1
|
Martín-Faivre L, Prince L, Cornu C, Villeret B, Sanchez-Guzman D, Rouzet F, Sallenave JM, Garcia-Verdugo I. Pulmonary delivery of silver nanoparticles prevents influenza infection by recruiting and activating lymphoid cells. Biomaterials 2025; 312:122721. [PMID: 39106817 DOI: 10.1016/j.biomaterials.2024.122721] [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: 04/16/2024] [Revised: 07/12/2024] [Accepted: 07/27/2024] [Indexed: 08/09/2024]
Abstract
Silver nanoparticles (AgNPs) are a potential antiviral agent due to their ability to disrupt the viral particle or alter the virus metabolism inside the host cell. In vitro, AgNPs exhibit antiviral activity against the most common human respiratory viruses. However, their capacity to modulate immune responses during respiratory viral infections has yet to be explored. This study demonstrates that administering AgNPs directly into the lungs prior to infection can reduce viral loads and therefore virus-induced cytokines in mice infected with influenza virus or murine pneumonia virus. The prophylactic effect was diminished in mice with depleted lymphoid cells. We showed that AgNPs-treatment resulted in the recruitment and activation of lymphocytes in the lungs, particularly natural killer (NK) cells. Mechanistically, AgNPs enhanced the ability of alveolar macrophages to promote both NK cell migration and IFN-γ production. By contrast, following infection, in mice treated with AgNPs, NK cells exhibited decreased activation, indicating that these nanoparticles can regulate the potentially deleterious activation of these cells. Overall, the data suggest that AgNPs may possess prophylactic antiviral properties by recruiting and controlling the activation of lymphoid cells through interaction with alveolar macrophages.
Collapse
Affiliation(s)
- Lydie Martín-Faivre
- Université Paris Cité, Inflamex Excellence Laboratory, INSERM UMR-1152-PHERE, F-75018, Paris, France
| | - Lisa Prince
- Université Paris Cité, Inflamex Excellence Laboratory, INSERM UMR-1152-PHERE, F-75018, Paris, France
| | - Clémentine Cornu
- Université Paris Cité, Inflamex Excellence Laboratory, INSERM UMR-1152-PHERE, F-75018, Paris, France
| | - Bérengère Villeret
- Université Paris Cité, Inflamex Excellence Laboratory, INSERM UMR-1152-PHERE, F-75018, Paris, France
| | - Daniel Sanchez-Guzman
- Université Paris Cité, Inflamex Excellence Laboratory, INSERM UMR-1152-PHERE, F-75018, Paris, France
| | - François Rouzet
- Nuclear Medicine Department, Hôpital Bichat-Claude Bernard, AP-HP, Université Paris Cité and Inserm U1148, F-75018, Paris, France
| | - Jean-Michel Sallenave
- Université Paris Cité, Inflamex Excellence Laboratory, INSERM UMR-1152-PHERE, F-75018, Paris, France
| | - Ignacio Garcia-Verdugo
- Université Paris Cité, Inflamex Excellence Laboratory, INSERM UMR-1152-PHERE, F-75018, Paris, France.
| |
Collapse
|
2
|
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
|
3
|
Li M, Bei ZC, Yuan Y, Wang B, Zhang D, Xu L, Zhao L, Xu Q, Song Y. In-cell bioluminescence resonance energy transfer (BRET)-based assay uncovers ceritinib and CA-074 as SARS-CoV-2 papain-like protease inhibitors. J Enzyme Inhib Med Chem 2024; 39:2387417. [PMID: 39163165 PMCID: PMC11338211 DOI: 10.1080/14756366.2024.2387417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024] Open
Abstract
Papain-like protease (PLpro) is an attractive anti-coronavirus target. The development of PLpro inhibitors, however, is hampered by the limitations of the existing PLpro assay and the scarcity of validated active compounds. We developed a novel in-cell PLpro assay based on BRET and used it to evaluate and discover SARS-CoV-2 PLpro inhibitors. The developed assay demonstrated remarkable sensitivity for detecting the reduction of intracellular PLpro activity while presenting high reliability and performance for inhibitor evaluation and high-throughput screening. Using this assay, three protease inhibitors were identified as novel PLpro inhibitors that are structurally disparate from those previously known. Subsequent enzymatic assays and ligand-protein interaction analysis based on molecular docking revealed that ceritinib directly inhibited PLpro, showing high geometric complementarity with the substrate-binding pocket in PLpro, whereas CA-074 methyl ester underwent intracellular hydrolysis, exposing a free carboxyhydroxyl group essential for hydrogen bonding with G266 in the BL2 groove, resulting in PLpro inhibition.
Collapse
Affiliation(s)
- Mei Li
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Zhu-Chun Bei
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Yongtian Yuan
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Baogang Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Dongna Zhang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Likun Xu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Liangliang Zhao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Qin Xu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yabin Song
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Hoste ACR, Smeralda W, Cugnet A, Brostaux Y, Deleu M, Garigliany M, Jacques P. The structure of lipopeptides impacts their antiviral activity and mode of action against SARS-CoV-2 in vitro. Appl Environ Microbiol 2024:e0103624. [PMID: 39445780 DOI: 10.1128/aem.01036-24] [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: 05/25/2024] [Accepted: 08/28/2024] [Indexed: 10/25/2024] Open
Abstract
Microbial lipopeptides are synthesized by nonribosomal peptide synthetases and are composed of a hydrophobic fatty acid chain and a hydrophilic peptide moiety. These structurally diverse amphiphilic molecules can interact with biological membranes and possess various biological activities, including antiviral properties. This study aimed to evaluate the cytotoxicity and antiviral activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) of 15 diverse lipopeptides to understand their structure-activity relationships. Non-ionic lipopeptides were generally more cytotoxic than charged ones, with cationic lipopeptides being less cytotoxic than anionic and non-ionic variants. At 100 µg/mL, six lipopeptides reduced SARS-CoV-2 RNA to undetectable levels in infected Vero E6 cells, while six others achieved a 2.5- to 4.1-log reduction, and three had no significant effect. Surfactin, white line-inducing principle (WLIP), fengycin, and caspofungin emerged as the most promising anti-SARS-CoV-2 agents. Detailed analysis revealed that these four lipopeptides affected various stages of the viral life cycle involving the viral envelope. Surfactin and WLIP significantly reduced viral RNA levels in replication assays, comparable to neutralizing serum. Surfactin uniquely inhibited viral budding, while fengycin impacted viral binding after pre-infection treatment of the cells. Caspofungin demonstrated a lower antiviral effect compared to the others. Key structural traits of lipopeptides influencing their cytotoxic and antiviral activities were identified. Lipopeptides with a high number of amino acids, especially charged (preferentially anionic) amino acids, showed potent anti-SARS-CoV-2 activity. This research paves the way for designing new lipopeptides with low cytotoxicity and high antiviral efficacy, potentially leading to effective treatments. IMPORTANCE This study advances our understanding of how lipopeptides, which are molecules mostly produced by bacteria, with both fat and protein components, can be used to fight viruses like severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). By analyzing 15 different lipopeptides, researchers identified key structural features that make some of these molecules particularly effective at reducing viral levels while being less harmful to cells. Specifically, lipopeptides with certain charged amino acids were found to have the strongest antiviral effects. This research lays the groundwork for developing new antiviral treatments that are both potent against viruses and safe for human cells, offering hope for better therapeutic options in the future.
Collapse
Affiliation(s)
- Alexis C R Hoste
- MiPI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
- Veterinary Pathology, FARAH Research Centre, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Willy Smeralda
- LBMI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Aurélien Cugnet
- MiPI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Yves Brostaux
- Applied Statistics, Computer Science and Modelling laboratory, TERRA Teaching and Research Centre, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Magali Deleu
- LBMI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Mutien Garigliany
- Veterinary Pathology, FARAH Research Centre, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Philippe Jacques
- MiPI, TERRA Teaching and Research Centre, Joint Research Unit BioEcoAgro, UMRt 1158, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| |
Collapse
|
5
|
Nemethova V, Babiakova P, Selc M, Jakic K, Uhelska L, Teglasova B, Makovicky P, Babelova A, Razga F. Therapeutic oligonucleotide ASC1R shows excellent tolerability and remarkable efficacy in reducing SARS-CoV-2 mRNA levels in C57BL/6 mice. Biomed Pharmacother 2024; 180:117587. [PMID: 39442238 DOI: 10.1016/j.biopha.2024.117587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/01/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
The coronavirus pandemic has resulted in over 775 million cases and 7 million deaths worldwide, driving efforts to develop therapeutic strategies to control the viral infection. Therapeutic oligonucleotides have shown promise in treating many pathological conditions, including those of viral origin. The present study assessed the in vivo efficacy and safety of ASC1R, a novel therapeutic oligonucleotide of unconventional design targeting the conserved viral RdRp sequence essential for replication. In functional studies, ASC1R was administered to transfected C57BL/6 mice at doses of 1 and 10 mg/kg. Safety assessments included acute toxicity evaluations at doses ranging from 30 to 100 mg/kg, and subacute toxicity evaluations of repeated doses of 1 and 10 mg/kg. Evaluations included general clinical observations, findings at necropsy, measurements of organ weight, and histopathological examinations of the liver, lungs, spleen, and kidneys. ASC1R effectively reduced RdRp levels >94 % within 24 hours following a single 1 mg/kg dose, with no observed organ toxicity. Acute and subacute toxicity assessments found that mice receiving high (≥30 mg/kg) or repeated (10 mg/kg for 7 days) doses of ASC1R showed an increase in relative spleen weight, without histopathological changes. The marked ability of a single low dose of ASC1R (1 mg/kg) to reduce viral RNA suggests its potential for clinical applications, balancing therapeutic efficacy with minimal side effects. Our findings indicate that ASC1R has promise as a viable treatment option for patients with COVID-19.
Collapse
Affiliation(s)
| | | | - Michal Selc
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava 84505, Slovakia; Centre for Advanced Materials Application, Slovak Academy of Sciences, Bratislava 84511, Slovakia
| | - Kristina Jakic
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava 84505, Slovakia
| | | | | | - Peter Makovicky
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava 84505, Slovakia
| | - Andrea Babelova
- Department of Nanobiology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava 84505, Slovakia; Centre for Advanced Materials Application, Slovak Academy of Sciences, Bratislava 84511, Slovakia
| | - Filip Razga
- Selecta Biotech SE, Bratislava 84107, Slovakia
| |
Collapse
|
6
|
Wang Z, Li Z, Shen Y, Qian S, Tang M, He J, Lu H, Zhang N. Long-term effects of COVID-19 infection on bone mineral density. J Glob Health 2024; 14:05029. [PMID: 39421935 PMCID: PMC11487469 DOI: 10.7189/jogh.14.05029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024] Open
Abstract
Background In this study, we aimed to identify bone mineral density (BMD) trajectories of hospitalised patients with coronavirus disease 2019 (COVID-19) and to determine the prognostic role of the trajectory groups. Methods This is a retrospective study of hospitalised patients with COVID-19 treated in our hospital from November 2022 to February 2023. BMD was manually measured from the thoracic 12 (T12) and lumbar one (L1) vertebra using chest computed tomography images. We constructed group trajectory models using group-based trajectory modelling. We performed the logistic regression analysis to associate the BMD trajectory pattern with clinical outcomes. Results This study included 1767 patients. The mean follow-up time after discharge was 181.5 days (standard deviation (SD) = 9.7). There were 1137 (64.3%) male patients, and more than 80% of patients were aged >60 years. We successfully identified three latent BMD trajectories to reveal the dynamic effects of COVID-19 infection on bone health in patients, namely, the early low-normal decline group, the average, and the early high-rapid decline group. All groups demonstrated consistent overall declining trends. A significant association was observed between BMD trajectory pattern (T12 or L1) and baseline characteristics of sex, age, and penetrating keratoplasty (P < 0.05). Our study showed that the BMD trajectories were significantly associated with mortality. Furthermore, we found that these trajectories were also associated with the length of hospital stay. Conclusions This study provided evidence for the COVID-19 process to bone health, as well as evidence on strengthening bone health management before and after COVID-19 infection. BMD trajectories may help manage bone health and guide treatment in patients with COVID-19.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Orthopaedics Research Institute of Zhejiang University, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Zhejiang, China
- Zhejiang Provincial Clinical Medical Research Centre for Motor System Diseases, Zhejiang, China
- International Chinese Musculoskeletal Research Society, Zhejiang, China
| | - Zilong Li
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Orthopaedics Research Institute of Zhejiang University, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Zhejiang, China
- Zhejiang Provincial Clinical Medical Research Centre for Motor System Diseases, Zhejiang, China
- International Chinese Musculoskeletal Research Society, Zhejiang, China
| | - Yechao Shen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Orthopaedics Research Institute of Zhejiang University, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Zhejiang, China
- Zhejiang Provincial Clinical Medical Research Centre for Motor System Diseases, Zhejiang, China
- International Chinese Musculoskeletal Research Society, Zhejiang, China
| | - Shengjun Qian
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Orthopaedics Research Institute of Zhejiang University, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Zhejiang, China
- Zhejiang Provincial Clinical Medical Research Centre for Motor System Diseases, Zhejiang, China
- International Chinese Musculoskeletal Research Society, Zhejiang, China
| | - Mengling Tang
- Department of Epidemiology and Biostatistics, Zhejiang University School of Public Health, Zhejiang, China
| | - Jiaming He
- First Affiliated Hospital of Xian Jiaotong University, Shaanxi, China
| | - Haoda Lu
- Bioinformatics Institute, A*STAR, Singapore
| | - Ning Zhang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
- Orthopaedics Research Institute of Zhejiang University, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Zhejiang, China
- Zhejiang Provincial Clinical Medical Research Centre for Motor System Diseases, Zhejiang, China
- International Chinese Musculoskeletal Research Society, Zhejiang, China
| |
Collapse
|
7
|
Murdocca M, Andrade Santos-Filho O, De Masi C, Dos Santos Rodrigues E, Campos de Souza CV, De Santis R, Amatore D, Latini A, Schipani R, di Rienzo Businco L, Brandimarte B, Grilli G, Huang TL, Mayence AS, Lista F, Duranti A, Sangiuolo F, Vanden Eynde JJ, Novelli G. Characterization of the symmetrical benzimidazole twin drug TL1228: the role as viral entry inhibitor for fighting COVID-19. Biol Direct 2024; 19:93. [PMID: 39415197 PMCID: PMC11481581 DOI: 10.1186/s13062-024-00523-9] [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: 08/14/2024] [Accepted: 08/28/2024] [Indexed: 10/18/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is reliably one of the largest pandemics the world has suffered in recent years. In the search for non-biological antivirals, special emphasis was placed on drug repurposing to accelerate the clinical implementation of effective drugs.The life cycle of the virus has been extensively investigated and many human targets have been identified, such as the molecular chaperone GRP78, representing a host auxiliary factor for SARS-CoV-2 entry. Here we report the inhibitor capacity of TL1228, a small molecule discovered through an in silico screening approach, which could interfere with the interaction of SARS-CoV-2 and its target cells, blocking the recognition of the GRP78 cellular receptor by the viral Spike protein. TL1228 showed in vitro the ability to reduce significantly both pseudoviral and authentic viral activity even through the reduction of GRP78/ACE2 transcript levels. Importantly, TL1228 acts in modulating expression levels of innate immunity and as inflammation markers.
Collapse
Affiliation(s)
- Michela Murdocca
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Osvaldo Andrade Santos-Filho
- Center of Health Sciences Laboratory of Molecular Modelling & Computational Strutural Biology Cidade Universitária, Federal University of Rio de Janeiro IPPN, Av. Carlos Chagas Filho373, Bloco H, Rio de Janeiro, 21941-599, RJ, Brazil
| | - Claudia De Masi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Edivaldo Dos Santos Rodrigues
- Center of Health Sciences Laboratory of Molecular Modelling & Computational Strutural Biology Cidade Universitária, Federal University of Rio de Janeiro IPPN, Av. Carlos Chagas Filho373, Bloco H, Rio de Janeiro, 21941-599, RJ, Brazil
| | - Claudia Valeria Campos de Souza
- Center of Health Sciences Laboratory of Molecular Modelling & Computational Strutural Biology Cidade Universitária, Federal University of Rio de Janeiro IPPN, Av. Carlos Chagas Filho373, Bloco H, Rio de Janeiro, 21941-599, RJ, Brazil
| | - Riccardo De Santis
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy
- Defence Institute for Biomedical Sciences, Rome, 00184, Italy
| | | | - Andrea Latini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Rossella Schipani
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Lino di Rienzo Businco
- Otorhinolaryngology Department, Institute of Sport Medicine and Science CONI, Rome, Italy
| | - Bruno Brandimarte
- Electronic Measurements Physics Department, Sapienza University, Rome, Italy
| | - Giorgia Grilli
- Defence Institute for Biomedical Sciences, Rome, 00184, Italy
| | - Tien L Huang
- Formerly Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA, 70125, USA
| | - Annie S Mayence
- Formerly Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, 1 Drexel Drive, New Orleans, LA, 70125, USA
| | - Florigio Lista
- Defence Institute for Biomedical Sciences, Rome, 00184, Italy
| | - Andrea Duranti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, 61029, Italy
| | - Federica Sangiuolo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| | - Jean Jacques Vanden Eynde
- Formerly Department of Organic Chemistry (FS), University of Mons-UMONS, 1 place du Parc, Mons, 7000, Belgium
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
8
|
Colaneri M, Fama F, Fassio F, Holmes D, Scaglione G, Mariani C, Galli L, Lai A, Antinori S, Gori A, Riva A, Schiavini M. Impact of Early Antiviral Therapy on SARS-CoV-2 Clearance Time in High-Risk COVID-19 subjects: A Propensity Score Matching Study. Int J Infect Dis 2024:107265. [PMID: 39393523 DOI: 10.1016/j.ijid.2024.107265] [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: 05/02/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Effective treatments for COVID-19 are needed to mitigate disease progression and reduce the burden on healthcare systems. This study investigates the impact of early treatments on SARS-CoV-2 viral shedding duration among high-risk individuals with mild symptoms. METHODS A single-centre, retrospective observational study was conducted at Luigi Sacco Hospital in Milan from December 2021 to March 2023. Hospitalized and non-hospitalized adults with a confirmed SARS-CoV-2 infection and at high-risk of disease progression were enrolled. Unadjusted and adjusted negative binomial regression models and a Random Forest regression model were performed before and after matching subjects based on their propensity of being treated or not. RESULTS Results from 518 subjects (428 treated and 90 untreated) revealed a significant reduction in SARS-CoV-2 viral shedding duration among those who received early treatment compared to untreated individuals. Propensity score matching and multivariable regression analyses confirmed this finding. Early treatment significantly reduced the risk of COVID-19-related hospitalization and pneumonia development. Subgroup analysis identified COPD as a potential factor influencing effectiveness of early treatments. CONCLUSIONS Early treatments play a crucial role in reducing SARS-CoV-2 viral shedding and preventing disease progression among high-risk individuals. Shorter viral shedding duration also contributes to improved healthcare resource utilization and infection control measures.
Collapse
Affiliation(s)
- Marta Colaneri
- Infectious Diseases and Immunopathology, Department of Clinical Sciences, University of Milan, Luigi Sacco Hospital, Milan, Italy.; Centre for Multidisciplinary Research in Health Science (MACH), University of Milan, Milan, Italy
| | - Federico Fama
- Infectious Diseases and Immunopathology, Department of Clinical Sciences, University of Milan, Luigi Sacco Hospital, Milan, Italy.; Centre for Multidisciplinary Research in Health Science (MACH), University of Milan, Milan, Italy..
| | - Federico Fassio
- Department of Public Health, Experimental and Forensic Medicine, Unit of Biostatistics and Clinical Epidemiology, University of Pavia, Pavia, Italy
| | - Darcy Holmes
- Infectious Disease Unit Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Giovanni Scaglione
- Infectious Diseases and Immunopathology, Department of Clinical Sciences, University of Milan, Luigi Sacco Hospital, Milan, Italy
| | - Chiara Mariani
- Infectious Diseases and Immunopathology, Department of Clinical Sciences, University of Milan, Luigi Sacco Hospital, Milan, Italy
| | - Lucia Galli
- Infectious Diseases and Immunopathology, Department of Clinical Sciences, University of Milan, Luigi Sacco Hospital, Milan, Italy
| | - Alessia Lai
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Spinello Antinori
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.; III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Andrea Gori
- Infectious Diseases and Immunopathology, Department of Clinical Sciences, University of Milan, Luigi Sacco Hospital, Milan, Italy.; Centre for Multidisciplinary Research in Health Science (MACH), University of Milan, Milan, Italy.; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.; III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Agostino Riva
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.; Infectious Diseases Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Monica Schiavini
- Infectious Diseases and Immunopathology, Department of Clinical Sciences, University of Milan, Luigi Sacco Hospital, Milan, Italy
| |
Collapse
|
9
|
Suri C, Pande B, Sahithi LS, Sahu T, Verma HK. Interplay between Lung Diseases and Viral Infections: A Comprehensive Review. Microorganisms 2024; 12:2030. [PMID: 39458339 PMCID: PMC11510474 DOI: 10.3390/microorganisms12102030] [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: 08/25/2024] [Revised: 09/16/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
The intricate relationship between chronic lung diseases and viral infections is a significant concern in respiratory medicine. We explore how pre-existing lung conditions, including chronic obstructive pulmonary disease, asthma, and interstitial lung diseases, influence susceptibility, severity, and outcomes of viral infections. We also examine how viral infections exacerbate and accelerate the progression of lung disease by disrupting immune responses and triggering inflammatory pathways. By summarizing current evidence, this review highlights the bidirectional nature of these interactions, where underlying lung diseasesincrease vulnerability to viral infections, while these infections, in turn, worsen the clinical course. This review underscores the importance of preventive measures, such as vaccination, early detection, and targeted therapies, to mitigate adverse outcomes in patients with chronic lung conditions. The insights provided aim to inform clinical strategies that can improve patient management and reduce the burden of chronic lung diseases exacerbated by viral infections.
Collapse
Affiliation(s)
- Chahat Suri
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
| | - Babita Pande
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | | | - Tarun Sahu
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lungs Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, 85764 Munich, Germany
| |
Collapse
|
10
|
De Clercq E. A scientific career from the early 1960s till 2023: A tale of the various protagonists. Biochem Pharmacol 2024; 228:116248. [PMID: 38701868 DOI: 10.1016/j.bcp.2024.116248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
In this era spanning more than 60 years (from the early 1960s till today (2023), a broad variety of actors played a decisive role: Piet De Somer, Tom C. Merigan, Paul A. Janssen, Maurice Hilleman, and Georges Smets. Two protagonists (Antonín Holý and John C. Martin) formed with me a unique triangle (the Holý Trinity). Walter Fiers' group (with the help of Jean Content) contributed to the cloning of human β-interferon, and Piet Herdewijn accomplished the chemical synthesis of an array of anti-HIV 2',3'-dideoxynucleoside analogues. Rudi Pauwels, Masanori Baba, Dominique Schols, Johan Neyts, Lieve Naesens, Anita Van Lierde, Graciela Andrei, Robert Snoeck and Dirk Daelemans, as members of my team, helped me in achieving the intended goal, the development of a selective therapy for virus infections. The collaboration with "Lowie" (Guangdi Li) generated a new dimension for the future.
Collapse
Affiliation(s)
- Erik De Clercq
- KU Leuven, Rega Institute for Medical Research, Herestraat 49, B-3000 Leuven, Belgium.
| |
Collapse
|
11
|
Jaishwal P, Jha K, Singh SP. Revisiting the dimensions of universal vaccine with special focus on COVID-19: Efficacy versus methods of designing. Int J Biol Macromol 2024; 277:134012. [PMID: 39048013 DOI: 10.1016/j.ijbiomac.2024.134012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 05/28/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Even though the use of SARS-CoV-2 vaccines during the COVID-19 pandemic showed unprecedented success in a short time, it also exposed a flaw in the current vaccine design strategy to offer broad protection against emerging variants of concern. However, developing broad-spectrum vaccines is still a challenge for immunologists. The development of universal vaccines against emerging pathogens and their variants appears to be a practical solution to mitigate the economic and physical effects of the pandemic on society. Very few reports are available to explain the basic concept of universal vaccine design and development. This review provides an overview of the innate and adaptive immune responses generated against vaccination and essential insight into immune mechanisms helpful in designing universal vaccines targeting influenza viruses and coronaviruses. In addition, the characteristics, safety, and factors affecting the efficacy of universal vaccines have been discussed. Furthermore, several advancements in methods worthy of designing universal vaccines are described, including chimeric immunogens, heterologous prime-boost vaccines, reverse vaccinology, structure-based antigen design, pan-reactive antibody vaccines, conserved neutralizing epitope-based vaccines, mosaic nanoparticle-based vaccines, etc. In addition to the several advantages, significant potential constraints, such as defocusing the immune response and subdominance, are also discussed.
Collapse
Affiliation(s)
- Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | - Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | | |
Collapse
|
12
|
Latarissa IR, Rendrayani F, Iftinan GN, Suhandi C, Meiliana A, Sormin IP, Barliana MI, Lestari K. The Efficacy of Oral/Intravenous Corticosteroid Use in COVID-19 Patients: A Systematic Review. J Exp Pharmacol 2024; 16:321-337. [PMID: 39371262 PMCID: PMC11453156 DOI: 10.2147/jep.s484596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024] Open
Abstract
The COVID-19 pandemic is prompting extensive investigation into potential treatments, including the use of corticosteroids to manage inflammation and mitigate severe disease outcomes. Therefore, this systematic review aimed to evaluate the efficacy of oral/intravenous corticosteroids in the management of COVID-19. A comprehensive search was conducted across major scientific databases such as MEDLINE, Scopus, and Cochrane for relevant studies published from 2019-2024. The inclusion criteria included studies investigating the use of oral/intravenous corticosteroids in COVID-19 patients >18 years with a randomized placebo-controlled trial method. Non-placebo-controlled studies, studies using combined treatments with other drugs, as well as protocol articles, conference proceedings, review articles, and non-English studies were excluded. A narrative synthesis approach was adopted given the significant methodological diversity. The results showed that a total of 12 studies met the inclusion criteria covering the use of three drugs, including dexamethasone (three), hydrocortisone (two), and methylprednisolone (seven). The outcome parameters used for each study were different. Among the total 12 studies, five showed insignificant results for hydrocortisone (two) and methylprednisolone (three), while others reported significant results. This systematic review suggested that oral/intravenous corticosteroids might confer clinical benefits in the management of COVID-19, particularly in reducing mortality and severe disease outcomes. However, further investigation was needed to establish standardized protocols regarding dosage, duration, and safety considerations to optimize efficacy and minimize potential adverse effects.
Collapse
Affiliation(s)
- Irma Rahayu Latarissa
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
| | - Farida Rendrayani
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Ghina Nadhifah Iftinan
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
| | - Cecep Suhandi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Anna Meiliana
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Prodia Clinical Laboratory, Central Jakarta, Indonesia
| | - Ida Paulina Sormin
- Faculty of Pharmacy, University of 17 August 1945 Jakarta, Jakarta, Indonesia
- Prodia Diacro Laboratories, Jakarta, Indonesia
| | - Melisa Intan Barliana
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
| | - Keri Lestari
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
| |
Collapse
|
13
|
Wang B, Golubov J, Oswald EM, Poon P, Wei Q, Lett C, Shehadeh F, Kaczynski M, Felix LO, Mishra B, Mylona EK, Wipperman MF, Chio E, Hamon SC, Hooper AT, Somersan-Karakaya S, Musser BJ, Petro CD, Hamilton JD, Sleeman MA, Kalliolias GD, Mylonakis E, Skokos D. Potential immunomodulatory effects of CAS+IMD monoclonal antibody cocktail in hospitalized patients with COVID-19. EBioMedicine 2024; 108:105334. [PMID: 39270622 PMCID: PMC11415811 DOI: 10.1016/j.ebiom.2024.105334] [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: 01/19/2024] [Revised: 08/19/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Passive administration of SARS-CoV-2 neutralizing monoclonal antibodies (mAbs), such as CAS + IMD (Casirivimab + Imdevimab) antibody cocktail demonstrated beneficial effects on clinical outcomes in hospitalized patients with COVID-19 who were seronegative at baseline and outpatients. However, little is known about their impact on the host immunophenotypes. METHODS We conducted an immunoprofiling study in 46 patients from a single site of a multi-site trial of CAS + IMD in hospitalized patients. We collected longitudinal samples during October 2020 ∼ April 2021, prior to the emergence of the Delta and Omicron variants and the use of COVID-19 vaccines. All collected samples were analyzed without exclusion and post-hoc statistical analysis was performed. We examined the dynamic interplay of CAS + IMD with host immunity applying dimensional reduction approach on plasma proteomics and high dimensional flow cytometry data. FINDINGS Using an unbiased clustering method, we identified unique immunophenotypes associated with acute inflammation and disease resolution. Compared to placebo group, administration of CAS + IMD accelerated the transition from an acute inflammatory immunophenotype, to a less inflammatory or "resolving" immunophenotype, as characterized by reduced tissue injury, proinflammatory markers and restored lymphocyte/monocyte imbalance independent of baseline serostatus. Moreover, CAS + IMD did not impair the magnitude or the quality of host T cell immunity against SARS-CoV-2 spike protein. INTERPRETATION Our results identified immunophenotypic changes indicative of a possible SARS-CoV-2 neutralizing antibodies-induced anti-inflammatory effect, without an evident impairment of cellular antiviral immunity, suggesting that further studies of Mabs effects on SAS-CoV-2 or other viral mediated inflammation are warranted. FUNDING Regeneron Pharmaceuticals Inc and federal funds from the Department of Health and Human Services; Administration for Strategic Preparedness and Response; Biomedical Advanced Research and Development Authority, under OT number: HHSO100201700020C.
Collapse
Affiliation(s)
- Bei Wang
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | | | - Erin M Oswald
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | - Patrick Poon
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | - Qiaozhi Wei
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | - Clarissa Lett
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | - Fadi Shehadeh
- Division of Infectious Diseases, Department of Medicine, The Brown Alpert Medical School and the Miriam Hospital, Providence, RI, USA; Department of Medicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Matthew Kaczynski
- Division of Infectious Diseases, Department of Medicine, The Brown Alpert Medical School and the Miriam Hospital, Providence, RI, USA
| | - Lewis Oscar Felix
- Division of Infectious Diseases, Department of Medicine, The Brown Alpert Medical School and the Miriam Hospital, Providence, RI, USA; Department of Medicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Biswajit Mishra
- Division of Infectious Diseases, Department of Medicine, The Brown Alpert Medical School and the Miriam Hospital, Providence, RI, USA; Department of Medicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Evangelia K Mylona
- Division of Infectious Diseases, Department of Medicine, The Brown Alpert Medical School and the Miriam Hospital, Providence, RI, USA; Department of Medicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | | | - Erica Chio
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | - Sara C Hamon
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | - Andrea T Hooper
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | | | - Bret J Musser
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | | | | | - Matthew A Sleeman
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA
| | | | - Eleftherios Mylonakis
- Division of Infectious Diseases, Department of Medicine, The Brown Alpert Medical School and the Miriam Hospital, Providence, RI, USA; Department of Medicine, Houston Methodist Academic Institute, Houston, TX, 77030, USA
| | - Dimitris Skokos
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York, NY, 10591, USA.
| |
Collapse
|
14
|
Soni S, Antonescu L, Ro K, Horowitz JC, Mebratu YA, Nho RS. Influenza, SARS-CoV-2, and Their Impact on Chronic Lung Diseases and Fibrosis: Exploring Therapeutic Options. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1807-1822. [PMID: 39032604 PMCID: PMC11423761 DOI: 10.1016/j.ajpath.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/23/2024]
Abstract
Respiratory tract infections represent a significant global public health concern, disproportionately affecting vulnerable populations such as children, the elderly, and immunocompromised individuals. RNA viruses, particularly influenza viruses and coronaviruses, significantly contribute to respiratory illnesses, especially in immunosuppressed and elderly individuals. Influenza A viruses (IAVs) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continue to pose global health threats due to their capacity to cause annual epidemics, with profound implications for public health. In addition, the increase in global life expectancy is influencing the dynamics and outcomes of respiratory viral infections. Understanding the molecular mechanisms by which IAVs and SARS-CoV-2 contribute to lung disease progression is therefore crucial. The aim of this review is to comprehensively explore the impact of IAVs and SARS-CoV-2 on chronic lung diseases, with a specific focus on pulmonary fibrosis in the elderly. It also outlines potential preventive and therapeutic strategies and suggests directions for future research.
Collapse
Affiliation(s)
- Sourabh Soni
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine and The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Laura Antonescu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine and The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Kaylin Ro
- Scripps Research Institute, San Diego, California
| | - Jeffrey C Horowitz
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine and The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - Yohannes A Mebratu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine and The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio.
| | - Richard S Nho
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine and The Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
15
|
Michot JM, Dozio V, Rohmer J, Pommeret F, Roumier M, Yu H, Sklodowki K, Danlos FX, Ouali K, Kishazi E, Naigeon M, Griscelli F, Gachot B, Groh M, Bacciarello G, Stoclin A, Willekens C, Sakkal M, Bayle A, Zitvogel L, Silvin A, Soria JC, Barlesi F, Beeler K, André F, Vasse M, Chaput N, Ackermann F, Escher C, Marabelle A. Circulating Proteins Associated with Anti-IL6 Receptor Therapeutic Resistance in the Sera of Patients with Severe COVID-19. J Proteome Res 2024. [PMID: 39352225 DOI: 10.1021/acs.jproteome.2c00422] [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: 10/03/2024]
Abstract
Circulating proteomes provide a snapshot of the physiological state of a human organism responding to pathogenic challenges and drug interventions. The outcomes of patients with COVID-19 and acute respiratory distress syndrome triggered by the SARS-CoV2 virus remain uncertain. Tocilizumab is an anti-interleukin-6 treatment that exerts encouraging clinical activity by controlling the cytokine storm and improving respiratory distress in patients with COVID-19. We investigate the biological determinants of therapeutic outcomes after tocilizumab treatment. Overall, 28 patients hospitalized due to severe COVID-19 who were treated with tocilizumab intravenously were included in this study. Sera were collected before and after tocilizumab, and the patient's outcome was evaluated until day 30 post-tocilizumab infusion for favorable therapeutic response to tocilizumab and mortality. Hyperreaction monitoring measurements by liquid chromatography-mass spectrometry-based proteomic analysis with data-independent acquisition quantified 510 proteins and 7019 peptides in the serum of patients. Alterations in the serum proteome reflect COVID-19 outcomes in patients treated with tocilizumab. Our results suggested that circulating proteins associated with the most significant prognostic impact belonged to the complement system, platelet degranulation, acute-phase proteins, and the Fc-epsilon receptor signaling pathway. Among these, upregulation of the complement system by activation of the classical pathway was associated with poor response to tocilizumab, and upregulation of Fc-epsilon receptor signaling was associated with lower mortality.
Collapse
Affiliation(s)
- Jean-Marie Michot
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Vito Dozio
- Biognosys, Wagistrasse 21, Schlieren 8952, Switzerland
| | - Julien Rohmer
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | - Fanny Pommeret
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Mathilde Roumier
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | - Haochen Yu
- Biognosys, Wagistrasse 21, Schlieren 8952, Switzerland
| | | | - François-Xavier Danlos
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Kaissa Ouali
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Edina Kishazi
- Biognosys, Wagistrasse 21, Schlieren 8952, Switzerland
| | - Marie Naigeon
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Laboratoire d'Immunomonitoring en Oncologie, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- Université Paris Saclay, Faculté de Pharmacie, Chatenay-Malabry F-92296, France
| | - Franck Griscelli
- Département de biologie et pathologie, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Bertrand Gachot
- Unité de Pathologie Infectieuse, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Matthieu Groh
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | - Giulia Bacciarello
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Annabelle Stoclin
- Unité de Pathologie Infectieuse, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Christophe Willekens
- Département d'hématologie, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Madona Sakkal
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Arnaud Bayle
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | | | - Aymeric Silvin
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Jean-Charles Soria
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
| | - Fabrice Barlesi
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | | | - Fabrice André
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
- Unité INSERM U981, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Marc Vasse
- Université Paris Saclay, Faculté de Pharmacie, Chatenay-Malabry F-92296, France
- Service de biologie clinique, Hôpital Foch, Suresnes 92150, France
- Unité INSERM U1176, Le Kremlin-Bicêtre, Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
| | - Nathalie Chaput
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Laboratoire d'Immunomonitoring en Oncologie, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Felix Ackermann
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | | | - Aurélien Marabelle
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
- Centre d'investigation clinique - biothérapie, INSERM CICBT1428, Villejuif 94800, France
| |
Collapse
|
16
|
McCarthy MW. The first five years of SARS-CoV-2: inpatient treatment updates and future directions. Expert Opin Pharmacother 2024; 25:1873-1878. [PMID: 39305134 DOI: 10.1080/14656566.2024.2408375] [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: 08/14/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION In December 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first identified in adults with pneumonia in Wuhan, China. AREAS COVERED It is now believed that several billion humans have been infected with SARS-CoV-2 and more than ten million have died from coronavirus disease 2019 (COVID-19), the disease caused by SARS-CoV-2. EXPERT OPINION The first five years of the SARS-CoV-2 pandemic have been marked by unfathomable suffering as well as remarkable scientific progress. This manuscript examines what has been learned about the treatment of inpatients with COVID-19 and explores how the therapeutic approach may evolve in the years ahead.
Collapse
|
17
|
Jhanwar A, Sharma D, Das U. Unraveling the structural and functional dimensions of SARS-CoV2 proteins in the context of COVID-19 pathogenesis and therapeutics. Int J Biol Macromol 2024; 278:134850. [PMID: 39168210 DOI: 10.1016/j.ijbiomac.2024.134850] [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: 01/12/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) has emerged as the causative agent behind the global pandemic of Coronavirus Disease 2019 (COVID-19). As the scientific community strives to comprehend the intricate workings of this virus, a fundamental aspect lies in deciphering the myriad proteins it expresses. This knowledge is pivotal in unraveling the complexities of the viral machinery and devising targeted therapeutic interventions. The proteomic landscape of SARS-CoV2 encompasses structural, non-structural, and open-reading frame proteins, each playing crucial roles in viral replication, host interactions, and the pathogenesis of COVID-19. This comprehensive review aims to provide an updated and detailed examination of the structural and functional attributes of SARS-CoV2 proteins. By exploring the intricate molecular architecture, we have highlighted the significance of these proteins in viral biology. Insights into their roles and interplay contribute to a deeper understanding of the virus's mechanisms, thereby paving the way for the development of effective therapeutic strategies. As the global scientific community strives to combat the ongoing pandemic, this synthesis of knowledge on SARS-CoV2 proteins serves as a valuable resource, fostering informed approaches toward mitigating the impact of COVID-19 and advancing the frontier of antiviral research.
Collapse
Affiliation(s)
- Aniruddh Jhanwar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Dipika Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Uddipan Das
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
18
|
De Clercq E, Li G, Zhang Y, Huang J, Tan L. Unachieved antiviral strategies with acyclic nucleoside phosphonates: Dedicated to the memory of dr. Salvatore "Sam" Joseph Enna. Biochem Pharmacol 2024; 228:116448. [PMID: 39043335 DOI: 10.1016/j.bcp.2024.116448] [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: 05/04/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 07/25/2024]
Abstract
Many acyclic nucleoside phosphonates such as cidofovir, adefovir dipivoxil, tenofovir disoproxil fumarate, and tenofovir alafenamide have been marketed for the treatment or prophylaxis of infectious diseases. Here, this review highlights potent acyclic nucleoside phosphonates for their potential in the treatment of retrovirus (e.g., human immunodeficiency virus) and DNA virus (e.g., adeno-, papilloma-, herpes- and poxvirus) infections. If properly assessed and/or optimized, some potent acyclic nucleoside phosphonates can be possibly applied in the control of current and emerging infectious diseases.
Collapse
Affiliation(s)
- Erik De Clercq
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Leuven 3000, Belgium
| | - Guangdi Li
- Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Yun Zhang
- Huaihua City Maternal and Child Health Care Hospital, Huaihua 418000, China
| | - Jie Huang
- Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Li Tan
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Hunan Clinical Molecular Diagnosis Center, Molecular Diagnostic Technology Hunan Engineering Research Center, Clinical Medical Research Center for Molecular Diagnosis of Infectious Diseases in Hunan Province, Changsha 410011, China.
| |
Collapse
|
19
|
Gege C, Hahn F, Wangen C, Häge S, Herrmann A, Uhlig N, Eberlein V, Issmail L, Klopfleisch R, Grunwald T, Marschall M, Kohlhof H, Vitt D. Synthesis and Characterization of DHODH Inhibitors Based on the Vidofludimus Scaffold with Pronounced Anti-SARS-CoV-2 Activity. ChemMedChem 2024; 19:e202400292. [PMID: 38887198 DOI: 10.1002/cmdc.202400292] [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: 04/22/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/20/2024]
Abstract
New strategies for the rapid development of broad-spectrum antiviral therapies are urgently required for emerging and re-emerging viruses like the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Host-directed antivirals that target universal cellular metabolic pathways necessary for viral replication present a promising approach with broad-spectrum activity and low potential for development of viral resistance. Dihydroorotate dehydrogenase (DHODH) was identified as one of those universal host factors essential for the replication of many clinically relevant human pathogenic viruses. DHODH is the rate-limiting enzyme catalyzing the fourth step in the de novo pyrimidine synthesis. Therefore, it is also developed as a therapeutic target for many diseases relying on cellular pyrimidine resources, such as cancer, autoimmune diseases and viral or bacterial infection. Thus, several DHODH inhibitors, including vidofludimus calcium (VidoCa, IMU-838), are currently in development or have been investigated in clinical trials for the treatment of virus infections such as SARS-CoV-2-mediated coronavirus disease 19 (COVID-19). Here, we report the medicinal chemistry optimization of VidoCa that resulted in metabolically more stable derivatives with improved DHODH target inhibition in various mammalian species, which translated into improved efficacy against SARS-CoV-2.
Collapse
Affiliation(s)
- Christian Gege
- Immunic AG, Lochhamer Schlag 21, 82166, Gräfelfing, Germany
| | - Friedrich Hahn
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054, Erlangen, Germany
| | - Christina Wangen
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054, Erlangen, Germany
| | - Sigrun Häge
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054, Erlangen, Germany
| | | | - Nadja Uhlig
- Department of Vaccines and Infection Models, Unit Preclinical Validation, Fraunhofer-Institute for Cell Therapy and Immunology IZI, Perlickstrasse 1, 04103, Leipzig, Germany
| | - Valentina Eberlein
- Department of Vaccines and Infection Models, Unit Preclinical Validation, Fraunhofer-Institute for Cell Therapy and Immunology IZI, Perlickstrasse 1, 04103, Leipzig, Germany
| | - Leila Issmail
- Department of Vaccines and Infection Models, Unit Preclinical Validation, Fraunhofer-Institute for Cell Therapy and Immunology IZI, Perlickstrasse 1, 04103, Leipzig, Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Str. 15, 14163, Berlin, Germany
| | - Thomas Grunwald
- Department of Vaccines and Infection Models, Unit Preclinical Validation, Fraunhofer-Institute for Cell Therapy and Immunology IZI, Perlickstrasse 1, 04103, Leipzig, Germany
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054, Erlangen, Germany
| | - Hella Kohlhof
- Immunic AG, Lochhamer Schlag 21, 82166, Gräfelfing, Germany
| | - Daniel Vitt
- Immunic AG, Lochhamer Schlag 21, 82166, Gräfelfing, Germany
| |
Collapse
|
20
|
Gunasekaran V, Sathishkumar P. Scutellaria baicalensis baicalein as a novel therapeutic agent to treat SARS-CoV-2 infections. Nat Prod Res 2024:1-2. [PMID: 39353035 DOI: 10.1080/14786419.2024.2407512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024]
Affiliation(s)
- Vinothini Gunasekaran
- Green Lab, Department of Prosthodontics, Saveetha Dental College and Hospital, SIMATS, Saveetha University, Chennai, India
| | - Palanivel Sathishkumar
- Green Lab, Department of Prosthodontics, Saveetha Dental College and Hospital, SIMATS, Saveetha University, Chennai, India
| |
Collapse
|
21
|
Balck A, Lange LM, Neumann A, Royl G, Jung P, Schaumberg J, Brüggemann N, Koch PJ. Highly beneficial outcome in severe acute necrotizing encephalopathy with tocilizumab treatment. J Neurol 2024; 271:7042-7045. [PMID: 39242371 PMCID: PMC11447148 DOI: 10.1007/s00415-024-12661-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024]
Affiliation(s)
- Alexander Balck
- Department of Neurology, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Lara M Lange
- Department of Neurology, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Alexander Neumann
- Department of Neuroradiology, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Georg Royl
- Department of Neurology, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Philipp Jung
- Department of Paediatrics, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Jens Schaumberg
- Department of Neurology, SANA Clinic Lübeck, Lübeck, Germany
| | - Norbert Brüggemann
- Department of Neurology, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany.
| | - Philipp J Koch
- Department of Neurology, University of Lübeck and University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| |
Collapse
|
22
|
Green AL, De Bellis D, Cowell E, Lenchine RV, Penn T, Kris LP, McEvoy-May J, Bihari S, Dixon DL, Carr JM. The Y498T499-SARS-CoV-2 spike (S) protein interacts poorly with rat ACE2 and does not affect the rat lung. Access Microbiol 2024; 6:000839.v3. [PMID: 39346684 PMCID: PMC11432600 DOI: 10.1099/acmi.0.000839.v3] [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: 05/07/2024] [Accepted: 09/05/2024] [Indexed: 10/01/2024] Open
Abstract
The rat is a useful laboratory model for respiratory diseases. SARS-CoV-2 proteins, such as the spike (S) protein, can induce inflammation. This study has investigated the ability of the Q498Y, P499T (QP-YT) amino acid change, described in the S-protein of the mouse-adapted laboratory SARS-CoV-2 MA strain, to interact with rat angiotensin converting enzyme-2 (ACE2) and stimulate responses in rat lungs. A real-time S-ACE2 quantitative fusion assay shows that ancestral and L452R S-proteins fuse with human but not rat ACE2 expressed on HEK293 (human embryonic kidney-293) cells. The QP-YT S-protein retains the ability to fuse with human ACE2 and increases the binding to rat ACE2. Although lower lung of the rat contains both ACE2 and TMPRSS2 (transmembrane serine protease 2) target cells, intratracheal delivery of ancestral or QP-YT S-protein pseudotyped lentivirus did not induce measurable respiratory changes, inflammatory infiltration or innate mRNA responses. Isolation of primary cells from rat alveoli demonstrated the presence of cells expressing ACE2 and TMPRSS2. Infection of these cells, however, with ancestral or QP-YT S-protein pseudotyped lentivirus was not observed, and the QP-YT S-protein pseudotyped lentivirus poorly infected HEK293 cells expressing rat ACE2. Analysis of the amino acid changes across the S-ACE2 interface highlights not only the Y498 interaction with H353 as a likely facilitator of binding to rat ACE2 but also other amino acids that could improve this interaction. Thus, rat lungs contain cells expressing receptors for SARS-CoV-2, and the QP-YT S-protein variant can bind to rat ACE2, but this does not result in infection or stimulate responses in the lung. Further, amino acid changes in S-protein may enhance this interaction to improve the utility of the rat model for defining the role of the S-protein in driving lung inflammation.
Collapse
Affiliation(s)
- Amy L Green
- College of Medicine and Public Health, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Dylan De Bellis
- College of Medicine and Public Health, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
| | - Evangeline Cowell
- College of Medicine and Public Health, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Roman V Lenchine
- College of Medicine and Public Health, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Timothy Penn
- College of Medicine and Public Health, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Luke P Kris
- College of Medicine and Public Health, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - James McEvoy-May
- College of Medicine and Public Health, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
| | - Shailesh Bihari
- College of Medicine and Public Health, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
| | - Dani-Louise Dixon
- College of Medicine and Public Health, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
| | - Jillian M Carr
- College of Medicine and Public Health, Flinders University, GPO Box 2100, Adelaide, South Australia 5001, Australia
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
23
|
van der Horst D, Carter-Timofte ME, Danneels A, Silva da Costa L, Kurmasheva N, Thielke AL, Hansen AL, Chorošajev V, Holm CK, Belouzard S, de Weber I, Beny C, Olagnier D. Large-scale deep learning identifies the antiviral potential of PKI-179 and MTI-31 against coronaviruses. Antiviral Res 2024; 231:106012. [PMID: 39332537 DOI: 10.1016/j.antiviral.2024.106012] [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: 03/01/2024] [Revised: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has led to the global pandemic of Coronavirus Disease (2019) (COVID-19), underscoring the urgency for effective antiviral drugs. Despite the development of different vaccination strategies, the search for specific antiviral compounds remains crucial. Here, we combine machine learning (ML) techniques with in vitro validation to efficiently identify potential antiviral compounds. We overcome the limited amount of SARS-CoV-2 data available for ML using various techniques, supplemented with data from diverse biomedical assays, which enables end-to-end training of a deep neural network architecture. We use its predictions to identify and prioritize compounds for in vitro testing. Two top-hit compounds, PKI-179 and MTI-31, originally identified as Pi3K-mTORC1/2 pathway inhibitors, exhibit significant antiviral activity against SARS-CoV-2 at low micromolar doses. Notably, both compounds outperform the well-known mTOR inhibitor rapamycin. Furthermore, PKI-179 and MTI-31 demonstrate broad-spectrum antiviral activity against SARS-CoV-2 variants of concern and other coronaviruses. In a physiologically relevant model, both compounds show antiviral effects in primary human airway epithelial (HAE) cultures derived from healthy donors cultured in an air-liquid interface (ALI). This study highlights the potential of ML combined with in vitro testing to expedite drug discovery, emphasizing the adaptability of AI-driven approaches across different viruses, thereby contributing to pandemic preparedness.
Collapse
Affiliation(s)
| | | | - Adeline Danneels
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL- Center for Infection and Immunity of Lille, Lille, 59000, France
| | | | - Naziia Kurmasheva
- Aarhus University, Department of Biomedicine, Aarhus C, 8000, Denmark
| | - Anne L Thielke
- Aarhus University, Department of Biomedicine, Aarhus C, 8000, Denmark
| | | | | | - Christian K Holm
- Aarhus University, Department of Biomedicine, Aarhus C, 8000, Denmark
| | - Sandrine Belouzard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL- Center for Infection and Immunity of Lille, Lille, 59000, France
| | | | | | - David Olagnier
- Aarhus University, Department of Biomedicine, Aarhus C, 8000, Denmark.
| |
Collapse
|
24
|
Kiso M, Uraki R, Yamayoshi S, Imai M, Kawaoka Y. Drug susceptibility and the potential for drug-resistant SARS-CoV-2 emergence in immunocompromised animals. iScience 2024; 27:110729. [PMID: 39280602 PMCID: PMC11402253 DOI: 10.1016/j.isci.2024.110729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/24/2024] [Accepted: 08/09/2024] [Indexed: 09/18/2024] Open
Abstract
The reduced susceptibility of mRNA vaccines and diminished neutralizing activity of therapeutic monoclonal antibodies against Omicron variants, including BQ.1.1, XBB, and their descendants, highlight the importance of antiviral therapies. Here, we assessed the efficacy of two antivirals, molnupiravir, targeting a viral RNA-dependent RNA polymerase, and nirmatrelvir, targeting a main protease, against BQ.1.1 in hamsters. We found that prophylactic or therapeutic treatment with either drug significantly reduced the viral load in the lungs of infected hamsters. We also evaluated the risk of emergence of drug-resistant viruses in immunocompromised hamsters. Although 13 days of drug treatment reduced viral titers, the immunocompromised hosts could not completely clear the virus. Viruses isolated from drug-treated immunocompromised hamsters did not show reduced susceptibility to the drugs. Molnupiravir and nirmatrelvir remain effective in vivo against variants with reduced susceptibility to monoclonal antibodies and mRNA vaccine-induced antibodies, with limited emergence of drug-resistant variants under the conditions tested.
Collapse
Affiliation(s)
- Maki Kiso
- Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), The University of Tokyo, Tokyo 108-8639, Japan
| | - Ryuta Uraki
- Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), The University of Tokyo, Tokyo 108-8639, Japan
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo 162-8655, Japan
| | - Seiya Yamayoshi
- Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), The University of Tokyo, Tokyo 108-8639, Japan
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo 162-8655, Japan
- International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Masaki Imai
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo 162-8655, Japan
- International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Yoshihiro Kawaoka
- Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), The University of Tokyo, Tokyo 108-8639, Japan
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo 162-8655, Japan
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53711, USA
| |
Collapse
|
25
|
Al Saihati HA, Dessouky AA, Salim RF, Elgohary I, El-Sherbiny M, Ali FEM, Moustafa MMA, Shaheen D, Forsyth NR, Badr OA, Ebrahim N. MSC-extracellular vesicle microRNAs target host cell-entry receptors in COVID-19: in silico modeling for in vivo validation. Stem Cell Res Ther 2024; 15:316. [PMID: 39304926 PMCID: PMC11416018 DOI: 10.1186/s13287-024-03889-9] [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: 03/25/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has created a global pandemic with significant morbidity and mortality. SARS-CoV-2 primarily infects the lungs and is associated with various organ complications. Therapeutic approaches to combat COVID-19, including convalescent plasma and vaccination, have been developed. However, the high mutation rate of SARS-CoV-2 and its ability to inhibit host T-cell activity pose challenges for effective treatment. Mesenchymal stem cells (MSCs) and their extracellular vesicles (MSCs-EVs) have shown promise in COVID-19 therapy because of their immunomodulatory and regenerative properties. MicroRNAs (miRNAs) play crucial regulatory roles in various biological processes and can be manipulated for therapeutic purposes. OBJECTIVE We aimed to investigate the role of lyophilized MSC-EVs and their microRNAs in targeting the receptors involved in SARS-CoV-2 entry into host cells as a strategy to limit infection. In silico microRNA prediction, structural predictions of the microRNA-mRNA duplex, and molecular docking with the Argonaut protein were performed. METHODS Male Syrian hamsters infected with SARS-CoV-2 were treated with human Wharton's jelly-derived Mesenchymal Stem cell-derived lyophilized exosomes (Bioluga Company)via intraperitoneal injection, and viral shedding was assessed. The potential therapeutic effects of MSCs-EVs were measured via histopathology of lung tissues and PCR for microRNAs. RESULTS The results revealed strong binding potential between miRNA‒mRNA duplexes and the AGO protein via molecular docking. MSCs-EVs reduced inflammation markers and normalized blood indices via the suppression of viral entry by regulating ACE2 and TMPRSS2 expression. MSCs-EVs alleviated histopathological aberrations. They improved lung histology and reduced collagen fiber deposition in infected lungs. CONCLUSION We demonstrated that MSCs-EVs are a potential therapeutic option for treating COVID-19 by preventing viral entry into host cells.
Collapse
Affiliation(s)
- Hajer A Al Saihati
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Albatin, Hafar Al-Batin, Saudi Arabia.
| | - Arigue A Dessouky
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Rabab F Salim
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Islam Elgohary
- Researcher of Pathology, Animal Health Research Institute, Agriculture Research Center, Giza, Egypt
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, 11597, Riyadh, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, Egypt
| | - Mahmoud M A Moustafa
- Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Benha, Egypt
| | - Dalia Shaheen
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nicholas Robert Forsyth
- PhD Molecular Genetics, Vice Principals' Office, Kings College, University of Aberdeen, Aberdeen, AB24 3FX, UK
- Cell and Tissue Engineering, School of pharmacy and bioengineering, Keele University, Keele, UK
| | - Omnia A Badr
- Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Benha, Egypt.
| | - Nesrine Ebrahim
- Department of Medical Histology and Cell Biology Faculty of Medicine, Benha University, Benha, Egypt.
- Stem Cell Unit, Faculty of Medicine, Benha University, Benha, Egypt.
- Faculty of Medicine, Benha National University, Al Obour City, Egypt.
- Cell and Tissue Engineering, School of pharmacy and bioengineering, Keele University, Keele, UK.
| |
Collapse
|
26
|
Chhetri SP, Bhandari VS, Maharjan R, Lamichhane TR. Identification of lead inhibitors for 3CLpro of SARS-CoV-2 target using machine learning based virtual screening, ADMET analysis, molecular docking and molecular dynamics simulations. RSC Adv 2024; 14:29683-29692. [PMID: 39297030 PMCID: PMC11408992 DOI: 10.1039/d4ra04502e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/04/2024] [Indexed: 09/21/2024] Open
Abstract
The SARS-CoV-2 3CLpro is a critical target for COVID-19 therapeutics due to its role in viral replication. We employed a screening pipeline to identify novel inhibitors by combining machine learning classification with similarity checks of approved medications. A voting classifier, integrating three machine learning classifiers, was used to filter a large database (∼10 million compounds) for potential inhibitors. This ensemble-based machine learning technique enhances overall performance and robustness compared to individual classifiers. From the screening, three compounds M1, M2 and M3 were selected for further analysis. Absorption, distribution, metabolism, excretion, and toxicity (ADMET) analysis compared these candidates to nirmatrelvir and azvudine. Molecular docking followed by 200 ns MD simulations showed that only M1 (6-[2,4-bis(dimethylamino)-6,8-dihydro-5H-pyrido[3,4-d]pyrimidine-7-carbonyl]-1H-pyrimidine-2,4-dione) remained stable. For azvudine and M1, the estimated median lethal doses are 1000 and 550 mg kg-1, respectively, with maximum tolerated doses of 0.289 and 0.614 log mg per kg per day. The predicted inhibitory activity of M1 is 7.35, similar to that of nirmatrelvir. The binding free energy based on Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) of M1 is -18.86 ± 4.38 kcal mol-1, indicating strong binding interactions. These findings suggest that M1 merits further investigation as a potential SARS-CoV-2 treatment.
Collapse
Affiliation(s)
| | | | - Rajesh Maharjan
- Central Department of Physics, Tribhuvan University Kathmandu 44600 Nepal
| | | |
Collapse
|
27
|
Rahmani D, Jafari A, Kesharwani P, Sahebkar A. Molecular targets in SARS-CoV-2 infection: An update on repurposed drug candidates. Pathol Res Pract 2024; 263:155589. [PMID: 39276508 DOI: 10.1016/j.prp.2024.155589] [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: 05/20/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024]
Abstract
The 2019 widespread contagion of the human coronavirus novel type (SARS-CoV-2) led to a pandemic declaration by the World Health Organization. A daily increase in patient numbers has formed an urgent necessity to find suitable targets and treatment options for the novel coronavirus (COVID-19). Despite scientists' struggles to discover quick treatment solutions, few effective specific drugs are approved to control SARS-CoV-2 infections thoroughly. Drug repositioning or Drug repurposing and target-based approaches are promising strategies for facilitating the drug discovery process. Here, we review current in silico, in vitro, in vivo, and clinical updates regarding proposed drugs for prospective treatment options for COVID-19. Drug targets that can direct pharmaceutical sciences efforts to discover new drugs against SARS-CoV-2 are divided into two categories: Virus-based targets, for example, Spike glycoprotein and Nucleocapsid Protein, and host-based targets, for instance, inflammatory cytokines and cell receptors through which the virus infects the cell. A broad spectrum of drugs has been found to show anti-SARS-CoV-2 potential, including antiviral drugs and monoclonal antibodies, statins, anti-inflammatory agents, and herbal products.
Collapse
Affiliation(s)
- Dibachehr Rahmani
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ameneh Jafari
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
28
|
Tripp RA, Martin DE. Antiviral agents and therapeutics against respiratory viruses. Expert Opin Investig Drugs 2024:1-5. [PMID: 39245955 DOI: 10.1080/13543784.2024.2401911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION Respiratory viruses are responsible for significant worldwide morbidity and mortality. While vaccines are highly effective at reducing the morbidity and mortality associated with viral infections, this protection is incomplete. It requires a high degree of compliance, which is hindered by vaccine hesitancy. To address these gaps, antiviral agents and therapeutics are crucial in combating diseases caused by respiratory viruses. Antiviral agents are broadly classified into two groups: 1) direct-acting antivirals (DAA) and 2) host-directed antivirals (HDA). AREAS COVERED This review comprehensively examines Phase II FDA-approved antiviral drugs for influenza virus, SARS-CoV-2, and RSV as published in clinicaltrials.gov. It focuses on DAAs and various monoclonal antibodies (mAbs) that have been approved for the prevention and treatment of viral respiratory tract infections. EXPERT OPINION Antiviral drugs being developed assess different mechanisms of action to combat viruses and other delivery routes (i.e. oral, inhalation, or parenteral). The associated clinical trials address the impact on disease while determining the appropriate dosage levels for further investigation in Phase III. A robust pipeline of agents is necessary to meet the global need for effective antiviral therapeutics.
Collapse
Affiliation(s)
- Ralph A Tripp
- College of Veterinary Medicine, Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| | | |
Collapse
|
29
|
Wang WW, Zeng P, Liu T, Zhou XL, Lin C, Guo L, Wang QS, Li J. Structural Basis of Main Proteases of Coronavirus Bound to Bofutrelvir. J Mol Biol 2024; 436:168784. [PMID: 39245318 DOI: 10.1016/j.jmb.2024.168784] [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: 08/09/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Globally, the continuous spread and evolution of SARS-CoV-2, along with its variants, profoundly impact human well-being, health, security, and the growth of socio-economic. In the field of development of drugs against COVID-19, the main protease (Mpro) is a critical target as it plays a core role in the lifecycle of SARS-CoV-2. Bofutrelvir acts as a potent inhibitor of SARS-CoV-2 Mpro, demonstrating high efficacy and broad-spectrum antiviral activity. Compared to therapies that require pharmacokinetic boosters, such as ritonavir, the monotherapy approach of Bofutrelvir reduces the risk of potential drug interactions, making it suitable for a wider patient population. However, further studies on the potency and mechanism of inhibition of Bofutrelvir against the Mpro of COVID-19 and its variants, together with other coronaviruses, are needed to prepare for the possibility of a possible re-emerging threat from an analogous virus in the future. Here, we reveal the effective inhibition of Bofutrelvir against the Mpro of SARS-CoV-2, SARS-CoV, and HCoV-229E through FRET and crystallographic analysis. Furthermore, the inhibitory mechanisms of Bofutrelvir against two SARS-CoV-2 Mpro mutants (G15S and K90R) were also elucidated through FRET and crystallographic studies. Through detailed analysis and comparison of these crystal structures, we identified crucial structural determinants of inhibition and elucidated the binding mode of Bofutrelvir to Mpros from different coronaviruses. These findings are hopeful to accelerate the development of safer and more potent inhibitors against the Mpro of coronavirus, and to provide important references for the prevention and treatment of similar viruses that may emerge in the future.
Collapse
Affiliation(s)
- Wei-Wei Wang
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Pei Zeng
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, School of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Tongchao Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xue-Lan Zhou
- Shenzhen Crystalo Biopharmaceutical Co, Ltd, Shenzhen 518118, China
| | - Cheng Lin
- Jiangxi Jmerry Biopharmaceutical Co, Ltd, Ganzhou 341000, China
| | - Li Guo
- Jiangxi Jmerry Biopharmaceutical Co, Ltd, Ganzhou 341000, China
| | - Qi-Sheng Wang
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China.
| | - Jian Li
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, School of Pharmacy, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
30
|
Deng J, Qin C, Lee M, Lee Y, You M, Liu J. Effects of rehabilitation interventions for old adults with long COVID: A systematic review and meta-analysis of randomised controlled trials. J Glob Health 2024; 14:05025. [PMID: 39238359 PMCID: PMC11377967 DOI: 10.7189/jogh.14.05025] [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: 09/07/2024] Open
Abstract
Background There is limited evidence on the effectiveness of the existing rehabilitation interventions for old adults with long coronavirus disease (COVID), which is of particular concern among old adults. Methods We systematically searched studies published in PubMed, EMBASE, Web of Science, Scopus, and Cochrane Library databases from their inception to 15 November 2023. Randomised controlled trials (RCTs) compared rehabilitation interventions with other controls in old adults (mean/median age of 60 or older) with long COVID were included. We performed a meta-analysis to compare the effects of the rehabilitation interventions with the common control group. Mean difference (MD) or standardised mean difference (SMD) with its 95% confidence intervals (CI) were used as summary statistics. Moreover, subgroup analyses based on the intervention programmes, the severity of acute infection, and the age of participants were carried out. Results A total of 11 RCTs involving 832 participants (64.37 ± 7.94 years, 52.2% were men) were included in the analysis. Compared with the control groups, rehabilitation interventions significantly improved 6-minute walking test (6 MWT; MD = 15.77 metres (m), 95% CI = 5.40, 26.13, P < 0.01), 30-second sit-to-stand test (MD = 4.11 number of stands (n), 95% CI = 2.46, 5.76, P < 0.001), all aspects of quality of life, independence in activities of daily living (SMD = 0.31, 95% CI = 0.14, 0.48, P < 0.001), and relieved fatigue (SMD = -0.66, 95% CI = -1.13, -0.19, P < 0.01), depression (SMD = -0.89, 95% CI = -1.76, -0.02, P < 0.05) and anxiety (SMD = -0.81, 95% CI = -1.58, -0.05, P < 0.05). However, the improvement of hand grip strength and pulmonary function was not statistically significant (P > 0.05). Subgroup analyses showed that improvements in 6 MWT, fatigue, anxiety, and depression were more pronounced in old patients who received exercise training, while those who received respiratory rehabilitation had more pronounced improvements in pulmonary function and quality of life. Conclusions Old adults with long COVID who underwent rehabilitation interventions experienced significant improvement in functional capacity, fatigue, quality of life, independence in activities of daily living, and mental health outcomes compared with usual/standard care. These findings suggest that screening, management, and rehabilitation interventions for long COVID in older adults should be strengthened to improve their complete health status and functional status, thereby reducing the long-term disease burden caused by long COVID and fostering healthy aging during the post-pandemic era.
Collapse
Affiliation(s)
- Jie Deng
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Chenyuan Qin
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Minjung Lee
- Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Yubin Lee
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Myoungsoon You
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - Jue Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute for Global Health and Development, Peking University, Beijing, China
| |
Collapse
|
31
|
Porebski B, Christ W, Corman A, Haraldsson M, Barz M, Lidemalm L, Häggblad M, Ilmain J, Wright SC, Murga M, Schlegel J, Jarvius M, Lapins M, Sezgin E, Bhabha G, Lauschke VM, Carreras-Puigvert J, Lafarga M, Klingström J, Hühn D, Fernandez-Capetillo O. Discovery of a novel inhibitor of macropinocytosis with antiviral activity. Mol Ther 2024; 32:3012-3024. [PMID: 38956870 PMCID: PMC11403221 DOI: 10.1016/j.ymthe.2024.06.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/04/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024] Open
Abstract
Several viruses hijack various forms of endocytosis in order to infect host cells. Here, we report the discovery of a molecule with antiviral properties that we named virapinib, which limits viral entry by macropinocytosis. The identification of virapinib derives from a chemical screen using high-throughput microscopy, where we identified chemical entities capable of preventing infection with a pseudotype virus expressing the spike (S) protein from SARS-CoV-2. Subsequent experiments confirmed the capacity of virapinib to inhibit infection by SARS-CoV-2, as well as by additional viruses, such as mpox virus and TBEV. Mechanistic analyses revealed that the compound inhibited macropinocytosis, limiting this entry route for the viruses. Importantly, virapinib has no significant toxicity to host cells. In summary, we present the discovery of a molecule that inhibits macropinocytosis, thereby limiting the infectivity of viruses that use this entry route such as SARS-CoV2.
Collapse
Affiliation(s)
- Bartlomiej Porebski
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 21 Stockholm, Sweden
| | - Wanda Christ
- Center of Infectious Medicine, Department of Medicine, Karolinska Institutet, 141-86 Huddinge, Sweden
| | - Alba Corman
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 21 Stockholm, Sweden
| | - Martin Haraldsson
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Myriam Barz
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 21 Stockholm, Sweden
| | - Louise Lidemalm
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 21 Stockholm, Sweden
| | - Maria Häggblad
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 21 Stockholm, Sweden
| | - Juliana Ilmain
- Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Shane C Wright
- Department of Physiology and Pharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Matilde Murga
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Jan Schlegel
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Malin Jarvius
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Box 591, SE-751 24 Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-751 24 Uppsala, Sweden
| | - Maris Lapins
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Box 591, SE-751 24 Uppsala, Sweden
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Gira Bhabha
- Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, S-171 77 Stockholm, Sweden; Margarete Fischer-Bosch Institute of Clinical Pharmacology, D-70376 Stuttgart, Germany; University of Tuebingen, 72074 Tuebingen, Germany
| | - Jordi Carreras-Puigvert
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Box 591, SE-751 24 Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-751 24 Uppsala, Sweden
| | - Miguel Lafarga
- Departament of Anatomy and Cellular Biology, Neurodegenerative Diseases Network (CIBERNED), University of Cantabria-IDIVAL, 39011 Santander, Spain
| | - Jonas Klingström
- Center of Infectious Medicine, Department of Medicine, Karolinska Institutet, 141-86 Huddinge, Sweden; Department of Biomedical and Clinical Sciences, Linköping University, 581 83 Linköping, Sweden
| | - Daniela Hühn
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 21 Stockholm, Sweden
| | - Oscar Fernandez-Capetillo
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 21 Stockholm, Sweden; Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain.
| |
Collapse
|
32
|
Do TND, Abdelnabi R, Boda B, Constant S, Neyts J, Jochmans D. The triple combination of Remdesivir (GS-441524), Molnupiravir and Ribavirin is highly efficient in inhibiting coronavirus replication in human nasal airway epithelial cell cultures and in a hamster infection model. Antiviral Res 2024; 231:105994. [PMID: 39237005 DOI: 10.1016/j.antiviral.2024.105994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/20/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024]
Abstract
The use of fixed dose-combinations of antivirals with different mechanisms of action has proven key in the successful treatment of infections with HIV and HCV. For the treatment of infections with SARS-CoV-2 and possible future epi-/pandemic coronaviruses, it will be important to explore the efficacy of combinations of different drugs, in particular to avoid resistance development, such as in patients with immunodeficiencies. This work explores the effect of a combination of 3 broad-spectrum antiviral nucleosides on the replication of coronaviruses. To that end, we made use of primary human airway epithelial cell (HAEC) cultures grown at the air-liquid interface that were infected with the beta coronavirus OC43. We found that the triple combination of GS-441524 (the parent nucleoside of remdesivir), molnupiravir and ribavirin resulted in a more pronounced antiviral efficacy than what could be expected from a purely additive antiviral effect. The potency of this triple combination was next tested in SARS-CoV-2 infected hamsters in a prophylactic setup. To that end, for each of the drugs, intentionally suboptimal or even ineffective doses were selected. Yet, in the lungs of all hamsters that received triple prophylactic therapy (but not in those that received the respective double combinations) no infectious virus was detectable. Our findings indicate that co-administration of approved drugs for the treatment of coronavirus infections should be further explored but also against other families of viruses with epidemic and pandemic potential for which no effective antiviral treatment is available.
Collapse
Affiliation(s)
- Thuc Nguyen Dan Do
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, 3000, Leuven, Belgium
| | - Rana Abdelnabi
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, 3000, Leuven, Belgium; The VirusBank Platform, Gaston Geenslaan, B-3000, Leuven, Belgium
| | - Bernadett Boda
- Epithelix Sàrl, 18 Chemin des Aulx, Plan-les-Ouates, CH-1228, Geneva, Switzerland
| | - Samuel Constant
- Epithelix Sàrl, 18 Chemin des Aulx, Plan-les-Ouates, CH-1228, Geneva, Switzerland
| | - Johan Neyts
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, 3000, Leuven, Belgium; The VirusBank Platform, Gaston Geenslaan, B-3000, Leuven, Belgium.
| | - Dirk Jochmans
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, 3000, Leuven, Belgium.
| |
Collapse
|
33
|
Huynh PNH, Khamplong P, Phan MH, Nguyen TP, Vu PNL, Tang QV, Chamsodsai P, Seetaha S, Tuong TL, Vu TY, Vo DD, Choowongkomon K, Vo CVT. Asymmetric imidazole-4,5-dicarboxamide derivatives as SARS-CoV-2 main protease inhibitors: design, synthesis and biological evaluation. RSC Med Chem 2024:d4md00414k. [PMID: 39345712 PMCID: PMC11423687 DOI: 10.1039/d4md00414k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
The SARS-CoV-2 main protease, a vital enzyme for virus replication, is a potential target for developing drugs in COVID-19 treatment. Until now, three SARS-CoV-2 main protease inhibitors have been approved for COVID-19 treatment. This study explored the inhibitory potency of asymmetric imidazole-4,5-dicarboxamide derivatives against the SARS-CoV-2 main protease. Fourteen derivatives were designed based on the structure of the SARS-CoV-2 main protease active site, the hydrolysis mechanism, and the experience gained from the reported inhibitor structures. They were synthesized through a four-step procedure from benzimidazole and 2-methylbenzimidazole. SARS-CoV-2 main protease inhibition was evaluated in vitro by fluorogenic assay with lopinavir, ritonavir, and ebselen as positive references. N-(4-Chlorophenyl)-2-methyl-4-(morpholine-4-carbonyl)-1H-imidazole-5-carboxamide (5a2) exhibited the highest potency against the SARS-CoV-2 main protease with an IC50 of 4.79 ± 1.37 μM relative to ebselen with an IC50 of 0.04 ± 0.013 μM. Enzyme kinetic and molecular docking studies were carried out to clarify the inhibitory mechanism and to prove that the compound interacts at the active site. We also performed cytotoxicity assay to confirm that these compounds are not toxic to human cells.
Collapse
Affiliation(s)
- Phuong Nguyen Hoai Huynh
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City Vietnam
| | - Phatcharin Khamplong
- Department of Biochemistry, Faculty of Science, Kasetsart University Bangkok 10900 Thailand
| | - Minh-Hoang Phan
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City Vietnam
| | - Thanh-Phuc Nguyen
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City Vietnam
| | - Phuong Ngoc Lan Vu
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City Vietnam
| | - Quang-Vinh Tang
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City Vietnam
| | - Phumin Chamsodsai
- Genetic Engineering Interdisciplinary Program, Graduate School, Kasetsart University Bangkok 10900 Thailand
| | - Supaphorn Seetaha
- Genetic Engineering Interdisciplinary Program, Graduate School, Kasetsart University Bangkok 10900 Thailand
| | - Truong Lam Tuong
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City Vietnam
| | - Thien Y Vu
- Faculty of Pharmacy, Ton Duc Thang University Ho Chi Minh City Vietnam
| | - Duc-Duy Vo
- Department of Cell and Molecular Biology, Biomedical Centre, Uppsala University Husargatan 3 75237 Uppsala Sweden
- Department of Chemistry, Biomedical Centre, Uppsala University Husargatan 3 75237 Uppsala Sweden
- School of Applied Chemistry, Tra Vinh University 126 Nguyen Thien Thanh Street, Ward 5 Tra Vinh City Vietnam
| | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University Bangkok 10900 Thailand
- Genetic Engineering Interdisciplinary Program, Graduate School, Kasetsart University Bangkok 10900 Thailand
| | - Cam-Van T Vo
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City Vietnam
| |
Collapse
|
34
|
Pellman J, Goldstein A, Słabicki M. Human E3 ubiquitin ligases: accelerators and brakes for SARS-CoV-2 infection. Biochem Soc Trans 2024:BST20230324. [PMID: 39222407 DOI: 10.1042/bst20230324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
E3 ubiquitin ligases regulate the composition of the proteome. These enzymes mono- or poly-ubiquitinate their substrates, directly altering protein function or targeting proteins for degradation by the proteasome. In this review, we discuss the opposing roles of human E3 ligases as effectors and targets in the evolutionary battle between host and pathogen, specifically in the context of SARS-CoV-2 infection. Through complex effects on transcription, translation, and protein trafficking, human E3 ligases can either attenuate SARS-CoV-2 infection or become vulnerabilities that are exploited by the virus to suppress the host's antiviral defenses. For example, the human E3 ligase RNF185 regulates the stability of SARS-CoV-2 envelope protein through the ubiquitin-proteasome pathway, and depletion of RNF185 significantly increases SARS-CoV-2 viral titer (iScience (2023) 26, 106601). We highlight recent advances that identify functions for numerous human E3 ligases in the SARS-CoV-2 life cycle and we assess their potential as novel antiviral agents.
Collapse
Affiliation(s)
- Jesse Pellman
- Broad Institute of MIT and Harvard, Cambridge, MA, U.S.A
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, U.S.A
| | - Anna Goldstein
- Broad Institute of MIT and Harvard, Cambridge, MA, U.S.A
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, U.S.A
| | - Mikołaj Słabicki
- Broad Institute of MIT and Harvard, Cambridge, MA, U.S.A
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, U.S.A
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, U.S.A
| |
Collapse
|
35
|
Huang Y, Wang T, Zhong L, Zhang W, Zhang Y, Yu X, Yuan S, Ni T. Molecular architecture of coronavirus double-membrane vesicle pore complex. Nature 2024; 633:224-231. [PMID: 39143215 PMCID: PMC11374677 DOI: 10.1038/s41586-024-07817-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/11/2024] [Indexed: 08/16/2024]
Abstract
Coronaviruses remodel the intracellular host membranes during replication, forming double-membrane vesicles (DMVs) to accommodate viral RNA synthesis and modifications1,2. SARS-CoV-2 non-structural protein 3 (nsp3) and nsp4 are the minimal viral components required to induce DMV formation and to form a double-membrane-spanning pore, essential for the transport of newly synthesized viral RNAs3-5. The mechanism of DMV pore complex formation remains unknown. Here we describe the molecular architecture of the SARS-CoV-2 nsp3-nsp4 pore complex, as resolved by cryogenic electron tomography and subtomogram averaging in isolated DMVs. The structures uncover an unexpected stoichiometry and topology of the nsp3-nsp4 pore complex comprising 12 copies each of nsp3 and nsp4, organized in 4 concentric stacking hexamer rings, mimicking a miniature nuclear pore complex. The transmembrane domains are interdigitated to create a high local curvature at the double-membrane junction, coupling double-membrane reorganization with pore formation. The ectodomains form extensive contacts in a pseudo-12-fold symmetry, belting the pore complex from the intermembrane space. A central positively charged ring of arginine residues coordinates the putative RNA translocation, essential for virus replication. Our work establishes a framework for understanding DMV pore formation and RNA translocation, providing a structural basis for the development of new antiviral strategies to combat coronavirus infection.
Collapse
Affiliation(s)
- Yixin Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Tongyun Wang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lijie Zhong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Wenxin Zhang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yu Zhang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiulian Yu
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Tao Ni
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, China.
| |
Collapse
|
36
|
Xu L, Yu D, Xu M, Liu Y, Yang LX, Zou QC, Feng XL, Li MH, Sheng N, Yao YG. Primate-specific BTN3A2 protects against SARS-CoV-2 infection by interacting with and reducing ACE2. EBioMedicine 2024; 107:105281. [PMID: 39142074 PMCID: PMC11367481 DOI: 10.1016/j.ebiom.2024.105281] [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: 01/04/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is an immune-related disorder caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The complete pathogenesis of the virus remains to be determined. Unraveling the molecular mechanisms governing SARS-CoV-2 interactions with host cells is crucial for the formulation of effective prophylactic measures and the advancement of COVID-19 therapeutics. METHODS We analyzed human lung single-cell RNA sequencing dataset to discern the association of butyrophilin subfamily 3 member A2 (BTN3A2) expression with COVID-19. The BTN3A2 gene edited cell lines and transgenic mice were infected by live SARS-CoV-2 in a biosafety level 3 (BSL-3) laboratory. Immunoprecipitation, flow cytometry, biolayer interferometry and competition ELISA assays were performed in BTN3A2 gene edited cells. We performed quantitative real-time PCR, histological and/or immunohistochemical analyses for tissue samples from mice with or without SARS-CoV-2 infection. FINDINGS The BTN3A2 mRNA level was correlated with COVID-19 severity. BTN3A2 expression was predominantly identified in epithelial cells, elevated in pathological epithelial cells from COVID-19 patients and co-occurred with ACE2 expression in the same lung cell subtypes. BTN3A2 targeted the early stage of the viral life cycle by inhibiting SARS-CoV-2 attachment through interactions with the receptor-binding domain (RBD) of the Spike protein and ACE2. BTN3A2 inhibited ACE2-mediated SARS-CoV-2 infection by reducing ACE2 in vitro and in vivo. INTERPRETATION These results reveal a key role of BTN3A2 in the fight against COVID-19. Identifying potential monoclonal antibodies which mimic BTN3A2 may facilitate disruption of SARS-CoV-2 infection, providing a therapeutic avenue for COVID-19. FUNDING This study was supported by the National Natural Science Foundation of China (32070569, U1902215, and 32371017), the CAS "Light of West China" Program, and Yunnan Province (202305AH340006).
Collapse
Affiliation(s)
- Ling Xu
- Key Laboratory of Genetic Evolution and Animal Models, Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China; Kunming National High-Level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China.
| | - Dandan Yu
- Key Laboratory of Genetic Evolution and Animal Models, Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China; Kunming National High-Level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Min Xu
- Key Laboratory of Genetic Evolution and Animal Models, Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Yamin Liu
- Key Laboratory of Genetic Evolution and Animal Models, Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Lu-Xiu Yang
- Key Laboratory of Genetic Evolution and Animal Models, Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, China
| | - Qing-Cui Zou
- Kunming National High-Level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Xiao-Li Feng
- Kunming National High-Level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Ming-Hua Li
- Kunming National High-Level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China
| | - Nengyin Sheng
- Key Laboratory of Genetic Evolution and Animal Models, Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China.
| | - Yong-Gang Yao
- Key Laboratory of Genetic Evolution and Animal Models, Key Laboratory of Animal Models and Human Disease Mechanisms of Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650204, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China; Kunming National High-Level Biosafety Research Center for Non-Human Primates, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China; National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), National Resource Center for Non-Human Primates, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650107, China.
| |
Collapse
|
37
|
Guo ZY, Tang YQ, Zhang ZB, Liu J, Zhuang YX, Li T. COVID-19: from immune response to clinical intervention. PRECISION CLINICAL MEDICINE 2024; 7:pbae015. [PMID: 39139990 PMCID: PMC11319938 DOI: 10.1093/pcmedi/pbae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/11/2024] [Indexed: 08/15/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has highlighted the pivotal role of the immune response in determining the progression and severity of viral infections. In this paper, we review the most recent studies on the complicated dynamics between SARS-CoV-2 and the host immune system, highlight the importance of understanding these dynamics in developing effective treatments and formulate potent management strategies for COVID-19. We describe the activation of the host's innate immunity and the subsequent adaptive immune response following infection with SARS-CoV-2. In addition, the review emphasizes the immune evasion strategies of the SARS-CoV-2, including inhibition of interferon production and induction of cytokine storms, along with the resulting clinical outcomes. Finally, we assess the efficacy of current treatment strategies, including antiviral drugs, monoclonal antibodies, and anti-inflammatory treatments, and discuss their role in providing immunity and preventing severe disease.
Collapse
Affiliation(s)
- Zheng-yang Guo
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, China
| | - Yan-qing Tang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, China
| | - Zi-bo Zhang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, China
| | - Juan Liu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, China
| | - Yu-xin Zhuang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau 999078, China
| |
Collapse
|
38
|
Liang Z, Wang J, Zhang H, Gao L, Xu J, Li P, Yang J, Fu X, Duan H, Liu J, Liu T, Ma W, Wu K. Peptide S4 is an entry inhibitor of SARS-CoV-2 infection. Virology 2024; 597:110149. [PMID: 38917689 DOI: 10.1016/j.virol.2024.110149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a significant socioeconomic burden, and combating COVID-19 is imperative. Blocking the SARS-CoV-2 RBD-ACE2 interaction is a promising therapeutic approach for viral infections, as SARS-CoV-2 binds to the ACE2 receptors of host cells via the RBD of spike proteins to infiltrate these cells. We used computer-aided drug design technology and cellular experiments to screen for peptide S4 with high affinity and specificity for the human ACE2 receptor through structural analysis of SARS-CoV-2 and ACE2 interactions. Cellular experiments revealed that peptide S4 effectively inhibited SARS-CoV-2 and HCoV-NL63 viruses from infecting host cells and was safe for cells at effective concentrations. Based on these findings, peptide S4 may be a potential pharmaceutical agent for clinical application in the treatment of the ongoing SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Zhiyu Liang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China; Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jiamei Wang
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Huan Zhang
- Guangdong Center for Disease Control and Prevention, Guangdong, China
| | - Lixia Gao
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jun Xu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Peiran Li
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jie Yang
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Xinting Fu
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Han Duan
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jiayan Liu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China; Institute of Antibody Engineering, School of Laboratory Medicine & Biotechnology, Southern Medical University, Guangzhou, China
| | - Tiancai Liu
- Institute of Antibody Engineering, School of Laboratory Medicine & Biotechnology, Southern Medical University, Guangzhou, China
| | - Weifeng Ma
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| | - Kun Wu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
39
|
Bolinger AA, Li J, Xie X, Li H, Zhou J. Lessons learnt from broad-spectrum coronavirus antiviral drug discovery. Expert Opin Drug Discov 2024; 19:1023-1041. [PMID: 39078037 PMCID: PMC11390334 DOI: 10.1080/17460441.2024.2385598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
INTRODUCTION Highly pathogenic coronaviruses (CoVs), such as severe acute respiratory syndrome CoV (SARS-CoV), Middle East respiratory syndrome CoV (MERS-CoV), and the most recent SARS-CoV-2 responsible for the COVID-19 pandemic, pose significant threats to human populations over the past two decades. These CoVs have caused a broad spectrum of clinical manifestations ranging from asymptomatic to severe distress syndromes (ARDS), resulting in high morbidity and mortality. AREAS COVERED The accelerated advancements in antiviral drug discovery, spurred by the COVID-19 pandemic, have shed new light on the imperative to develop treatments effective against a broad spectrum of CoVs. This perspective discusses strategies and lessons learnt in targeting viral non-structural proteins, structural proteins, drug repurposing, and combinational approaches for the development of antivirals against CoVs. EXPERT OPINION Drawing lessons from the pandemic, it becomes evident that the absence of efficient broad-spectrum antiviral drugs increases the vulnerability of public health systems to the potential onslaught by highly pathogenic CoVs. The rapid and sustained spread of novel CoVs can have devastating consequences without effective and specifically targeted treatments. Prioritizing the effective development of broad-spectrum antivirals is imperative for bolstering the resilience of public health systems and mitigating the potential impact of future highly pathogenic CoVs.
Collapse
Affiliation(s)
- Andrew A. Bolinger
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jun Li
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Hongmin Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
40
|
Otsuka Y, Zhang L, Mou H, Shumate J, Kitzmiller CE, Scampavia L, Bannister TD, Farzan M, Choe H, Spicer TP. Simultaneous screening for selective SARS-CoV-2, Lassa, and Machupo virus entry inhibitors. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100178. [PMID: 39159824 DOI: 10.1016/j.slasd.2024.100178] [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: 05/24/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024]
Abstract
Emerging highly pathogenic viruses can pose profound impacts on global health, the economy, and society. To meet that challenge, the National Institute of Allergy and Infectious Diseases (NIAID) established nine Antiviral Drug Discovery (AViDD) centers for early-stage identification and validation of novel antiviral drug candidates against viruses with pandemic potential. As part of this initiative, we established paired entry assays that simultaneously screen for inhibitors specifically targeting SARS-CoV-2 (SARS2), Lassa virus (LASV) and Machupo virus (MACV) entry. To do so we employed a dual pseudotyped virus (PV) infection system allowing us to screen ∼650,000 compounds efficiently and cost-effectively. Adaptation of these paired assays into 1536 well-plate format for ultra-high throughput screening (uHTS) resulted in the largest screening ever conducted in our facility, with over 2.4 million wells completed. The paired infection system allowed us to detect two PV infections simultaneously: LASV + MACV, MACV + SARS2, and SARS2 + LASV. Each PV contains a different luciferase reporter gene which enabled us to measure the infection of each PV exclusively, albeit in the same well. Each PV was screened at least twice utilizing different reporters, which allowed us to select the inhibitors specific to a particular PV and to exclude those that hit off targets, including cellular components or the reporter proteins. All assays were robust with an average Z' value ranging from 0.5 to 0.8. The primary screening of ∼650,000 compounds resulted in 1812, 1506, and 2586 unique hits for LASV, MACV, and SARS2, respectively. The confirmation screening narrowed this list further to 60, 40, and 90 compounds that are unique to LASV, MACV, and SARS2, respectively. Of these compounds, 8, 35, and 50 compounds showed IC50 value < 10 μM, some of which have much greater potency and excellent antiviral activity profiles specific to LASV, MACV, and SARS2, and none are cytotoxic. These selected compounds are currently being studied for their mechanism of action and to improve their specificity and potency through chemical modification.
Collapse
Affiliation(s)
- Yuka Otsuka
- Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL 33458, United States
| | - Lizhou Zhang
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, United States; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, United States
| | - Huihui Mou
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, United States; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, United States
| | - Justin Shumate
- Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL 33458, United States
| | - Claire E Kitzmiller
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, United States
| | - Louis Scampavia
- Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL 33458, United States
| | - Thomas D Bannister
- Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL 33458, United States
| | - Michael Farzan
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, United States; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, United States
| | - Hyeryun Choe
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA 02115, United States; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, United States
| | - Timothy P Spicer
- Department of Molecular Medicine, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL 33458, United States.
| |
Collapse
|
41
|
Otsuka Y, Kim E, Krueger A, Shumate J, Wang C, Bdiri B, Ullah S, Park H, Scampavia L, Bannister TD, Chung D, Spicer TP. High throughput screening for SARS-CoV-2 helicase inhibitors. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100180. [PMID: 39173831 DOI: 10.1016/j.slasd.2024.100180] [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: 03/26/2024] [Revised: 07/26/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for nearly 7 million deaths worldwide since its outbreak in late 2019. Even with the rapid development and production of vaccines and intensive research, there is still a huge need for specific anti-viral drugs that address the rapidly arising new variants. To address this concern, the National Institute of Allergy and Infectious Diseases (NIAID) established nine Antiviral Drug Discovery (AViDD) Centers, tasked with exploring approaches to target pathogens with pandemic potential, including SARS-CoV-2. In this study, we sought inhibitors of SARS-CoV2 non-structural protein 13 (nsP13) as potential antivirals, first developing a HTS-compatible assay to measure SARS-CoV2 nsP13 helicase activity. Here we present our effort in implementing the assay in a 1,536 well-plate format and in identifying nsP13 inhibitor hit compounds from a ∼650,000 compound library. The primary screen was robust (average Z' = 0.86 ± 0.05) and resulted in 7,009 primary hits. 1,763 of these compounds upon repeated retests were further confirmed, showing consistent inhibition. Following in-silico analysis, an additional orthogonal assay and titration assays, we identified 674 compounds with IC50 <10 μM. We confirmed activity of independent compound batches from de novo powders while also incorporating multiple counterscreen assays. Our study highlights the potential of this assay for use on HTS platforms to discover novel compounds inhibiting SARS-CoV2 nsP13, which merit further development as an effective SARS-CoV2 antiviral.
Collapse
Affiliation(s)
- Yuka Otsuka
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Department of Molecular Medicine, Midwest AViDD HTS Core B, Jupiter, FL 33458, United States
| | - Eunjung Kim
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Department of Chemistry, Midwest AViDD Chemistry Core C, Jupiter, FL 33458, United States
| | - Austin Krueger
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Department of Chemistry, Midwest AViDD Chemistry Core C, Jupiter, FL 33458, United States
| | - Justin Shumate
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Department of Molecular Medicine, Midwest AViDD HTS Core B, Jupiter, FL 33458, United States
| | - Chao Wang
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Department of Chemistry, Midwest AViDD Chemistry Core C, Jupiter, FL 33458, United States
| | - Bilel Bdiri
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Department of Chemistry, Midwest AViDD Chemistry Core C, Jupiter, FL 33458, United States
| | - Sultan Ullah
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Department of Chemistry, Midwest AViDD Chemistry Core C, Jupiter, FL 33458, United States
| | - HaJeung Park
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Department of Chemistry, Midwest AViDD Chemistry Core C, Jupiter, FL 33458, United States
| | - Louis Scampavia
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Department of Molecular Medicine, Midwest AViDD HTS Core B, Jupiter, FL 33458, United States
| | - Thomas D Bannister
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Department of Chemistry, Midwest AViDD Chemistry Core C, Jupiter, FL 33458, United States
| | - Donghoon Chung
- Center for Predictive Medicine, Department of Microbiology Immunology, School of Medicine, Midwest AViDD Project 5, University of Louisville, Louisville, KY 40202, United States
| | - Timothy P Spicer
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Department of Molecular Medicine, Midwest AViDD HTS Core B, Jupiter, FL 33458, United States.
| |
Collapse
|
42
|
Choi G, Rejinold NS, Piao H, Ryu YB, Kwon HJ, Lee IC, Seo JI, Yoo HH, Jin GW, Choy JH. The Next Generation COVID-19 Antiviral; Niclosamide-Based Inorganic Nanohybrid System Kills SARS-CoV-2. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305148. [PMID: 37635100 DOI: 10.1002/smll.202305148] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/31/2023] [Indexed: 08/29/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is a serious global threat with surging new variants of concern. Although global vaccinations have slowed the pandemic, their longevity is still unknown. Therefore, new orally administrable antiviral agents are highly demanded. Among various repurposed drugs, niclosamide (NIC) is the most potential one for various viral diseases such as COVID-19, SARS (severe acute respiratory syndrome), MERS (middle east respiratory syndrome), influenza, RSV (respiratory syncytial virus), etc. Since NIC cannot be effectively absorbed, a required plasma concentration for antiviral potency is hard to maintain, thereby restricting its entry into the infected cells. Such a 60-year-old bioavailability challenging issue has been overcome by engineering with MgO and hydroxypropyl methylcellulose (HPMC), forming hydrophilic NIC-MgO-HPMC, with improved intestinal permeability without altering NIC metabolism as confirmed by parallel artificial membrane permeability assay. The inhibitory effect on SARS-CoV-2 replication is confirmed in the Syrian hamster model to reduce lung injury. Clinical studies reveal that the bioavailability of NIC hybrid drug can go 4 times higher than the intact NIC. The phase II clinical trial shows a dose-dependent bioavailability of NIC from hybrid drug suggesting its potential applicability as a game changer in achieving the much-anticipated endemic phase.
Collapse
Affiliation(s)
- Goeun Choi
- Intelligent Nanohybrid Materials Laboratory (INML), Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- College of Science and Technology, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - N Sanoj Rejinold
- Intelligent Nanohybrid Materials Laboratory (INML), Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Huiyan Piao
- Intelligent Nanohybrid Materials Laboratory (INML), Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
| | - Young Bae Ryu
- Functional Biomaterials Research Center, Korea Research Institute of Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 34141, Republic of Korea
| | - Hyung-Jun Kwon
- Functional Biomaterials Research Center, Korea Research Institute of Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 34141, Republic of Korea
| | - In Chul Lee
- Functional Biomaterials Research Center, Korea Research Institute of Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 34141, Republic of Korea
| | - Jeong In Seo
- Institute of Pharmaceutical Science and Technology, College of Pharmacy, Hanyang University, Ansan, 15588, Republic of Korea
| | - Hye Hyun Yoo
- Institute of Pharmaceutical Science and Technology, College of Pharmacy, Hanyang University, Ansan, 15588, Republic of Korea
| | - Geun-Woo Jin
- R&D Center, CnPharm Co. LTD., Seoul, 03759, Republic of Korea
| | - Jin-Ho Choy
- Intelligent Nanohybrid Materials Laboratory (INML), Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea
- Department of Pre-Medical Course, College of Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- International Research Frontier Initiative (IRFI), Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, 226-8503, Japan
| |
Collapse
|
43
|
Rut W, Groborz K, Sun X, Hilgenfeld R, Drag M. Profiling of coronaviral M pro and enteroviral 3C pro specificity provides a framework for the development of broad-spectrum antiviral compounds. Protein Sci 2024; 33:e5139. [PMID: 39150063 PMCID: PMC11328108 DOI: 10.1002/pro.5139] [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: 05/06/2024] [Revised: 07/04/2024] [Accepted: 07/20/2024] [Indexed: 08/17/2024]
Abstract
The main protease from coronaviruses and the 3C protease from enteroviruses play a crucial role in processing viral polyproteins, making them attractive targets for the development of antiviral agents. In this study, we employed a combinatorial chemistry approach-HyCoSuL-to compare the substrate specificity profiles of the main and 3C proteases from alphacoronaviruses, betacoronaviruses, and enteroviruses. The obtained data demonstrate that coronavirus Mpros exhibit overlapping substrate specificity in all binding pockets, whereas the 3Cpro from enterovirus displays slightly different preferences toward natural and unnatural amino acids at the P4-P2 positions. However, chemical tools such as substrates, inhibitors, and activity-based probes developed for SARS-CoV-2 Mpro can be successfully applied to investigate the activity of the Mpro from other coronaviruses as well as the 3Cpro from enteroviruses. Our study provides a structural framework for the development of broad-spectrum antiviral compounds.
Collapse
Affiliation(s)
- Wioletta Rut
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Katarzyna Groborz
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Xinyuanyuan Sun
- Institute of Molecular Medicine, University of Lübeck, Lübeck, Germany
| | - Rolf Hilgenfeld
- Institute of Molecular Medicine, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems Site, University of Lübeck, Lübeck, Germany
| | - Marcin Drag
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| |
Collapse
|
44
|
Delgado R, Vishwakarma J, Moghadasi SA, Otsuka Y, Shumate J, Cuell A, Tansiongco M, Cooley CB, Chen Y, Dabrowska A, Basu R, Anindita PD, Luo D, Dosa PI, Harki DA, Bannister T, Scampavia L, Spicer TP, Harris RS. SARS-CoV-2 M pro inhibitor identification using a cellular gain-of-signal assay for high-throughput screening. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100181. [PMID: 39173830 DOI: 10.1016/j.slasd.2024.100181] [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: 05/22/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2, SARS2) is responsible for the COVID-19 pandemic and infections that continue to affect the lives of millions of people worldwide, especially those who are older and/or immunocompromised. The SARS2 main protease enzyme, Mpro (also called 3C-like protease, 3CLpro), is a bona fide drug target as evidenced by potent inhibition with nirmatrelvir and ensitrelvir, the active components of the drugs Paxlovid and Xocova, respectively. However, the existence of nirmatrelvir and ensitrelvir-resistant isolates underscores the need to develop next-generation drugs with different resistance profiles and/or distinct mechanisms of action. Here, we report the results of a high-throughput screen of 649,568 compounds using a cellular gain-of-signal assay. In this assay, Mpro inhibits expression of a luciferase reporter, and 8,777 small molecules were considered hits by causing a gain in luciferase activity 3x SD above the sample field activity (6.8% gain-of-signal relative to 100 µM GC376). Single concentration and dose-response gain-of-signal experiments confirmed 3,522/8,762 compounds as candidate inhibitors. In parallel, all initial high-throughput screening hits were tested in a peptide cleavage assay with purified Mpro and only 39/8,762 showed inhibition. Importantly, 19/39 compounds (49%) re-tested positive in both SARS2 assays, including two previously reported Mpro inhibitors, demonstrating the efficacy of the overall screening strategy. This approach led to the rediscovery of known Mpro inhibitors such as calpain inhibitor II, as well as to the discovery of novel compounds that provide chemical information for future drug development efforts.
Collapse
Affiliation(s)
- Renee Delgado
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Jyoti Vishwakarma
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Seyed Arad Moghadasi
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA; Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | - Yuka Otsuka
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
| | - Justin Shumate
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
| | - Ashley Cuell
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Megan Tansiongco
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Department of Chemistry, Trinity University, San Antonio, TX 78212, USA
| | | | - Yanjun Chen
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Agnieszka Dabrowska
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Rahul Basu
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Paulina Duhita Anindita
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore; Institute of Structural Biology, Nanyang Technological University, Singapore, 639798, Singapore
| | - Dahai Luo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore; Institute of Structural Biology, Nanyang Technological University, Singapore, 639798, Singapore
| | - Peter I Dosa
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel A Harki
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Thomas Bannister
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
| | - Louis Scampavia
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
| | - Timothy P Spicer
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL 33458, USA
| | - Reuben S Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX 78229, USA; Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
45
|
Stancheva VG, Sanyal S. Positive-strand RNA virus replication organelles at a glance. J Cell Sci 2024; 137:jcs262164. [PMID: 39254430 PMCID: PMC11423815 DOI: 10.1242/jcs.262164] [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] [Indexed: 09/11/2024] Open
Abstract
Membrane-bound replication organelles (ROs) are a unifying feature among diverse positive-strand RNA viruses. These compartments, formed as alterations of various host organelles, provide a protective niche for viral genome replication. Some ROs are characterised by a membrane-spanning pore formed by viral proteins. The RO membrane separates the interior from immune sensors in the cytoplasm. Recent advances in imaging techniques have revealed striking diversity in RO morphology and origin across virus families. Nevertheless, ROs share core features such as interactions with host proteins for their biogenesis and for lipid and energy transfer. The restructuring of host membranes for RO biogenesis and maintenance requires coordinated action of viral and host factors, including membrane-bending proteins, lipid-modifying enzymes and tethers for interorganellar contacts. In this Cell Science at a Glance article and the accompanying poster, we highlight ROs as a universal feature of positive-strand RNA viruses reliant on virus-host interplay, and we discuss ROs in the context of extensive research focusing on their potential as promising targets for antiviral therapies and their role as models for understanding fundamental principles of cell biology.
Collapse
Affiliation(s)
- Viktoriya G. Stancheva
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| |
Collapse
|
46
|
Yang L, He J, Liu J, Xie T, Tang Q. Application of chimeric antigen receptor therapy beyond oncology: A bibliometric and visualized analysis. Curr Res Transl Med 2024; 72:103442. [PMID: 38452444 DOI: 10.1016/j.retram.2024.103442] [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: 09/16/2023] [Revised: 01/17/2024] [Accepted: 02/04/2024] [Indexed: 03/09/2024]
Abstract
PURPOSE Chimeric antigen receptor therapy beyond oncology has gained increasing attention. While a substantial number of publications have emerged in recent years, there has been a paucity of conducted bibliometric studies. Our objective is to systematically summarize and visually analyze the literature in the field of chimeric antigen receptors therapy beyond oncology and explore hotspots in this field. METHODS Web of Science Core Collection was selected as the data source, and the data was retrieved on December 23, 2022, according to the search strategy. CiteSpace 6.1.R6 and Vosviewer 1.6.18 were used to analyze publications and explore research hotspots and directions. RESULTS A total of 338 publications written by 1832 authors from 516 institutions in 42 countries/regions were selected for the analysis. The number of publications is steadily increasing annually. The United States emerged as the primary contributor, and University of Pennsylvania was the leading institution. Frontiers in Immunology boasted the highest number of published papers. Kitchen SG, Riley JL, and Scott DW were the most productive researchers in this field. The keyword cluster analysis identified HIV, autoimmune diseases, transplant related diseases and COVID-19 as the primary focus areas within the realm of chimeric antigen receptor therapy beyond oncology. CONCLUSION The advancement of chimeric antigen receptor therapy beyond oncology has witnessed rapid progress in recent years. We have explored the hotspots and research directions in this field. It is hoped that this study could provide references and directions for future clinical researches.
Collapse
Affiliation(s)
- Linxin Yang
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Systemic Autoimmune Diseases in Hunan Province, Changsha, Hunan, China
| | - Jinshen He
- Department of Orthopaedic Surgery, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiahao Liu
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Systemic Autoimmune Diseases in Hunan Province, Changsha, Hunan, China
| | - Tianjian Xie
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Systemic Autoimmune Diseases in Hunan Province, Changsha, Hunan, China
| | - Qi Tang
- Department of Rheumatology and Immunology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Clinical Medical Research Center for Systemic Autoimmune Diseases in Hunan Province, Changsha, Hunan, China.
| |
Collapse
|
47
|
Suryawanshi RK, Jaishankar P, Correy GJ, Rachman MM, O’Leary PC, Taha TY, Zapatero-Belinchón FJ, McCavittMalvido M, Doruk YU, Stevens MGV, Diolaiti ME, Jogalekar MP, Richards AL, Montano M, Rosecrans J, Matthay M, Togo T, Gonciarz RL, Gopalkrishnan S, Neitz RJ, Krogan NJ, Swaney DL, Shoichet BK, Ott M, Renslo AR, Ashworth A, Fraser JS. The Mac1 ADP-ribosylhydrolase is a Therapeutic Target for SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.606661. [PMID: 39149230 PMCID: PMC11326214 DOI: 10.1101/2024.08.08.606661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
SARS-CoV-2 continues to pose a threat to public health. Current therapeutics remain limited to direct acting antivirals that lack distinct mechanisms of action and are already showing signs of viral resistance. The virus encodes an ADP-ribosylhydrolase macrodomain (Mac1) that plays an important role in the coronaviral lifecycle by suppressing host innate immune responses. Genetic inactivation of Mac1 abrogates viral replication in vivo by potentiating host innate immune responses. However, it is unknown whether this can be achieved by pharmacologic inhibition and can therefore be exploited therapeutically. Here we report a potent and selective lead small molecule, AVI-4206, that is effective in an in vivo model of SARS-CoV-2 infection. Cellular models indicate that AVI-4206 has high target engagement and can weakly inhibit viral replication in a gamma interferon- and Mac1 catalytic activity-dependent manner; a stronger antiviral effect for AVI-4206 is observed in human airway organoids. In an animal model of severe SARS-CoV-2 infection, AVI-4206 reduces viral replication, potentiates innate immune responses, and leads to a survival benefit. Our results provide pharmacological proof of concept that Mac1 is a valid therapeutic target via a novel immune-restoring mechanism that could potentially synergize with existing therapies targeting distinct, essential aspects of the coronaviral life cycle. This approach could be more widely used to target other viral macrodomains to develop antiviral therapeutics beyond COVID-19.
Collapse
Affiliation(s)
| | - Priyadarshini Jaishankar
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Galen J. Correy
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA
| | - Moira M. Rachman
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Patrick C. O’Leary
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA
| | - Taha Y. Taha
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA
- Quantitative Biosciences Institute (QBI), University of California San Francisco, San Francisco, CA
| | | | | | - Yagmur U. Doruk
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA
| | - Maisie G. V. Stevens
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA
| | - Morgan E. Diolaiti
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA
| | - Manasi P. Jogalekar
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA
| | - Alicia L. Richards
- Quantitative Biosciences Institute (QBI), University of California San Francisco, San Francisco, CA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA
- Data Science and Biotechnology Institute, Gladstone Institutes, San Francisco, CA
| | - Mauricio Montano
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA
| | - Julia Rosecrans
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA
| | - Michael Matthay
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Takaya Togo
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Ryan L. Gonciarz
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Saumya Gopalkrishnan
- Quantitative Biosciences Institute (QBI), University of California San Francisco, San Francisco, CA
| | - R. Jeffrey Neitz
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Small Molecule Discovery Center, University of California San Francisco, San Francisco, CA
| | - Nevan J. Krogan
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA
- Quantitative Biosciences Institute (QBI), University of California San Francisco, San Francisco, CA
- Data Science and Biotechnology Institute, Gladstone Institutes, San Francisco, CA
| | - Danielle L. Swaney
- Quantitative Biosciences Institute (QBI), University of California San Francisco, San Francisco, CA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA
- Data Science and Biotechnology Institute, Gladstone Institutes, San Francisco, CA
| | - Brian K. Shoichet
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Melanie Ott
- Gladstone Institute of Virology, Gladstone Institutes, San Francisco, CA
- Department of Medicine, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub- San Francisco, San Francisco, CA
| | - Adam R. Renslo
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Alan Ashworth
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA
| | - James S. Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA
| |
Collapse
|
48
|
Greenhalgh T, Sivan M, Perlowski A, Nikolich JŽ. Long COVID: a clinical update. Lancet 2024; 404:707-724. [PMID: 39096925 DOI: 10.1016/s0140-6736(24)01136-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/07/2024] [Accepted: 05/30/2024] [Indexed: 08/05/2024]
Abstract
Post-COVID-19 condition (also known as long COVID) is generally defined as symptoms persisting for 3 months or more after acute COVID-19. Long COVID can affect multiple organ systems and lead to severe and protracted impairment of function as a result of organ damage. The burden of this disease, both on the individual and on health systems and national economies, is high. In this interdisciplinary Review, with a coauthor with lived experience of severe long COVID, we sought to bring together multiple streams of literature on the epidemiology, pathophysiology (including the hypothesised mechanisms of organ damage), lived experience and clinical manifestations, and clinical investigation and management of long COVID. Although current approaches to long COVID care are largely symptomatic and supportive, recent advances in clinical phenotyping, deep molecular profiling, and biomarker identification might herald a more mechanism-informed and personally tailored approach to clinical care. We also cover the organisation of services for long COVID, approaches to preventing long COVID, and suggestions for future research.
Collapse
Affiliation(s)
- Trisha Greenhalgh
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, Oxford, UK.
| | - Manoj Sivan
- Academic Department of Rehabilitation Medicine, Leeds Institute of Rheumatic and Musculoskeletal Medicine University of Leeds, Leeds General Infirmary, Leeds, UK
| | | | - Janko Ž Nikolich
- Department of Immunobiology and University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The Aegis Consortium for Pandemic-Free Future, University of Arizona Health Sciences, Tucson, AZ, USA
| |
Collapse
|
49
|
Friedhoff R, Elfayres G, Mérindol N, Desgagné-Penix I, Berthoux L. RNA replication-independent, DNA linearization-dependent expression of reporter genes from a SARS-CoV-2 replicon-encoding DNA in human cells. PLoS One 2024; 19:e0300491. [PMID: 39150942 PMCID: PMC11329111 DOI: 10.1371/journal.pone.0300491] [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: 02/28/2024] [Accepted: 07/13/2024] [Indexed: 08/18/2024] Open
Abstract
Replicons, derived from RNA viruses, are genetic constructs retaining essential viral enzyme genes while lacking key structural protein genes. Upon introduction into cells, the genes carried by the replicon RNA are expressed, and the RNA self-replicates, yet viral particle production does not take place. Typically, RNA replicons are transcribed in vitro and are then electroporated in cells. However, it would be advantageous for the replicon to be generated in cells following DNA transfection instead of RNA. In this study, a bacterial artificial chromosome (BAC) DNA encoding a SARS-CoV-2 replicon under control of a T7 promoter was transfected into HEK293T cells engineered to functionally express the T7 RNA polymerase (T7 RNAP). Upon transfection of the BAC DNA, we observed low, but reproducible expression of reporter proteins GFP and luciferase carried by this replicon. Expression of the reporter proteins required linearization of the BAC DNA prior to transfection. Moreover, expression occurred independently of T7 RNAP. Gene expression was also insensitive to remdesivir treatment, suggesting that it did not involve self-replication of replicon RNA. Similar results were obtained in highly SARS-CoV-2 infection-permissive Calu-3 cells. Strikingly, prior expression of the SARS-CoV-2 N protein boosted expression from transfected SARS-CoV-2 RNA replicon but not from the replicon BAC DNA. In conclusion, transfection of a large DNA encoding a coronaviral replicon led to reproducible replicon gene expression through an unidentified mechanism. These findings highlight a novel pathway toward replicon gene expression from transfected replicon cDNA, offering valuable insights for the development of methods for DNA-based RNA replicon applications.
Collapse
Affiliation(s)
- Ronja Friedhoff
- Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Ghada Elfayres
- Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Natacha Mérindol
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Isabel Desgagné-Penix
- Department of Chemistry, Biochemistry and Physics, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Lionel Berthoux
- Department of Medical Biology, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| |
Collapse
|
50
|
Yu F, Liu X, Ou H, Li X, Liu R, Lv X, Xiao S, Hu M, Liang T, Chen T, Wei X, Zhang Z, Liu S, Liu H, Zhu Y, Liu G, Tu T, Li P, Zhang H, Pan T, Ma X. The histamine receptor H1 acts as an alternative receptor for SARS-CoV-2. mBio 2024; 15:e0108824. [PMID: 38953634 PMCID: PMC11324024 DOI: 10.1128/mbio.01088-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/29/2024] [Indexed: 07/04/2024] Open
Abstract
Numerous host factors, in addition to human angiotensin-converting enzyme 2 (hACE2), have been identified as coreceptors of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), demonstrating broad viral tropism and diversified druggable potential. We and others have found that antihistamine drugs, particularly histamine receptor H1 (HRH1) antagonists, potently inhibit SARS-CoV-2 infection. In this study, we provided compelling evidence that HRH1 acts as an alternative receptor for SARS-CoV-2 by directly binding to the viral spike protein. HRH1 also synergistically enhanced hACE2-dependent viral entry by interacting with hACE2. Antihistamine drugs effectively prevent viral infection by competitively binding to HRH1, thereby disrupting the interaction between the spike protein and its receptor. Multiple inhibition assays revealed that antihistamine drugs broadly inhibited the infection of various SARS-CoV-2 mutants with an average IC50 of 2.4 µM. The prophylactic function of these drugs was further confirmed by authentic SARS-CoV-2 infection assays and humanized mouse challenge experiments, demonstrating the therapeutic potential of antihistamine drugs for combating coronavirus disease 19.IMPORTANCEIn addition to human angiotensin-converting enzyme 2, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can utilize alternative cofactors to facilitate viral entry. In this study, we discovered that histamine receptor H1 (HRH1) not only functions as an independent receptor for SARS-CoV-2 but also synergistically enhances ACE2-dependent viral entry by directly interacting with ACE2. Further studies have demonstrated that HRH1 facilitates the entry of SARS-CoV-2 by directly binding to the N-terminal domain of the spike protein. Conversely, antihistamine drugs, primarily HRH1 antagonists, can competitively bind to HRH1 and thereby prevent viral entry. These findings revealed that the administration of repurposable antihistamine drugs could be a therapeutic intervention to combat coronavirus disease 19.
Collapse
Affiliation(s)
- Fei Yu
- Medical Research
Institute, Guangdong Provincial People’s Hospital (Guangdong
Academy of Medical Sciences), Southern Medical
University, Guangzhou,
Guangdong, China
| | - Xiaoqing Liu
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
- Institute of Human
Virology, Zhongshan School of Medicine, Sun Yat-sen
University, Guangzhou,
Guangdong, China
| | - Hailan Ou
- Medical Research
Institute, Guangdong Provincial People’s Hospital (Guangdong
Academy of Medical Sciences), Southern Medical
University, Guangzhou,
Guangdong, China
| | - Xinyu Li
- Shenzhen Key
Laboratory of Systems Medicine for Inflammatory Diseases, Shenzhen
Campus of Sun Yat-sen University,
Shenzhen, Guangdong,
China
| | - Ruxin Liu
- Shenzhen Key
Laboratory of Systems Medicine for Inflammatory Diseases, Shenzhen
Campus of Sun Yat-sen University,
Shenzhen, Guangdong,
China
| | - Xi Lv
- Medical Research
Institute, Guangdong Provincial People’s Hospital (Guangdong
Academy of Medical Sciences), Southern Medical
University, Guangzhou,
Guangdong, China
- School of Medicine,
South China University of Technology,
Guangzhou, Guangdong,
China
| | - Shiqi Xiao
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
| | - Meilin Hu
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
- Department of Breast
Surgery, The Second Affiliated Hospital of Guangzhou Medical
University, Guangzhou,
Guangdong, China
| | - Taizhen Liang
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
- State Key Laboratory
of Respiratory Disease, National Clinical Research Center for
Respiratory Disease, Guangzhou Institute of Respiratory Health, the
First Affiliated Hospital of Guangzhou Medical
University, Guangzhou,
Guangdong, China
| | - Tao Chen
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
- State Key Laboratory
of Respiratory Disease, National Clinical Research Center for
Respiratory Disease, Guangzhou Institute of Respiratory Health, the
First Affiliated Hospital of Guangzhou Medical
University, Guangzhou,
Guangdong, China
| | - Xuepeng Wei
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
| | - Zhenglai Zhang
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
| | - Sen Liu
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
- School of Biology and
Biological Engineering, South China University of
Technology, Guangzhou,
Guangdong, China
| | - Han Liu
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
| | - Yiqiang Zhu
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
| | - Guangyan Liu
- Department of Pathogen
Biology, Shenyang Medical College,
Shenyang, Liaoning,
China
| | - Tianyong Tu
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
| | - Peiwen Li
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
| | - Hui Zhang
- Institute of Human
Virology, Zhongshan School of Medicine, Sun Yat-sen
University, Guangzhou,
Guangdong, China
| | - Ting Pan
- Shenzhen Key
Laboratory of Systems Medicine for Inflammatory Diseases, Shenzhen
Campus of Sun Yat-sen University,
Shenzhen, Guangdong,
China
| | - Xiancai Ma
- Medical Research
Institute, Guangdong Provincial People’s Hospital (Guangdong
Academy of Medical Sciences), Southern Medical
University, Guangzhou,
Guangdong, China
- Guangzhou National
Laboratory, Guangzhou International
Bio-Island, Guangzhou,
Guangdong, China
- State Key Laboratory
of Respiratory Disease, National Clinical Research Center for
Respiratory Disease, Guangzhou Institute of Respiratory Health, the
First Affiliated Hospital of Guangzhou Medical
University, Guangzhou,
Guangdong, China
| |
Collapse
|