Lamendola-Essel MF, Young CE, Kewlani D, Akinbo PA, Parent B, Chung A, Cato TC, Mudumbi PC, Esquenazi-Karonika S, Wood MJ, Chan J, Monteiro J, Shinnick DJ, Thaweethai T, Nguyen AN, Fitzgerald ML, Perlowski AA, Stiles LE, Paskett ML, Katz SD, Foulkes AS. Researching COVID to enhance recovery (RECOVER) tissue pathology study protocol: Rationale, objectives, and design. PLoS One 2024; 19:e0285645. [PMID: 38198481 PMCID: PMC10781091 DOI: 10.1371/journal.pone.0285645] [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: 04/25/2023] [Accepted: 11/23/2023] [Indexed: 01/12/2024] Open
Abstract
IMPORTANCE SARS-CoV-2 infection can result in ongoing, relapsing, or new symptoms or organ dysfunction after the acute phase of infection, termed Post-Acute Sequelae of SARS-CoV-2 (PASC), or long COVID. The characteristics, prevalence, trajectory and mechanisms of PASC are poorly understood. The objectives of the Researching COVID to Enhance Recovery (RECOVER) tissue pathology study (RECOVER-Pathology) are to: (1) characterize prevalence and types of organ injury/disease and pathology occurring with PASC; (2) characterize the association of pathologic findings with clinical and other characteristics; (3) define the pathophysiology and mechanisms of PASC, and possible mediation via viral persistence; and (4) establish a post-mortem tissue biobank and post-mortem brain imaging biorepository. METHODS RECOVER-Pathology is a cross-sectional study of decedents dying at least 15 days following initial SARS-CoV-2 infection. Eligible decedents must meet WHO criteria for suspected, probable, or confirmed infection and must be aged 18 years or more at the time of death. Enrollment occurs at 7 sites in four U.S. states and Washington, DC. Comprehensive autopsies are conducted according to a standardized protocol within 24 hours of death; tissue samples are sent to the PASC Biorepository for later analyses. Data on clinical history are collected from the medical records and/or next of kin. The primary study outcomes include an array of pathologic features organized by organ system. Causal inference methods will be employed to investigate associations between risk factors and pathologic outcomes. DISCUSSION RECOVER-Pathology is the largest autopsy study addressing PASC among US adults. Results of this study are intended to elucidate mechanisms of organ injury and disease and enhance our understanding of the pathophysiology of PASC.
Collapse
Affiliation(s)
- Andrea B. Troxel
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Marie-Abele C. Bind
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | - Thomas J. Flotte
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, MN, United States of America
| | - Carlos Cordon-Cardo
- Department of Pathology, Molecular and Cell-Based Medicine, Mount Sinai Health System, New York, NY, United States of America
| | - Lauren A. Decker
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Aloke V. Finn
- Department of Pathology, CVPath Institute, Gaithersburg, MD, United States of America
| | - Robert F. Padera
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, United States of America
| | - R. Ross Reichard
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, MN, United States of America
| | - James R. Stone
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Natalie L. Adolphi
- Office of the Medical Investigator, University of New Mexico School of Medicine, Albuquerque, NM, United States of America
| | | | - John F. Crary
- Department of Pathology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, United States of America
| | - Jamie Elifritz
- Departments of Radiology and Pathology, University of New Mexico, Albuquerque, NM, United States of America
| | - Arline Faustin
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Saikat Kumar B. Ghosh
- Department of Molecular Biology and Genomics, CVPath Institute, Gaithersburg, MD, United States of America
| | - Amanda Krausert
- Department of Pathology, Molecular and Cell-Based Medicine, Mount Sinai Health System, New York, NY, United States of America
| | - Maria Martinez-Lage
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Jonathan Melamed
- Department of Anatomical Pathology, NYU Langone Hospital—Long Island, Mineola, NY, United States of America
| | - Roger A. Mitchell
- Department of Pathology, Howard University College of Medicine, Washington DC, United States of America
| | - Barbara A. Sampson
- Department of Pathology, Molecular and Cell-Based Medicine, Mount Sinai Health System, New York, NY, United States of America
| | - Alan C. Seifert
- Biomedical Engineering and Imaging Institute, Department of Radiology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Aylin Simsir
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Cheryle Adams
- Department of Pathology, Howard University College of Medicine, Washington DC, United States of America
| | - Stephanie Haasnoot
- Department of Pathology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY, United States of America
| | - Stephanie Hafner
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, Rochester, MN, United States of America
| | - Michelle A. Siciliano
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, United States of America
| | - Brittany B. Vallejos
- Office of the Medical Investigators, Department of Research, University of New Mexico, Albuquerque, NM, United States of America
| | - Phoebe Del Boccio
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Michelle F. Lamendola-Essel
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Chloe E. Young
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Deepshikha Kewlani
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Precious A. Akinbo
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Brendan Parent
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Alicia Chung
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Teresa C. Cato
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Praveen C. Mudumbi
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Shari Esquenazi-Karonika
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Marion J. Wood
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States of America
| | - James Chan
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | - Jonathan Monteiro
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | - Daniel J. Shinnick
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | - Tanayott Thaweethai
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | - Amber N. Nguyen
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | - Megan L. Fitzgerald
- Patient-Led Research Collaborative on COVID-19, Washington DC, United States of America
| | | | - Lauren E. Stiles
- Department of Neurology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY, United States of America
| | - Moira L. Paskett
- Department of Anatomical Pathology, NYU Langone Hospital—Long Island, Mineola, NY, United States of America
| | - Stuart D. Katz
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States of America
| | - Andrea S. Foulkes
- Department of Biostatistics, Massachusetts General Hospital, Boston, MA, United States of America
| | | |
Collapse
|
23
|
Narayanan SA, Jamison DA, Guarnieri JW, Zaksas V, Topper M, Koutnik AP, Park J, Clark KB, Enguita FJ, Leitão AL, Das S, Moraes-Vieira PM, Galeano D, Mason CE, Trovão NS, Schwartz RE, Schisler JC, Coelho-Dos-Reis JGA, Wurtele ES, Beheshti A. A comprehensive SARS-CoV-2 and COVID-19 review, Part 2: host extracellular to systemic effects of SARS-CoV-2 infection. Eur J Hum Genet 2024; 32:10-20. [PMID: 37938797 PMCID: PMC10772081 DOI: 10.1038/s41431-023-01462-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 09/01/2023] [Accepted: 09/13/2023] [Indexed: 11/09/2023] Open
Abstract
COVID-19, the disease caused by SARS-CoV-2, has caused significant morbidity and mortality worldwide. The betacoronavirus continues to evolve with global health implications as we race to learn more to curb its transmission, evolution, and sequelae. The focus of this review, the second of a three-part series, is on the biological effects of the SARS-CoV-2 virus on post-acute disease in the context of tissue and organ adaptations and damage. We highlight the current knowledge and describe how virological, animal, and clinical studies have shed light on the mechanisms driving the varied clinical diagnoses and observations of COVID-19 patients. Moreover, we describe how investigations into SARS-CoV-2 effects have informed the understanding of viral pathogenesis and provide innovative pathways for future research on the mechanisms of viral diseases.
Collapse
Affiliation(s)
- S Anand Narayanan
- COVID-19 International Research Team, Medford, MA, 02155, USA.
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL, 32301, USA.
| | - David A Jamison
- COVID-19 International Research Team, Medford, MA, 02155, USA
| | - Joseph W Guarnieri
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Victoria Zaksas
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Center for Translational Data Science, University of Chicago, Chicago, IL, 60637, USA
- Clever Research Lab, Springfield, IL, 62704, USA
| | - Michael Topper
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Departments of Oncology and Medicine and the Sidney Comprehensive Cancer Center, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Andrew P Koutnik
- Human Healthspan, Resilience, and Performance, Florida Institute for Human and Machine Cognition, Pensacola, FL, 32502, USA
- Sansum Diabetes Research Institute, Santa Barbara, CA, 93015, USA
| | - Jiwoon Park
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, 10065, USA
| | - Kevin B Clark
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Cures Within Reach, Chicago, IL, 60602, USA
- Campus and Domain Champions Program, Multi-Tier Assistance, Training, and Computational Help (MATCH) Track, National Science Foundation's Advanced Cyberinfrastructure Coordination Ecosystem: Services and Support (ACCESS), Philadelphia, PA, USA
- Expert Network, Penn Center for Innovation, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Biometrics and Nanotechnology Councils, Institute for Electrical and Electronics Engineers, New York, NY, 10016, USA
- Peace Innovation Institute, The Hague 2511, Netherlands and Stanford University, Palo Alto, 94305, CA, USA
| | - Francisco J Enguita
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Ana Lúcia Leitão
- MEtRICs, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal
| | - Saswati Das
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Atal Bihari Vajpayee Institute of Medical Sciences and Dr Ram Mannohar Lohia Hospital, New Delhi, 110001, India
| | - Pedro M Moraes-Vieira
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Department of Genetics, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- Experimental Medicine Research Cluster (EMRC) and Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, Brazil
| | - Diego Galeano
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Facultad de Ingeniería, Universidad Nacional de Asunción, San Lorenzo, Paraguay
| | - Christopher E Mason
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Nídia S Trovão
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Fogarty International Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Robert E Schwartz
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Jonathan C Schisler
- COVID-19 International Research Team, Medford, MA, 02155, USA
- McAllister Heart Institute and Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jordana G A Coelho-Dos-Reis
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Basic and Applied Virology Lab, Department of Microbiology, Institute for Biological Sciences (ICB), Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Eve Syrkin Wurtele
- COVID-19 International Research Team, Medford, MA, 02155, USA
- Genetics Program, Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 90011, USA
- Bioinformatics and Computational Biology Program, Center for Metabolomics, Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 90011, USA
| | - Afshin Beheshti
- COVID-19 International Research Team, Medford, MA, 02155, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, Santa Clara, CA, 94035, USA.
| |
Collapse
|
24
|
Magaki S, Zhang T, Han K, Hilda M, Yong WH, Achim C, Fishbein G, Fishbein MC, Garner O, Salamon N, Williams CK, Valdes-Sueiras MA, Hsu JJ, Kelesidis T, Mathisen GE, Lavretsky H, Singer EJ, Vinters HV. HIV and COVID-19: two pandemics with significant (but different) central nervous system complications. FREE NEUROPATHOLOGY 2024; 5:5-5. [PMID: 38469363 PMCID: PMC10925920 DOI: 10.17879/freeneuropathology-2024-5343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/02/2024] [Indexed: 03/13/2024]
Abstract
Human immunodeficiency virus (HIV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cause significant neurologic disease. Central nervous system (CNS) involvement of HIV has been extensively studied, with well-documented invasion of HIV into the brain in the initial stage of infection, while the acute effects of SARS-CoV-2 in the brain are unclear. Neuropathologic features of active HIV infection in the brain are well characterized whereas neuropathologic findings in acute COVID-19 are largely non-specific. On the other hand, neuropathologic substrates of chronic dysfunction in both infections, as HIV-associated neurocognitive disorders (HAND) and post-COVID conditions (PCC)/long COVID are unknown. Thus far, neuropathologic studies on patients with HAND in the era of combined antiretroviral therapy have been inconclusive, and autopsy studies on patients diagnosed with PCC have yet to be published. Further longitudinal, multidisciplinary studies on patients with HAND and PCC and neuropathologic studies in comparison to controls are warranted to help elucidate the mechanisms of CNS dysfunction in both conditions.
Collapse
Affiliation(s)
- Shino Magaki
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA, USA
| | - Ting Zhang
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA, USA
| | - Karam Han
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA, USA
| | - Mirbaha Hilda
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA, USA
| | - William H. Yong
- Department of Pathology and Laboratory Medicine, University of California-Irvine School of Medicine, Irvine, CA, USA
| | - Cristian Achim
- Department of Psychiatry, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Gregory Fishbein
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael C. Fishbein
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Omai Garner
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Noriko Salamon
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Christopher K. Williams
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA, USA
| | - Miguel A. Valdes-Sueiras
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jeffrey J. Hsu
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Theodoros Kelesidis
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Glenn E. Mathisen
- Department of Infectious Diseases, Olive View-University of California Los Angeles Medical Center, Sylmar, CA, USA
| | - Helen Lavretsky
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Elyse J. Singer
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Harry V. Vinters
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Brain Research Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
25
|
Reina-Reina A, Barrera J, Maté A, Trujillo J, Valdivieso B, Gas ME. Developing an interpretable machine learning model for predicting COVID-19 patients deteriorating prior to intensive care unit admission using laboratory markers. Heliyon 2023; 9:e22878. [PMID: 38125502 PMCID: PMC10731083 DOI: 10.1016/j.heliyon.2023.e22878] [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: 05/27/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Coronavirus disease (COVID-19) remains a significant global health challenge, prompting a transition from emergency response to comprehensive management strategies. Furthermore, the emergence of new variants of concern, such as BA.2.286, underscores the need for early detection and response to new variants, which continues to be a crucial strategy for mitigating the impact of COVID-19, especially among the vulnerable population. This study aims to anticipate patients requiring intensive care or facing elevated mortality risk throughout their COVID-19 infection while also identifying laboratory predictive markers for early diagnosis of patients. Therefore, haematological, biochemical, and demographic variables were retrospectively evaluated in 8,844 blood samples obtained from 2,935 patients before intensive care unit admission using an interpretable machine learning model. Feature selection techniques were applied using precision-recall measures to address data imbalance and evaluate the suitability of the different variables. The model was trained using stratified cross-validation with k=5 and internally validated, achieving an accuracy of 77.27%, sensitivity of 78.55%, and area under the receiver operating characteristic (AUC) of 0.85; successfully identifying patients at increased risk of severe progression. From a medical perspective, the most important features of the progression or severity of patients with COVID-19 were lactate dehydrogenase, age, red blood cell distribution standard deviation, neutrophils, and platelets, which align with findings from several prior investigations. In light of these insights, diagnostic processes can be significantly expedited through the use of laboratory tests, with a greater focus on key indicators. This strategic approach not only improves diagnostic efficiency but also extends its reach to a broader spectrum of patients. In addition, it allows healthcare professionals to take early preventive measures for those most at risk of adverse outcomes, thereby optimising patient care and prognosis.
Collapse
Affiliation(s)
- A. Reina-Reina
- Lucentia Research. Department of Software and Computing System, University of Alicante, Carretera San Vicente del Raspeig s/n, 03690, Alicante, Spain
- Lucentia Lab, Av. Pintor Pérez Gil, 16, 03540, Alicante, Spain
| | - J.M. Barrera
- Lucentia Research. Department of Software and Computing System, University of Alicante, Carretera San Vicente del Raspeig s/n, 03690, Alicante, Spain
- Lucentia Lab, Av. Pintor Pérez Gil, 16, 03540, Alicante, Spain
| | - A. Maté
- Lucentia Research. Department of Software and Computing System, University of Alicante, Carretera San Vicente del Raspeig s/n, 03690, Alicante, Spain
- Lucentia Lab, Av. Pintor Pérez Gil, 16, 03540, Alicante, Spain
| | - J.C. Trujillo
- Lucentia Research. Department of Software and Computing System, University of Alicante, Carretera San Vicente del Raspeig s/n, 03690, Alicante, Spain
- Lucentia Lab, Av. Pintor Pérez Gil, 16, 03540, Alicante, Spain
| | - B. Valdivieso
- The University and Polytechnic La Fe Hospital of Valencia, Avenida Fernando Abril Martorell, 106 Torre H 1st floor, 46026, Valencia, Spain
- The Medical Research Institute of Hospital La Fe, Avenida Fernando Abril Martorell, 106 Torre F 7th floor, 46026, Valencia, Spain
| | - María-Eugenia Gas
- The Medical Research Institute of Hospital La Fe, Avenida Fernando Abril Martorell, 106 Torre F 7th floor, 46026, Valencia, Spain
| |
Collapse
|
26
|
Nasyrov RA, Ivanov DO, Krasnogorskaya OL, Timchenko VN, Fedotova EP, Chepelev AS, Galichina VA, Sidorova NA, Anichkov NM. COVID-19 in Children: Molecular Profile and Pathological Features. Int J Mol Sci 2023; 24:16750. [PMID: 38069078 PMCID: PMC10706827 DOI: 10.3390/ijms242316750] [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: 10/09/2023] [Revised: 11/15/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Although the World Health Organization has declared the end of the COVID-19 pandemic, doctors continue to register new cases of the disease among both adults and children. Unfortunately, the course of COVID-19 in children can have a severe form, with death being a potential outcome. The absence of published works discussing the pathological morphology of COVID-19 in children prevents the objective analysis of the disease's pathogenesis, including among the adult population. In this vein, the objective of our study is to identify the morphological features of the lungs' involvement and evaluate virus-host interactions in the case of COVID-19 in patients at a pediatric medical practice. We present the results of the study of the lungs of three children who died due to COVID-19, highlighting the predominant involvement of their respiratory organs at different stages of the disease (5, 21, and 50 days). This article presents data obtained from histopathological and immunohistochemical investigations, taking into account the results of clinical and laboratory indicators and intravital and postmortem SARS-CoV-2 PCR investigations. The common finding of all of the examined COVID-19 cases is the involvement of the endothelium in microcirculation vessels, which are considered to be a primary target of various pathogenic influencing factors. We also discuss both the significance of apoptosis as a result of virus-host interactions and the most likely cause of endothelium cell destruction. The results of this study could be useful for the development of endothelium-protective therapy to prevent the progression of disseminated intravascular coagulation syndrome.
Collapse
Affiliation(s)
- Ruslan A. Nasyrov
- The Prof. D.D. Lohov Department of Pathological Anatomy with Course of Forensic Medicine, Saint Petersburg State Pediatric Medical University Ministry of Public Health Care of the Russian Federation, St. Litovskaya, 2, 194100 St. Petersburg, Russia; (D.O.I.); (O.L.K.); (V.N.T.); (E.P.F.); (A.S.C.); (V.A.G.); (N.A.S.); (N.M.A.)
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Cappelletti P, Gallo G, Marino R, Palaniappan S, Corbo M, Savoia C, Feligioni M. From cardiovascular system to brain, the potential protective role of Mas Receptors in COVID-19 infection. Eur J Pharmacol 2023; 959:176061. [PMID: 37775018 DOI: 10.1016/j.ejphar.2023.176061] [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: 07/13/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 10/01/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has been declared a new pandemic in March 2020. Although most patients are asymptomatic, those with underlying cardiovascular comorbidities may develop a more severe systemic infection which is often associated with fatal pneumonia. Nonetheless, neurological and cardiovascular manifestations could be present even without respiratory symptoms. To date, no COVID-19-specific drugs are able for preventing or treating the infection and generally, the symptoms are relieved with general anti-inflammatory drugs. Angiotensin-converting-enzyme 2 (ACE2) may function as the receptor for virus entry within the cells favoring the progression of infection in the organism. On the other hand, ACE2 is a relevant enzyme in renin angiotensin system (RAS) cascade fostering Ang1-7/Mas receptor activation which promotes protective effects in neurological and cardiovascular systems. It is known that RAS is composed by two functional countervailing axes the ACE/AngII/AT1 receptor and the ACE/AngII/AT2 receptor which counteracts the actions mediated by AngII/AT1 receptor by inducing anti-inflammatory, antioxidant and anti-growth functions. Subsequently an "alternative" ACE2/Ang1-7/Mas receptor axis has been described with functions similar to the latter protective arm. Here, we discuss the neurological and cardiovascular effects of COVID-19 highlighting the role of the stimulation of the RAS "alternative" protective arm in attenuating pulmonary, cerebral and cardiovascular damages. In conclusion, only two clinical trials are running for Mas receptor agonists but few other molecules are in preclinical phase and if successful these drugs might represent a successful strategy for the treatment of the acute phase of COVID-19 infection.
Collapse
Affiliation(s)
- Pamela Cappelletti
- Department of Neuro-Rehabilitation Sciences, Casa di Cura Igea, Milan, Italy.
| | - Giovanna Gallo
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Rachele Marino
- European Brain Research Institute (EBRI) Rita Levi Montalcini Foundation, Rome, Italy
| | | | - Massimo Corbo
- Department of Neuro-Rehabilitation Sciences, Casa di Cura Igea, Milan, Italy
| | - Carmine Savoia
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Marco Feligioni
- Department of Neuro-Rehabilitation Sciences, Casa di Cura Igea, Milan, Italy; European Brain Research Institute (EBRI) Rita Levi Montalcini Foundation, Rome, Italy.
| |
Collapse
|
28
|
Noack P, Grosse C, Bodingbauer J, Almeder M, Lohfink-Schumm S, Salzer HJF, Meier J, Lamprecht B, Schmitt CA, Langer R. Minimally invasive autopsies for the investigation of pulmonary pathology of COVID-19-experiences of a longitudinal series of 92 patients. Virchows Arch 2023; 483:611-619. [PMID: 37653260 PMCID: PMC10673967 DOI: 10.1007/s00428-023-03622-6] [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: 01/24/2023] [Revised: 07/20/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023]
Abstract
Minimally invasive autopsies (MIAs) allow the collection of tissue samples for diagnostic and research purposes in special situations, e.g., when there is a high risk of infection which is the case in the context of COVID-19 or restrictions due to legal or personal reasons. We performed MIA to analyze lung tissue from 92 COVID-19 patients (mean age 78 years; range 48-98; 35 women, 57 men), representing 44% of all patients who died from the disease between October 2020 and April 2021. An intercostal approach was used with removal of a 5-cm rib section followed by manual collection of four lung tissue samples (5-8 cm in size). Diffuse alveolar damage (DAD) was found in 89 (97%) patients at various stages. Exudative DAD (eDAD) predominated in 18 (20%) patients, proliferative DAD (pDAD) in 43 (47%) patients, and mixed DAD (mDAD) in 31 (34%) patients. There were no significant differences in the predominant DAD pattern between tissue samples from the same patient. Additional purulent components were present in 46 (50%) cases. Fungi were detected in 11 (12%) patients. The pDAD pattern was associated with longer hospital stay including intensive care unit (p=0.026 and p<0.001) and younger age (p=0.019). Positive bronchoalveolar lavage and blood cultures were observed more frequently in pDAD patterns (p<0.001; p=0.018). In contrast, there was no significant association between intravital positive microbiological results and superimposed bronchopneumonia or fungal infection at autopsy. Having demonstrated the characteristic lung changes in a large longitudinal autopsy series, we conclude that the presented MIA approach can be considered a reliable and safe method for performing post mortem lung diagnostics in COVID-19 and other high-risk situations. The lack of correlation between histological changes indicative of bacterial or fungal superinfection and microbiology could have clinical implications for disease and treatment surveillance.
Collapse
Affiliation(s)
- Petar Noack
- Institute of Clinical Pathology, Kepler University Hospital, Krankenhausstr. 9, 4021, Linz, Austria
- Medical Faculty, Johannes Kepler University, Linz, Austria
| | - Claudia Grosse
- Institute of Clinical Pathology, Kepler University Hospital, Krankenhausstr. 9, 4021, Linz, Austria
| | - Jacob Bodingbauer
- Institute of Clinical Pathology, Kepler University Hospital, Krankenhausstr. 9, 4021, Linz, Austria
- Medical Faculty, Johannes Kepler University, Linz, Austria
| | - Marion Almeder
- Institute of Clinical Pathology, Kepler University Hospital, Krankenhausstr. 9, 4021, Linz, Austria
- Medical Faculty, Johannes Kepler University, Linz, Austria
| | - Sylvia Lohfink-Schumm
- Institute of Clinical Pathology, Kepler University Hospital, Krankenhausstr. 9, 4021, Linz, Austria
- Medical Faculty, Johannes Kepler University, Linz, Austria
| | - Helmut J F Salzer
- Medical Faculty, Johannes Kepler University, Linz, Austria
- Division of Infectious Diseases and Tropical Medicine, Department of Pulmonary Medicine, Kepler University Hospital, Linz, Austria
- Ignaz-Semmelweis-Institute, Interuniversity Institute for Infection Research, Vienna, Austria
| | - Jens Meier
- Medical Faculty, Johannes Kepler University, Linz, Austria
- Department of Anesthesiology and Intensive Care Medicine, Kepler University Hospital, Linz, Austria
| | - Bernd Lamprecht
- Medical Faculty, Johannes Kepler University, Linz, Austria
- Department of Pulmonary Medicine, Kepler University Hospital, Linz, Austria
| | - Clemens A Schmitt
- Medical Faculty, Johannes Kepler University, Linz, Austria
- Department of Hematology and Medical Oncology, Kepler University Hospital, Linz, Austria
| | - Rupert Langer
- Institute of Clinical Pathology, Kepler University Hospital, Krankenhausstr. 9, 4021, Linz, Austria.
- Medical Faculty, Johannes Kepler University, Linz, Austria.
| |
Collapse
|
29
|
Mortazavi S, de Peralta-Venturina M, Marchevsky AM. Nonspecific interstitial pneumonia pattern is a frequent finding in patients with post-acute COVID-19 syndrome treated with bilateral orthotopic lung transplantation: current best evidence. Hum Pathol 2023; 141:90-101. [PMID: 37364827 PMCID: PMC10290180 DOI: 10.1016/j.humpath.2023.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023]
Abstract
Patients with post-acute COVID-19 (PA-COVID) syndrome or long COVID-19 syndrome develop persistent symptoms and complications that last beyond 4 weeks of the initial infection. There is limited information regarding the pulmonary pathology in PA-COVID patients who require bilateral orthotopic lung transplantation (BOLT). Our experience with 40 lung explants from 20 PA-COVID patients who underwent BOLT is described. Clinicopathologic findings are correlated with best evidence from literature. The lung parenchyma showed bronchiectasis (n = 20) and severe interstitial fibrosis with areas resembling the nonspecific interstitial pneumonia (NSIP) pattern of fibrosis (n = 20), interstitial fibrosis not otherwise specified (n = 20), and fibrotic cysts (n = 9). None of the explants exhibited a usual interstitial pneumonia pattern of fibrosis. Other parenchymal changes included multinucleated giant cells (n = 17), hemosiderosis (n = 16), peribronchiolar metaplasia (n = 19), obliterative bronchiolitis (n = 6), and microscopic honeycombing (n = 5). Vascular abnormalities included thrombosis of a lobar artery (n = 1) and microscopic thrombi in small vessels (n = 7). Systematic literature review identified 7 articles reporting the presence in 12 patients of interstitial fibrosis showing the NSIP pattern (n = 3), organizing pneumonia/diffuse alveolar damage (n = 4) and not otherwise specified (n = 3) patterns. All but one of these studies also reported the presence of multinucleated giant cells and none of the studies reported the presence of severe vascular abnormalities. PA-COVID patients undergoing BOLT show a pattern of fibrosis that resembles a mixed cellular-fibrotic NSIP pattern and generally lack severe vascular complications. As the NSIP pattern of fibrosis is often associated with autoimmune diseases, additional studies are needed to understand the mechanism of disease and learn whether this information can be used for therapeutic purposes.
Collapse
Affiliation(s)
- Samira Mortazavi
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| | | | - Alberto M Marchevsky
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| |
Collapse
|
30
|
Heinrich F, Mertz KD, Glatzel M, Beer M, Krasemann S. Using autopsies to dissect COVID-19 pathogenesis. Nat Microbiol 2023; 8:1986-1994. [PMID: 37798476 DOI: 10.1038/s41564-023-01488-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 09/04/2023] [Indexed: 10/07/2023]
Abstract
The emergence of SARS-CoV-2 has resulted in millions of deaths as a result of COVID-19. Suitable models were missing at the beginning of the pandemic, and studies investigating disease pathogenesis relied on patients who had succumbed to COVID-19. Since then, autopsies of patients have substantially contributed to our understanding of the pathogenesis of COVID-19 and associated major organ complications. Here we summarize how autopsies have complemented experimental studies, mainly in animal models, and how they have facilitated critical knowledge of COVID-19 to improve daily clinical practice and develop therapeutic interventions. Employing advanced histopathologic and molecular genetic methods in post-mortem tissues, the COVID-19 pandemic has highlighted the importance of autopsies for virology research and clinical practice in current and emerging infectious diseases.
Collapse
Affiliation(s)
- Fabian Heinrich
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kirsten D Mertz
- Institute of Pathology, Kantonsspital Baselland, Liestal, Switzerland
- University of Basel, Basel, Switzerland
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Beer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
31
|
Stüdle C, Nishihara H, Wischnewski S, Kulsvehagen L, Perriot S, Ishikawa H, Schroten H, Frank S, Deigendesch N, Du Pasquier R, Schirmer L, Pröbstel AK, Engelhardt B. SARS-CoV-2 infects epithelial cells of the blood-cerebrospinal fluid barrier rather than endothelial cells or pericytes of the blood-brain barrier. Fluids Barriers CNS 2023; 20:76. [PMID: 37875964 PMCID: PMC10598911 DOI: 10.1186/s12987-023-00479-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/13/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND As a consequence of SARS-CoV-2 infection various neurocognitive and neuropsychiatric symptoms can appear, which may persist for several months post infection. However, cell type-specific routes of brain infection and underlying mechanisms resulting in neuroglial dysfunction are not well understood. METHODS Here, we investigated the susceptibility of cells constituting the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) of the choroid plexus (ChP) to SARS-CoV-2 infection using human induced pluripotent stem cell (hiPSC)-derived cellular models and a ChP papilloma-derived epithelial cell line as well as ChP tissue from COVID-19 patients, respectively. RESULTS We noted a differential infectibility of hiPSC-derived brain microvascular endothelial cells (BMECs) depending on the differentiation method. Extended endothelial culture method (EECM)-BMECs characterized by a complete set of endothelial markers, good barrier properties and a mature immune phenotype were refractory to SARS-CoV-2 infection and did not exhibit an activated phenotype after prolonged SARS-CoV-2 inoculation. In contrast, defined medium method (DMM)-BMECs, characterized by a mixed endothelial and epithelial phenotype and excellent barrier properties were productively infected by SARS-CoV-2 in an ACE2-dependent manner. hiPSC-derived brain pericyte-like cells (BPLCs) lacking ACE2 expression were not susceptible to SARS-CoV-2 infection. Furthermore, the human choroid plexus papilloma-derived epithelial cell line HIBCPP, modeling the BCSFB was productively infected by SARS-CoV-2 preferentially from the basolateral side, facing the blood compartment. Assessment of ChP tissue from COVID-19 patients by RNA in situ hybridization revealed SARS-CoV-2 transcripts in ChP epithelial and ChP stromal cells. CONCLUSIONS Our study shows that the BCSFB of the ChP rather than the BBB is susceptible to direct SARS-CoV-2 infection. Thus, neuropsychiatric symptoms because of COVID-19 may rather be associated with dysfunction of the BCSFB than the BBB. Future studies should consider a role of the ChP in underlying neuropsychiatric symptoms following SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Chiara Stüdle
- Theodor Kocher Institute, University of Bern, Bern, Switzerland.
| | - Hideaki Nishihara
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
- Department of Neurotherapeutics, Yamaguchi University, Yamaguchi, Japan
| | - Sven Wischnewski
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Laila Kulsvehagen
- Departments of Neurology, Biomedicine and Clinical Research, Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | - Sylvain Perriot
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, University of Tsukuba, Tsukuba, 305-8575, Ibaraki, Japan
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephan Frank
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Nikolaus Deigendesch
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Renaud Du Pasquier
- Laboratory of Neuroimmunology, Neuroscience Research Centre, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Service of Neurology, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Center for Translational Neuroscience and Institute for Innate Immunoscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Anne-Katrin Pröbstel
- Departments of Neurology, Biomedicine and Clinical Research, Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel and University of Basel, Basel, Switzerland
| | | |
Collapse
|
32
|
Abenavoli L, Aquila I, Sacco MA, Scarlata GGM, Procopio AC, Boccuto L, Scarpellini E, Greco M, Foti DP, Ricci P, Luzza F. Liver Damage and Impaired Coagulation in COVID-19 Patients: A Case Series. Diseases 2023; 11:141. [PMID: 37873785 PMCID: PMC10594514 DOI: 10.3390/diseases11040141] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/02/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has generated an unprecedented challenge for healthcare systems worldwide. Currently, the scientific community wonders if liver injury in patients suffering from severe forms is a direct consequence of the virus or secondary manifestations of systemic inflammation. The liver plays an essential role in the development of the inflammatory storm typical of this disease, and its involvement is associated with worse clinical outcomes and a higher risk of morbidity and mortality from Coronavirus disease 2019 (COVID-19). METHODS Ten patients suffering from severe COVID-19 disease who died between January 2020 and December 2021 were included in the present analysis. These subjects underwent a post mortem examination with a focused evaluation of the hepatic injury. Also, several laboratory parameters have been evaluated, with a primary focus on prothrombin time, partial thromboplastin time, fibrinogen, antithrombin III, and D-dimers to detect coagulative changes. RESULTS The main cause of death was represented by pulmonary thromboembolism events (50%). The analysis of coagulation laboratory parameters and liver biomarkers revealed a statistically significant rise in aPTT and ALP, and a decrease in albumin, when comparing the blood value at admission and death. We also found high levels of D-dimers in most of the subjects at the time of hospitalization. Interestingly, the post mortem analysis of the liver showed ample morphologic variability, with several disease features. In detail, the liver histology revealed the following: the presence of a variable degree of micro- and macrovacuolar steatosis, inflammation (also, hepato-cholangitis), and variable fibrosis. Of mention, we were also able to detect organized fibrinous material. CONCLUSIONS Our results indicate that in subjects with a severe form of COVID-19, liver disease is related to changes in coagulative and fibrinolytic pathways. In particular, we noted low fibrinogen levels and high D-dimer levels with histological liver findings. Our data suggest that fibrinogen and D-dimers may be used as prognostic markers to detect the severity of liver disease in patients with COVID-19. Finally, we underline the crucial role of coagulation balance in subjects with severe forms of COVID-19.
Collapse
Affiliation(s)
- Ludovico Abenavoli
- Department of Health Sciences, University “Magna Græcia”, Viale Europa, 88100 Catanzaro, Italy; (G.G.M.S.); (A.C.P.); (M.G.); (F.L.)
| | - Isabella Aquila
- Institute of Legal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia”, Viale Europa, 88100 Catanzaro, Italy; (I.A.); (M.A.S.); (P.R.)
| | - Matteo Antonio Sacco
- Institute of Legal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia”, Viale Europa, 88100 Catanzaro, Italy; (I.A.); (M.A.S.); (P.R.)
| | - Giuseppe Guido Maria Scarlata
- Department of Health Sciences, University “Magna Græcia”, Viale Europa, 88100 Catanzaro, Italy; (G.G.M.S.); (A.C.P.); (M.G.); (F.L.)
| | - Anna Caterina Procopio
- Department of Health Sciences, University “Magna Græcia”, Viale Europa, 88100 Catanzaro, Italy; (G.G.M.S.); (A.C.P.); (M.G.); (F.L.)
| | - Luigi Boccuto
- Healthcare Genetics and Genomics Doctoral Program, School of Nursing, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC 29631, USA;
| | - Emidio Scarpellini
- Translationeel Onderzoek van Gastroenterologische Aandoeningen (T.A.R.G.I.D.), Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Leuven, Belgium;
| | - Marta Greco
- Department of Health Sciences, University “Magna Græcia”, Viale Europa, 88100 Catanzaro, Italy; (G.G.M.S.); (A.C.P.); (M.G.); (F.L.)
| | - Daniela Patrizia Foti
- Department of Experimental and Clinical Medicine, University “Magna Græcia”, Viale Europa, 88100 Catanzaro, Italy;
| | - Pietrantonio Ricci
- Institute of Legal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia”, Viale Europa, 88100 Catanzaro, Italy; (I.A.); (M.A.S.); (P.R.)
| | - Francesco Luzza
- Department of Health Sciences, University “Magna Græcia”, Viale Europa, 88100 Catanzaro, Italy; (G.G.M.S.); (A.C.P.); (M.G.); (F.L.)
| |
Collapse
|
33
|
Lim JK, Njei B. Clinical and Histopathological Discoveries in Patients with Hepatic Injury and Cholangiopathy Who Have Died of COVID-19: Insights and Opportunities for Intervention. Hepat Med 2023; 15:151-164. [PMID: 37814605 PMCID: PMC10560482 DOI: 10.2147/hmer.s385133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/28/2023] [Indexed: 10/11/2023] Open
Abstract
The COVID-19 pandemic has had a profound impact on global health, necessitating a comprehensive understanding of its diverse manifestations. Cholangiopathy, a condition characterized by biliary dysfunction, has emerged as a significant complication in COVID-19 patients. In this review, we report the epidemiology of COVID-19, describe the hepatotropism of SARS-CoV-2, and present the histopathology of acute liver injury (ALI) in COVID-19. Additionally, we explore the relationship between pre-existing chronic liver disease and COVID-19, shedding light on the increased susceptibility of these individuals to develop cholangiopathy. Through an in-depth analysis of cholangiopathy in COVID-19 patients, we elucidate its clinical manifestations, diagnostic criteria, and underlying pathogenesis involving inflammation, immune dysregulation, and vascular changes. Furthermore, we provide a summary of studies investigating post-COVID-19 cholangiopathy, highlighting the long-term effects and potential management strategies for this condition, and discussing opportunities for intervention, including therapeutic targets, diagnostic advancements, supportive care, and future research needs.
Collapse
Affiliation(s)
- Joseph K Lim
- Yale Liver Center and Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Basile Njei
- Yale Liver Center and Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
34
|
Rose-John S, Jenkins BJ, Garbers C, Moll JM, Scheller J. Targeting IL-6 trans-signalling: past, present and future prospects. Nat Rev Immunol 2023; 23:666-681. [PMID: 37069261 PMCID: PMC10108826 DOI: 10.1038/s41577-023-00856-y] [Citation(s) in RCA: 101] [Impact Index Per Article: 101.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2023] [Indexed: 04/19/2023]
Abstract
Interleukin-6 (IL-6) is a key immunomodulatory cytokine that affects the pathogenesis of diverse diseases, including autoimmune diseases, chronic inflammatory conditions and cancer. Classical IL-6 signalling involves the binding of IL-6 to the membrane-bound IL-6 receptor α-subunit (hereafter termed 'mIL-6R') and glycoprotein 130 (gp130) signal-transducing subunit. By contrast, in IL-6 trans-signalling, complexes of IL-6 and the soluble form of IL-6 receptor (sIL-6R) signal via membrane-bound gp130. A third mode of IL-6 signalling - known as cluster signalling - involves preformed complexes of membrane-bound IL-6-mIL-6R on one cell activating gp130 subunits on target cells. Antibodies and small molecules have been developed that block all three forms of IL-6 signalling, but in the past decade, IL-6 trans-signalling has emerged as the predominant pathway by which IL-6 promotes disease pathogenesis. The first selective inhibitor of IL-6 trans-signalling, sgp130, has shown therapeutic potential in various preclinical models of disease and olamkicept, a sgp130Fc variant, had promising results in phase II clinical studies for inflammatory bowel disease. Technological developments have already led to next-generation sgp130 variants with increased affinity and selectivity towards IL-6 trans-signalling, along with indirect strategies to block IL-6 trans-signalling. Here, we summarize our current understanding of the biological outcomes of IL-6-mediated signalling and the potential for targeting this pathway in the clinic.
Collapse
Affiliation(s)
- Stefan Rose-John
- Biochemical Institute, Medical Faculty, Christian-Albrechts-University, Kiel, Germany
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GC:I3), Otto-von-Guericke-University, Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke-University, Magdeburg, Germany
| | - Jens M Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
35
|
Fornazieri MA, da Silva JLB, Gameiro JG, Scussiato HO, Ramos RAMR, Cunha BM, Figueiredo AF, Takahashi EH, Marin GA, Caetano IRDA, Meli TK, Higuchi DI, dos Santos RRP, Rampazzo ACM, Pinna FDR, Voegels RL, Doty RL. Chemosensory Dysfunction 3-Months After COVID-19, Medications and Factors Associated with Complete Recovery. Ann Otol Rhinol Laryngol 2023; 132:1177-1185. [PMID: 36482672 PMCID: PMC9742352 DOI: 10.1177/00034894221138485] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To examine the longitudinal prevalence and recovery of olfactory, gustatory, and oral chemesthetic deficits in a sizable cohort of SARS-CoV-2 infected persons using quantitative testing. To determine whether demographic and clinical factors, mainly the medications used after the COVID-19 diagnosis, influence the test measures. METHODS Prospective cohort in a hospital with primary, secondary, tertiary, and quaternary care. Patients with confirmed COVID-19 were tested during the acute infection phase (within 15 days of initial symptom, n = 187) and one (n = 113) and 3 months later (n = 73). The University of Pennsylvania Smell Identification Test, the Global Gustatory Test, and a novel test for chemesthesis were administered at all visits. RESULTS During the acute phase, 93% were anosmic or microsmic and 29.4% were hypogeusic. No one was ageusic. A deficit in oral chemesthesis was present in 13.4%. By 3 months, taste and chemesthesis had largely recovered, however, some degree of olfactory dysfunction remained in 54.8%. Remarkably, patients who had been treated with anticoagulants tended to have more olfactory improvement. Recovery was greater in men than in women, but was unrelated to disease severity, smoking behavior, or the use of various medications prior to, or during, COVID-19 infection. CONCLUSIONS When using quantitative testing, olfactory disturbances were found in nearly all SARS-CoV-2 infected patients during the acute infection phase. Taste or chemesthetic deficits were low. Olfactory impairment persisted to some degree in over half of the patients at the 3-month follow-up evaluation, being more common in women and less common in those who had been treated earlier with anticoagulants. LEVEL OF EVIDENCE 3.
Collapse
Affiliation(s)
- Marco Aurélio Fornazieri
- Londrina State University, Londrina, Paraná, Brazil
- Pontifical Catholic University of Paraná, Londrina, Brazil
- University of São Paulo, São Paulo, State of São Paulo, Brazil
- Smell and Taste Center, Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Richard L. Doty
- Smell and Taste Center, Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
36
|
Olteanu GE, Pezzuto F, Lunardi F, Fortarezza F, Dubini A, Calabrese F. Exploring the pathologist's role in understanding COVID-19: from pneumonia to long-COVID lung sequelae. Pathologica 2023; 115:275-283. [PMID: 38054902 DOI: 10.32074/1591-951x-906] [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: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 12/07/2023] Open
Abstract
The crucial role of pathologists in enhancing our understanding of SARS-CoV-2-related disease, from initial pneumonia manifestations to persistent long COVID lung symptoms, is the focus of this review. Pathological explorations have offered unprecedented insights into the early stages of severe COVID-19, shedding light on the interplay between the virus and subsequent complications, thereby shaping clinical approaches. Growing interest is directed to residual lung abnormalities of COVID-19 survivors. Although various radiological studies reported long-lasting pulmonary changes (e.g., ground glass opacities, reticulations, and bronchiectasis), the true incidence of pulmonary fibrosis and corresponding pathological findings in these patients remains largely unknown. There are a few high-impact and knowledgeable works on late complications in COVID-19 survivors, several coming from explant or autopsy cases, and rare cases from in vivo sampling. The study of biopsy samples has further deepened our knowledge of the aftermath of COVID-19 on lung tissue, uncovering alterations at the cellular level and shifts in vascular and epithelial dynamics. Despite the substantial progress made, future research is needed to devise a uniform strategy for interpreting lung biopsies, with a focus on leveraging advanced tools such as molecular and digital pathology techniques, along with artificial intelligence.
Collapse
Affiliation(s)
- Gheorghe-Emilian Olteanu
- Center for Research and Innovation in Personalized Medicine of Respiratory Diseases, Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | - Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova Medical School, Padova, Italy
- University Hospital of Padova, Padova, Italy
| | - Francesca Lunardi
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova Medical School, Padova, Italy
- University Hospital of Padova, Padova, Italy
| | | | | | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova Medical School, Padova, Italy
- University Hospital of Padova, Padova, Italy
| |
Collapse
|
37
|
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
|
38
|
Hawley HB. Long COVID: Clinical Findings, Pathology, and Endothelial Molecular Mechanisms. Am J Med 2023:S0002-9343(23)00539-9. [PMID: 37704072 DOI: 10.1016/j.amjmed.2023.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/29/2023] [Accepted: 08/08/2023] [Indexed: 09/15/2023]
Abstract
Persistence of COVID-19 symptoms may follow severe acute respiratory syndrome coronavirus 2 infection. The incidence of long COVID increases with the severity of acute disease, but even mild disease can be associated with sequelae. The symptoms vary widely, with fatigue, shortness of breath, and cognitive dysfunction the most common. Abnormalities of multiple organs have been documented, and histopathology has revealed widespread microthrombi. Elevated levels of complement are present in acute COVID-19 patients and may persist at lower levels in long COVID. Evidence supports complement activation, with endotheliopathy-associated disease as the molecular mechanism causing both acute and long COVID.
Collapse
Affiliation(s)
- H Bradford Hawley
- Department of Medicine, Wright State University Boonshoft School of Medicine, Dayton, Ohio.
| |
Collapse
|
39
|
Fiel MI, Schiano TD. Systemic Disease and the Liver-Part 1: Systemic Lupus Erythematosus, Celiac Disease, Rheumatoid Arthritis, and COVID-19. Surg Pathol Clin 2023; 16:473-484. [PMID: 37536883 DOI: 10.1016/j.path.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
The development of liver dysfunction in patients having various systemic diseases is common and has a broad differential diagnosis, at times being the initial manifestation of the disorder. Liver injury associated with systemic lupus erythematosus is heterogeneous and may present with nonspecific histology. Differentiating autoimmune hepatitis from lupus hepatitis is challenging on histologic grounds alone. Other systemic diseases that may present mostly with nonspecific findings are rheumatoid arthritis and celiac disease. More recently COVID-19 cholangiopathy and secondary sclerosing cholangitis have become increasingly recognized as distinct liver conditions. Many patients may also have intrinsic liver disease or may develop drug-induced liver injury from the treatment of the systemic disease. Timely identification of the cause of the liver dysfunction is essential and liver biopsy may help the clinician in diagnosis and management.
Collapse
Affiliation(s)
- Maria Isabel Fiel
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA
| | - Thomas D Schiano
- Division of Liver Diseases, Recanati-Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place-Box 1104, New York, NY 10029, USA.
| |
Collapse
|
40
|
Evering TH, Moser CB, Jilg N, Yeh E, Sanusi B, Wohl DA, Daar ES, Li JZ, Klekotka P, Javan AC, Eron JJ, Currier JS, Hughes MD, Smith DM, Chew KW. Long COVID After Bamlanivimab Treatment. J Infect Dis 2023; 228:S126-S135. [PMID: 37650236 PMCID: PMC10686694 DOI: 10.1093/infdis/jiad286] [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: 05/15/2023] [Accepted: 07/20/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Prospective evaluations of long COVID in outpatients with coronavirus disease 2019 (COVID-19) are lacking. We aimed to determine the frequency and predictors of long COVID after treatment with the monoclonal antibody bamlanivimab in ACTIV-2/A5401. METHODS Data were analyzed from participants who received bamlanivimab 700 mg in ACTIV-2 from October 2020 to February 2021. Long COVID was defined as the presence of self-assessed COVID symptoms at week 24. Self-assessed return to pre-COVID health was also examined. Associations were assessed by regression models. RESULTS Among 506 participants, median age was 51 years. Half were female, 5% Black/African American, and 36% Hispanic/Latino. At 24 weeks, 18% reported long COVID and 15% had not returned to pre-COVID health. Smoking (adjusted risk ratio [aRR], 2.41 [95% confidence interval {CI}, 1.34- 4.32]), female sex (aRR, 1.91 [95% CI, 1.28-2.85]), non-Hispanic ethnicity (aRR, 1.92 [95% CI, 1.19-3.13]), and presence of symptoms 22-28 days posttreatment (aRR, 2.70 [95% CI, 1.63-4.46]) were associated with long COVID, but nasal severe acute respiratory syndrome coronavirus 2 RNA was not. CONCLUSIONS Long COVID occurred despite early, effective monoclonal antibody therapy and was associated with smoking, female sex, and non-Hispanic ethnicity, but not viral burden. The strong association between symptoms 22-28 days after treatment and long COVID suggests that processes of long COVID start early and may need early intervention. CLINICAL TRIALS REGISTRATION NCT04518410.
Collapse
Affiliation(s)
- Teresa H Evering
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Carlee B Moser
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health
| | - Nikolaus Jilg
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Eunice Yeh
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health
| | - Busola Sanusi
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health
| | - David A Wohl
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill
| | - Eric S Daar
- Lundquist Institute, Harbor-UCLA Medical Center, Torrance, California
| | - Jonathan Z Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | - Joseph J Eron
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill
| | - Judith S Currier
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Michael D Hughes
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Davey M Smith
- Department of Medicine, University of California, San Diego, La Jolla
| | - Kara W Chew
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| |
Collapse
|
41
|
Reuter N, Chen X, Kropff B, Peter AS, Britt WJ, Mach M, Überla K, Thomas M. SARS-CoV-2 Spike Protein Is Capable of Inducing Cell-Cell Fusions Independent from Its Receptor ACE2 and This Activity Can Be Impaired by Furin Inhibitors or a Subset of Monoclonal Antibodies. Viruses 2023; 15:1500. [PMID: 37515187 PMCID: PMC10384293 DOI: 10.3390/v15071500] [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: 05/15/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which was responsible for the COVID-19 pandemic, efficiently spreads cell-to-cell through mechanisms facilitated by its membrane glycoprotein spike. We established a dual split protein (DSP) assay based on the complementation of GFP and luciferase to quantify the fusogenic activity of the SARS-CoV-2 spike protein. We provide several lines of evidence that the spike protein of SARS-CoV-2, but not SARS-CoV-1, induced cell-cell fusion even in the absence of its receptor, angiotensin-converting enzyme 2 (ACE2). This poorly described ACE2-independent cell fusion activity of the spike protein was strictly dependent on the proteasomal cleavage of the spike by furin while TMPRSS2 was dispensable. Previous and current variants of concern (VOCs) differed significantly in their fusogenicity. The Delta spike was extremely potent compared to Alpha, Beta, Gamma and Kappa, while the Omicron spike was almost devoid of receptor-independent fusion activity. Nonetheless, for all analyzed variants, cell fusion was dependent on furin cleavage and could be pharmacologically inhibited with CMK. Mapping studies revealed that amino acids 652-1273 conferred the ACE2-independent fusion activity of the spike. Unexpectedly, residues proximal to the furin cleavage site were not of major relevance, whereas residue 655 critically regulated fusion. Finally, we found that the spike's fusion activity in the absence of ACE2 could be inhibited by antibodies directed against its N-terminal domain (NTD) but not by antibodies targeting its receptor-binding domain (RBD). In conclusion, our BSL-1-compatible DSP assay allowed us to screen for inhibitors or antibodies that interfere with the spike's fusogenic activity and may therefore contribute to both rational vaccine design and development of novel treatment options against SARS-CoV-2.
Collapse
Affiliation(s)
- Nina Reuter
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Xiaohan Chen
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Barbara Kropff
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Antonia Sophia Peter
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - William J Britt
- Departments of Pediatrics, Microbiology and Neurobiology, Children's Hospital of Alabama, School of Medicine, University of Alabama, Birmingham, AL 35233-1771, USA
| | - Michael Mach
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Klaus Überla
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Marco Thomas
- Virologisches Institut, Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
42
|
Su P, Wu Y, Xie F, Zheng Q, Chen L, Liu Z, Meng X, Zhou F, Zhang L. A Review of Extracellular Vesicles in COVID-19 Diagnosis, Treatment, and Prevention. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206095. [PMID: 37144543 PMCID: PMC10323633 DOI: 10.1002/advs.202206095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 04/15/2023] [Indexed: 05/06/2023]
Abstract
The 2019 novel coronavirus disease (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is ongoing, and has necessitated scientific efforts in disease diagnosis, treatment, and prevention. Interestingly, extracellular vesicles (EVs) have been crucial in these developments. EVs are a collection of various nanovesicles which are delimited by a lipid bilayer. They are enriched in proteins, nucleic acids, lipids, and metabolites, and naturally released from different cells. Their natural material transport properties, inherent long-term recycling ability, excellent biocompatibility, editable targeting, and inheritance of parental cell properties make EVs one of the most promising next-generation drug delivery nanocarriers and active biologics. During the COVID-19 pandemic, many efforts have been made to exploit the payload of natural EVs for the treatment of COVID-19. Furthermore, strategies that use engineered EVs to manufacture vaccines and neutralization traps have produced excellent efficacy in animal experiments and clinical trials. Here, the recent literature on the application of EVs in COVID-19 diagnosis, treatment, damage repair, and prevention is reviewed. And the therapeutic value, application strategies, safety, and biotoxicity in the production and clinical applications of EV agents for COVID-19 treatment, as well as inspiration for using EVs to block and eliminate novel viruses are discussed.
Collapse
Affiliation(s)
- Peng Su
- Department of Breast SurgeryZhejiang Provincial People's HospitalHangzhou310014P. R. China
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Yuchen Wu
- Department of Clinical MedicineThe First School of MedicineWenzhou Medical UniversityWenzhouZhejiang325035P. R. China
| | - Feng Xie
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Qinghui Zheng
- Department of Breast SurgeryZhejiang Provincial People's HospitalHangzhou310014P. R. China
| | - Long Chen
- Center for Translational MedicineThe Affiliated Zhangjiagang Hospital of Soochow UniversityZhangjiagangJiangsu215600China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhouJiangsu215123China
| | - Xuli Meng
- Department of Breast SurgeryZhejiang Provincial People's HospitalHangzhou310014P. R. China
| | - Fangfang Zhou
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Long Zhang
- Department of Breast SurgeryZhejiang Provincial People's HospitalHangzhou310014P. R. China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058P. R. China
| |
Collapse
|
43
|
Ding Q, Zhao H. Long-term effects of SARS-CoV-2 infection on human brain and memory. Cell Death Discov 2023; 9:196. [PMID: 37380640 DOI: 10.1038/s41420-023-01512-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/05/2023] [Accepted: 06/20/2023] [Indexed: 06/30/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants have caused several waves of outbreaks. From the ancestral strain to Omicron variant, SARS-CoV-2 has evolved with the high transmissibility and increased immune escape against vaccines. Because of the multiple basic amino acids in the S1-S2 junction of spike protein, the widespread distribution of angiotensin-converting enzyme 2 (ACE2) receptor in human body and the high transmissibility, SARS-CoV-2 can infect multiple organs and has led to over 0.7 billion infectious cases. Studies showed that SARS-CoV-2 infection can cause more than 10% patients with the Long-COVID syndrome, including pathological changes in brains. This review mainly provides the molecular foundations for understanding the mechanism of SARS-CoV-2 invading human brain and the molecular basis of SARS-CoV-2 infection interfering with human brain and memory, which are associated with the immune dysfunction, syncytia-induced cell death, the persistence of SARS-CoV-2 infection, microclots and biopsychosocial aspects. We also discuss the strategies for reducing the Long-COVID syndrome. Further studies and analysis of shared researches will allow for further clarity regarding the long-term health consequences.
Collapse
Affiliation(s)
- Qiulu Ding
- School of Finance and Business, Shanghai Normal University, Shanghai, China
- School of Education, Shanghai Normal University, Shanghai, China
| | - HanJun Zhao
- Department of Microbiology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.
- Centre for Virology, Vaccinology and Therapeutics, Science Park, Hong Kong Special Administrative Region, China.
| |
Collapse
|
44
|
Ilias I, Vassiliou AG, Keskinidou C, Vrettou CS, Orfanos S, Kotanidou A, Dimopoulou I. Changes in Cortisol Secretion and Corticosteroid Receptors in COVID-19 and Non COVID-19 Critically Ill Patients with Sepsis/Septic Shock and Scope for Treatment. Biomedicines 2023; 11:1801. [PMID: 37509441 PMCID: PMC10376106 DOI: 10.3390/biomedicines11071801] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Sepsis is associated with dysregulated cortisol secretion, leading to abnormal levels of cortisol in the blood. In the early stages of the condition, cortisol levels are typically elevated due to increased secretion from the adrenal glands. However, as the disease progresses, cortisol levels may decline due to impaired adrenal function, leading to relative adrenal insufficiency. The latter is thought to be caused by a combination of factors, including impaired adrenal function, decreased production of corticotropin-releasing hormone (CRH) and adrenocorticotropic hormone (ACTH) by the hypothalamus and pituitary gland, and increased breakdown of cortisol. The dysregulation of cortisol secretion in sepsis is thought to contribute to the pathophysiology of the disease by impairing the body's ability to mount an appropriate inflammatory response. Given the dysregulation of cortisol secretion and corticosteroid receptors in sepsis, there has been considerable interest in the use of steroids as a treatment. However, clinical trials have yielded mixed results and corticosteroid use in sepsis remains controversial. In this review, we will discuss the changes in cortisol secretion and corticosteroid receptors in critically ill patients with sepsis/septic shock. We will also make special note of COVID-19 patients, who presented a recent challenge for ICU management, and explore the scope for corticosteroid administration in both COVID-19 and non-COVID-19 septic patients.
Collapse
Affiliation(s)
- Ioannis Ilias
- Department of Endocrinology, Diabetes and Metabolism, Elena Venizelou Hospital, GR-11521 Athens, Greece
| | - Alice G Vassiliou
- 1st Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, GR-10676 Athens, Greece
| | - Chrysi Keskinidou
- 1st Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, GR-10676 Athens, Greece
| | - Charikleia S Vrettou
- 1st Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, GR-10676 Athens, Greece
| | - Stylianos Orfanos
- 1st Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, GR-10676 Athens, Greece
| | - Anastasia Kotanidou
- 1st Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, GR-10676 Athens, Greece
| | - Ioanna Dimopoulou
- 1st Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, GR-10676 Athens, Greece
| |
Collapse
|
45
|
Lichtenstein B, Zheng Y, Gjertson D, Ferbas KG, Rimoin AW, Yang OO, Aldrovandi GM, Schaenman JM, Reed EF, Fulcher JA. Vascular and Non-HLA autoantibody profiles in hospitalized patients with COVID-19. Front Immunol 2023; 14:1197326. [PMID: 37398658 PMCID: PMC10309004 DOI: 10.3389/fimmu.2023.1197326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction Severe COVID-19 illness is characterized by an overwhelming immune hyperactivation. Autoantibodies against vascular, tissue, and cytokine antigens have been detected across the spectrum of COVID-19. How these autoantibodies correlate with COVID-19 severity is not fully defined. Methods We performed an exploratory study to investigate the expression of vascular and non-HLA autoantibodies in 110 hospitalized patients with COVID-19 ranging from moderate to critically ill. Relationships between autoantibodies and COVID- 19 severity and clinical risk factors were examined using logistic regression analysis. Results There were no absolute differences in levels of expression of autoantibodies against angiotensin II receptor type 1 (AT1R) or endothelial cell proteins between COVID-19 severity groups. AT1R autoantibody expression also did not differ by age, sex, or diabetes status. Using a multiplex panel of 60 non- HLA autoantigens we did identify seven autoantibodies that differed by COVID-19 severity including myosin (myosin; p=0.02), SHC-transforming protein 3 (shc3; p=0.07), peroxisome proliferator-activated receptor gamma coactivator 1-beta (perc; p=0.05), glial-cell derived neurotrophic factor (gdnf; p=0.07), enolase 1 (eno1; p=0.08), latrophilin-1 (lphn1; p=0.08), and collagen VI (coll6; p=0.05) with greater breadth and higher expression levels seen in less severe COVID-19. Discussion Overall, we found that patients hospitalized with COVID-19 demonstrate evidence of auto-reactive antibodies targeting endothelial cells, angiotensin II receptors, and numerous structural proteins including collagens. Phenotypic severity did not correlate with specific autoantibodies. This exploratory study underscores the importance of better understanding of the role of autoimmunity in COVID-19 disease and sequelae.
Collapse
Affiliation(s)
- Brian Lichtenstein
- Division of Hospital Medicine, Department of Internal Medicine, Sharp Rees-Stealy Medical Group, Sharp Healthcare, San Diego, CA, United States
| | - Ying Zheng
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - David Gjertson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, United States
| | - Kathie G. Ferbas
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Anne W. Rimoin
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, United States
| | - Otto O. Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Grace M. Aldrovandi
- Division of Infectious Diseases, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Joanna M. Schaenman
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Jennifer A. Fulcher
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- Infectious Diseases Section, Veterans Affairs (VA) Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
46
|
Otani K, Fukushima H, Matsuishi K. COVID-19 delirium and encephalopathy: Pathophysiology assumed in the first 3 years of the ongoing pandemic. BRAIN DISORDERS 2023; 10:100074. [PMID: 37056914 PMCID: PMC10076074 DOI: 10.1016/j.dscb.2023.100074] [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: 01/12/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023] Open
Abstract
Background The coronavirus disease (COVID-19) continues to spread worldwide. It has a high rate of delirium, even in young patients without comorbidities. Infected patients required isolation because of the high infectivity and virulence of COVID-19. The high prevalence of delirium in COVID-19 primarily results from encephalopathy and neuroinflammation caused by acute respiratory distress syndrome (ARDS)-associated cytokine storm. Acute respiratory distress syndrome has been linked to delirium and psychotic symptoms in the subacute phase (4 to 12 weeks), termed post-acute COVID-19 syndrome (PACS), and to brain fog, cognitive dysfunction, and fatigue, termed "long COVID," which persists beyond 12 weeks. However, no review article that mentions "COVID-19 delirium" have never been reported. Basic Procedures This narrative review summarizes data on delirium associated with acute severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and related neurological symptoms of persistent post-infection illness (PACS or long COVID) after persistence of cognitive dysfunction. Thus, we describe the pathophysiological hypothesis of COVID-19 delirium and its continuation as long COVID. This review also describes the treatment of delirium complicated by COVID-19 pneumonia. Main Findings SARS-CoV-2 infection is associated with encephalopathy and delirium. An association between COVID-19 infection and Alzheimer's disease has been suggested, and studies are being conducted from multiple facets including genetics, cytology, and postmortem study. Principal Conclusions This review suggests that COVID-19 has important short and long-term neuropsychiatric effects. Several hypotheses have been proposed that highlight potential neurobiological mechanisms as causal factors, including neuronal-inflammatory pathways by cytokine storm and cellular senescence, and chronic inflammation.
Collapse
Affiliation(s)
- Kyohei Otani
- Department of Psychiatry, Kobe City Medical Center General Hospital, 2-1-1, Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
- Department of Psychiatry, Kakogawa Central City Hospital, 439, Kakogawa-cho honmachi, Kakogawa City, Hyogo, 675-8611, Japan
| | - Haruko Fukushima
- Department of Psychiatry, Kobe City Medical Center General Hospital, 2-1-1, Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Kunitaka Matsuishi
- Department of Psychiatry, Kobe City Medical Center General Hospital, 2-1-1, Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| |
Collapse
|
47
|
Chaudhary S, Yadav RP, Kumar S, Yadav SC. Ultrastructural study confirms the formation of single and heterotypic syncytial cells in bronchoalveolar fluids of COVID-19 patients. Virol J 2023; 20:97. [PMID: 37208729 PMCID: PMC10198030 DOI: 10.1186/s12985-023-02062-7] [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] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 05/02/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND SARS-CoV-2 was reported to induce cell fusions to form multinuclear syncytia that might facilitate viral replication, dissemination, immune evasion, and inflammatory responses. In this study, we have reported the types of cells involved in syncytia formation at different stages of COVID-19 disease through electron microscopy. METHODS Bronchoalveolar fluids from the mild (n = 8, SpO2 > 95%, no hypoxia, within 2-8 days of infection), moderate (n = 8, SpO2 90% to ≤ 93% on room air, respiratory rate ≥ 24/min, breathlessness, within 9-16 days of infection), and severe (n = 8, SpO2 < 90%, respiratory rate > 30/min, external oxygen support, after 17th days of infection) COVID-19 patients were examined by PAP (cell type identification), immunofluorescence (for the level of viral infection), scanning (SEM), and transmission (TEM) electron microscopy to identify the syncytia. RESULTS Immunofluorescence studies (S protein-specific antibodies) from each syncytium indicate a very high infection level. We could not find any syncytial cells in mildly infected patients. However, identical (neutrophils or type 2 pneumocytes) and heterotypic (neutrophils-monocytes) plasma membrane initial fusion (indicating initiation of fusion) was observed under TEM in moderately infected patients. Fully matured large-size (20-100 μm) syncytial cells were found in severe acute respiratory distress syndrome (ARDS-like) patients of neutrophils, monocytes, and macrophage origin under SEM. CONCLUSIONS This ultrastructural study on the syncytial cells from COVID-19 patients sheds light on the disease's stages and types of cells involved in the syncytia formations. Syncytia formation was first induced in type II pneumocytes by homotypic fusion and later with haematopoetic cells (monocyte and neutrophils) by heterotypic fusion in the moderate stage (9-16 days) of the disease. Matured syncytia were reported in the late phase of the disease and formed large giant cells of 20 to 100 μm.
Collapse
Affiliation(s)
- Shikha Chaudhary
- Electron Microscope Facility, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Ravi P Yadav
- Electron Microscope Facility, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Shailendra Kumar
- Department of Anaesthesiology, Pain Medicine and Critical Care, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Subhash Chandra Yadav
- Electron Microscope Facility, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India.
| |
Collapse
|
48
|
Lebrun L, Absil L, Remmelink M, De Mendonça R, D'Haene N, Gaspard N, Rusu S, Racu ML, Collin A, Allard J, Zindy E, Schiavo AA, De Clercq S, De Witte O, Decaestecker C, Lopes MB, Salmon I. SARS-Cov-2 infection and neuropathological findings: a report of 18 cases and review of the literature. Acta Neuropathol Commun 2023; 11:78. [PMID: 37165453 PMCID: PMC10170054 DOI: 10.1186/s40478-023-01566-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/15/2023] [Indexed: 05/12/2023] Open
Abstract
INTRODUCTION COVID-19-infected patients harbour neurological symptoms such as stroke and anosmia, leading to the hypothesis that there is direct invasion of the central nervous system (CNS) by SARS-CoV-2. Several studies have reported the neuropathological examination of brain samples from patients who died from COVID-19. However, there is still sparse evidence of virus replication in the human brain, suggesting that neurologic symptoms could be related to mechanisms other than CNS infection by the virus. Our objective was to provide an extensive review of the literature on the neuropathological findings of postmortem brain samples from patients who died from COVID-19 and to report our own experience with 18 postmortem brain samples. MATERIAL AND METHODS We used microscopic examination, immunohistochemistry (using two different antibodies) and PCR-based techniques to describe the neuropathological findings and the presence of SARS-CoV-2 virus in postmortem brain samples. For comparison, similar techniques (IHC and PCR) were applied to the lung tissue samples for each patient from our cohort. The systematic literature review was conducted from the beginning of the pandemic in 2019 until June 1st, 2022. RESULTS In our cohort, the most common neuropathological findings were perivascular haemosiderin-laden macrophages and hypoxic-ischaemic changes in neurons, which were found in all cases (n = 18). Only one brain tissue sample harboured SARS-CoV-2 viral spike and nucleocapsid protein expression, while all brain cases harboured SARS-CoV-2 RNA positivity by PCR. A colocalization immunohistochemistry study revealed that SARS-CoV-2 antigens could be located in brain perivascular macrophages. The literature review highlighted that the most frequent neuropathological findings were ischaemic and haemorrhagic lesions, including hypoxic/ischaemic alterations. However, few studies have confirmed the presence of SARS-CoV-2 antigens in brain tissue samples. CONCLUSION This study highlighted the lack of specific neuropathological alterations in COVID-19-infected patients. There is still no evidence of neurotropism for SARS-CoV-2 in our cohort or in the literature.
Collapse
Affiliation(s)
- Laetitia Lebrun
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB)Hôpital Universitaire de Bruxelles (HUB), CUB Hôpital Erasme, 808 Route de Lennik, B-1070, Brussels, Belgium
| | - Lara Absil
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB)Hôpital Universitaire de Bruxelles (HUB), CUB Hôpital Erasme, 808 Route de Lennik, B-1070, Brussels, Belgium
| | - Myriam Remmelink
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB)Hôpital Universitaire de Bruxelles (HUB), CUB Hôpital Erasme, 808 Route de Lennik, B-1070, Brussels, Belgium
| | - Ricardo De Mendonça
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB)Hôpital Universitaire de Bruxelles (HUB), CUB Hôpital Erasme, 808 Route de Lennik, B-1070, Brussels, Belgium
| | - Nicky D'Haene
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB)Hôpital Universitaire de Bruxelles (HUB), CUB Hôpital Erasme, 808 Route de Lennik, B-1070, Brussels, Belgium
| | - Nicolas Gaspard
- Department of Neurology, Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (HUB), CUB Hôpital Erasme, Erasme University Hospital, Brussels, Belgium
| | - Stefan Rusu
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB)Hôpital Universitaire de Bruxelles (HUB), CUB Hôpital Erasme, 808 Route de Lennik, B-1070, Brussels, Belgium
| | - Marie-Lucie Racu
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB)Hôpital Universitaire de Bruxelles (HUB), CUB Hôpital Erasme, 808 Route de Lennik, B-1070, Brussels, Belgium
| | - Amandine Collin
- DIAPath, Center for Microscopy and Molecular Imaging (CMMI), ULB, Gosselies, Belgium
| | - Justine Allard
- DIAPath, Center for Microscopy and Molecular Imaging (CMMI), ULB, Gosselies, Belgium
| | - Egor Zindy
- DIAPath, Center for Microscopy and Molecular Imaging (CMMI), ULB, Gosselies, Belgium
| | - Andrea Alex Schiavo
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB)Hôpital Universitaire de Bruxelles (HUB), CUB Hôpital Erasme, 808 Route de Lennik, B-1070, Brussels, Belgium
| | - Sarah De Clercq
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB)Hôpital Universitaire de Bruxelles (HUB), CUB Hôpital Erasme, 808 Route de Lennik, B-1070, Brussels, Belgium
| | - Olivier De Witte
- Department of Neurosurgery, Université Libre de Bruxelles (ULB)Hôpital Universitaire de Bruxelles (HUB), CUB Hôpital ErasmeErasme University Hospital, Brussels, Belgium
| | - Christine Decaestecker
- DIAPath, Center for Microscopy and Molecular Imaging (CMMI), ULB, Gosselies, Belgium
- Laboratory of Image Synthesis and Analysis, Brussels School of Engineering/École Polytechnique de Brussels, ULB, Brussels, Belgium
| | - Maria-Beatriz Lopes
- Department of Pathology, University of Virginia Health System, Charlottesville, VA, USA
| | - Isabelle Salmon
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles (ULB)Hôpital Universitaire de Bruxelles (HUB), CUB Hôpital Erasme, 808 Route de Lennik, B-1070, Brussels, Belgium.
- DIAPath, Center for Microscopy and Molecular Imaging (CMMI), ULB, Gosselies, Belgium.
| |
Collapse
|
49
|
Bommarito G, Garibotto V, Frisoni GB, Assal F, Lalive PH, Allali G. The Two-Way Route between Delirium Disorder and Dementia: Insights from COVID-19. NEURODEGENER DIS 2023; 22:91-103. [PMID: 37054684 DOI: 10.1159/000530566] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/23/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND Delirium disorder is a frequent neurological complication of SARS-CoV-2 infection and associated with increased disease severity and mortality. Cognitive impairment is a major risk factor for developing delirium disorder during COVID-19, which, in turn, increases the risk of subsequent neurological complications and cognitive decline. SUMMARY The bidirectional connection between delirium disorder and dementia likely resides at multiple levels, and its pathophysiological mechanisms during COVID-19 include endothelial damage, blood-brain barrier dysfunction, and local inflammation, with activation of microglia and astrocytes. Here, we describe the putative pathogenic pathways underlying delirium disorder during COVID-19 and highlight how they cross with the ones leading to neurodegenerative dementia. KEY MESSAGES The analysis of the two-sided link can offer useful insights for confronting with long-term neurological consequences of COVID-19 and framing future prevention and early treatment strategies.
Collapse
Affiliation(s)
- Giulia Bommarito
- Department of Clinical Neurosciences, Lausanne University Hospitals and University of Lausanne, Lausanne, Switzerland
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospitals and NIMTlab, University of Geneva, Geneva, Switzerland
| | - Giovanni B Frisoni
- Memory Center and LANVIE-Laboratory of Neuroimaging of Aging, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Frédéric Assal
- Department of Clinical Neurosciences, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Patrice H Lalive
- Department of Clinical Neurosciences, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
| | - Gilles Allali
- Department of Clinical Neurosciences, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
- Division of Cognitive and Motor Aging, Department of Neurology, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York, USA
- Leenaards Memory Center, Lausanne University Hospitals and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
50
|
Solomon IH, Singh A, Folkerth RD, Mukerji SS. What Can We Still Learn from Brain Autopsies in COVID-19? Semin Neurol 2023. [PMID: 37023787 DOI: 10.1055/s-0043-1767716] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Neuropathological findings have been published from ∼900 patients who died with or from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, representing less than 0.01% of the close to 6.4 million deaths reported to the World Health Organization 2 years into the coronavirus disease 2019 (COVID-19) pandemic. In this review, we extend our prior work summarizing COVID-19 neuropathology by including information on published autopsies up to June 2022, and neuropathological studies in children, COVID-19 variants, secondary brain infections, ex vivo brain imaging, and autopsies performed in countries outside of the United States or Europe. We also summarize research studies that investigate mechanisms of neuropathogenesis in nonhuman primates and other models. While a pattern of cerebrovascular pathology and microglial-predominant inflammation remains the primary COVID-19-associated neuropathological finding, there is no singular understanding of the mechanisms that underlie neurological symptoms in acute COVID-19 or the post-acute COVID-19 condition. Thus, it is paramount that we incorporate microscopic and molecular findings from brain tissue into what we know about the clinical disease so that we attain best practice guidance and direct research priorities for the study of the neurological morbidity of COVID-19.
Collapse
Affiliation(s)
- Isaac H Solomon
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Arjun Singh
- Division of Neuroimmunology and Neuro-Infectious Diseases, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts
- Healing Hospital, Chandigarh, India
| | - Rebecca D Folkerth
- Office of Chief Medical Examiner and Department of Forensic Medicine, New York University School of Medicine, New York, New York
| | - Shibani S Mukerji
- Harvard Medical School, Boston, Massachusetts
- Division of Neuroimmunology and Neuro-Infectious Diseases, Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts
- Division of Infectious Diseases, Vaccine and Immunotherapy Center, Massachusetts General Hospital, Charlestown, Massachusetts
| |
Collapse
|