151
|
Jumde RP, Jézéquel G, Saramago M, Frank N, Adam S, Cunha MV, Bader CD, Gunesch AP, Köhler NM, Johannsen S, Bousis S, Pietschmann T, Matos RG, Müller R, Arraiano CM, Hirsch AKH. Dynamic Combinatorial Chemistry Unveils Nsp10 Inhibitors with Antiviral Potential Against SARS-CoV-2. Chemistry 2025; 31:e202403390. [PMID: 39676060 PMCID: PMC11739841 DOI: 10.1002/chem.202403390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
The development of antiviral drugs against the Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) responsible for the recent Covid-19 pandemic is crucial, as treatment options remain limited and vaccination does not prevent (re)infection. Two relatively underexplored targets of this virus are the 3'-5' exoribonuclease (ExoN) and the 2'-O-methyltransferase (2'-O-MTase), both essential for viral viability. The non-structural proteins Nsp14 and Nsp16 exhibit enzymatic activities for ExoN and 2'-O-MTase, respectively, especially when in complex with their co-factor protein Nsp10. The study focuses on the use of target-directed dynamic combinatorial chemistry (tdDCC) to identify binders of Nsp10, aiming to disturb the protein-protein interactions (PPI) involving Nsp10-Nsp14, as well as Nsp10-Nsp16. We synthesised the hits and evaluated them to assess Nsp10 affinity, ExoN and 2'-O-MTase activities inhibition, and antiviral activity in hCoV-229E and SARS-CoV-2-infected whole-cell settings. This study reports a novel class of ExoN and/or 2'-O-MTase inhibitors exhibiting antiviral activity against coronaviruses.
Collapse
Affiliation(s)
- Ravindra P. Jumde
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
- Current addressGlobal Antibiotic Research & Development Partnership (GARDP)Chemin Camille-Vidart 151202GenevaSwitzerland
| | - Gwenaëlle Jézéquel
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
| | - Margarida Saramago
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAvenida da República2780-157OeirasPortugal
| | - Nicolas Frank
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
- Saarland UniversityDepartment of PharmacyCampus E 8.166123SaarbrückenGermany
| | - Sebastian Adam
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
| | - Marta V. Cunha
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAvenida da República2780-157OeirasPortugal
| | - Chantal D. Bader
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
| | - Antonia P. Gunesch
- Institute for Experimental VirologyTwincore – Centre for Experimental and Clinical Infection ResearchFeodor-Lynen-Str. 730625HannoverGermany
| | - Natalie M. Köhler
- Institute for Experimental VirologyTwincore – Centre for Experimental and Clinical Infection ResearchFeodor-Lynen-Str. 730625HannoverGermany
| | - Sandra Johannsen
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
- Saarland UniversityDepartment of PharmacyCampus E 8.166123SaarbrückenGermany
| | - Spyridon Bousis
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
- Saarland UniversityDepartment of PharmacyCampus E 8.166123SaarbrückenGermany
| | - Thomas Pietschmann
- Institute for Experimental VirologyTwincore – Centre for Experimental and Clinical Infection ResearchFeodor-Lynen-Str. 730625HannoverGermany
- Cluster of Excellence RESIST (EXC 2155)Hannover Medical School30625HannoverGermany
- Helmholtz International Lab for Anti-infectivesCampus E 8.166123SaarbrückenGermany
| | - Rute G. Matos
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAvenida da República2780-157OeirasPortugal
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
- Saarland UniversityDepartment of PharmacyCampus E 8.166123SaarbrückenGermany
- Cluster of Excellence RESIST (EXC 2155)Hannover Medical School30625HannoverGermany
- Helmholtz International Lab for Anti-infectivesCampus E 8.166123SaarbrückenGermany
| | - Cecília M. Arraiano
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaAvenida da República2780-157OeirasPortugal
| | - Anna K. H. Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) – Helmholtz Centre for Infection Research (HZI)Campus E 8.166123SaarbrückenGermany
- Saarland UniversityDepartment of PharmacyCampus E 8.166123SaarbrückenGermany
- Cluster of Excellence RESIST (EXC 2155)Hannover Medical School30625HannoverGermany
- Helmholtz International Lab for Anti-infectivesCampus E 8.166123SaarbrückenGermany
| |
Collapse
|
152
|
Kim SW, Han MJ, Rahman MS, Kim H, Noh JE, Lee MK, Kim M, Lee JO, Jang SK. Ultra-Sensitive Aptamer-Based Diagnostic Systems for Rapid Detection of All SARS-CoV-2 Variants. Int J Mol Sci 2025; 26:745. [PMID: 39859459 PMCID: PMC11766214 DOI: 10.3390/ijms26020745] [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: 11/27/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
The emergence of numerous SARS-CoV-2 variants, characterized by mutations in the viral RNA genome and target proteins, has presented challenges for accurate COVID-19 diagnosis. To address this, we developed universal aptamer probes capable of binding to the spike proteins of SARS-CoV-2 variants, including highly mutated strains like Omicron. These aptamers were identified through protein-based SELEX using spike proteins from three key variants (D614G-substituted Wuhan-Hu-1, Delta, and Omicron) and virus-based SELEX, known as viro-SELEX. Leveraging these universal aptamers, we created a highly sensitive lateral flow assay (LFA) and an ultra-sensitive molecular diagnostic platform that integrates a novel rapid PCR technique, enabling fast and reliable detection across all SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Sang Won Kim
- Department of Life Sciences, POSTECH Biotech Center, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang-si 37673, Republic of Korea; (S.W.K.)
| | - Min Jung Han
- Department of Life Sciences, POSTECH Biotech Center, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang-si 37673, Republic of Korea; (S.W.K.)
| | - Md Shafiqur Rahman
- Department of Life Sciences, POSTECH Biotech Center, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang-si 37673, Republic of Korea; (S.W.K.)
| | - Heesun Kim
- Division of Integrative Bioscience & Biotechnology, POSTECH Biotech Center, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang-si 37673, Republic of Korea
| | - Jung Eun Noh
- Department of Life Sciences, POSTECH Biotech Center, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang-si 37673, Republic of Korea; (S.W.K.)
| | - Myoung Kyu Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), 141 Gajeong-ro, Yuseong, Daejeon 34114, Republic of Korea
| | - Meehyein Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), 141 Gajeong-ro, Yuseong, Daejeon 34114, Republic of Korea
| | - Jie-Oh Lee
- Department of Life Sciences, POSTECH Biotech Center, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang-si 37673, Republic of Korea; (S.W.K.)
| | - Sung Key Jang
- Department of Life Sciences, POSTECH Biotech Center, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang-si 37673, Republic of Korea; (S.W.K.)
- Division of Integrative Bioscience & Biotechnology, POSTECH Biotech Center, Pohang University of Science and Technology, 77 Cheongam-ro, Nam-gu, Pohang-si 37673, Republic of Korea
| |
Collapse
|
153
|
Rustagi V, Rameshwari R, Kumar Singh I. Identification of potential inhibitors for MAP4K4 in glaucoma using meta-dynamics-based dissociation free energy calculation. Brain Res 2025; 1847:149300. [PMID: 39500479 DOI: 10.1016/j.brainres.2024.149300] [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: 05/23/2024] [Revised: 10/16/2024] [Accepted: 10/26/2024] [Indexed: 11/12/2024]
Abstract
Glaucoma, a prevalent eye ailment causing irreversible vision loss, affects over 295 million individuals globally, necessitating the exploration of novel therapeutic avenues. Despite extensive research on targets like the phosphodiesterase enzyme and rho kinase, the potential of MAP4K4 in glaucoma remains untapped. This study aims to identify potent MAP4K4 inhibitors to counteract retinal cell apoptosis and oxidative stress associated with glaucoma. Using HTVS and XP docking, 911,059 compounds were screened. The MMGBSA calculation and pharmacokinetics analysis were used to shortlist the compounds. After performing 75 molecular dynamics simulations, further meta-dynamics were employed to calculate dissociation-free energy and find potential MAP4K4 inhibitors. Findings indicated that ZINC06717217 and ZINC38836256 exhibited remarkable promise, with docking scores of -9.57 and -11.12 and MMGBSA binding energies of -91.07 kcal/mol and -87.52 kcal/mol, respectively. Comparative analysis with the reference compound Q27453723 underscored their superior stability, requiring dissociation-free energies of -15.11 kcal/mol and -12.46 kcal/mol to disengage from the docked complex. This underscored their robust binding affinity. ZINC06717217 and ZINC38836256 show promising stability and strong binding to the MAP4K4 protein. Hence, these findings are promising in inhibiting MAP4K4 for glaucoma treatment, potentially leading to more effective treatment and curing blindness. KEY MESSAGES: First to incorporate the dissociation-free energy for identifying compounds for glaucoma treatment. In-silico analysis showed that ZINC06717217 and ZINC38836256 are promising compounds for targeting MAP4K4.
Collapse
Affiliation(s)
- Vanshika Rustagi
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana 121004, India
| | - Rashmi Rameshwari
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana 121004, India.
| | - Indrakant Kumar Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India; Delhi School of Public Health, Institute of Eminence, University of Delhi, Delhi 110007, India.
| |
Collapse
|
154
|
Gen R, Addetia A, Asarnow D, Park YJ, Quispe J, Chan MC, Brown JT, Lee J, Campbell MG, Lapointe CP, Veesler D. SARS-CoV-2 nsp1 mediates broad inhibition of translation in mammals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.633005. [PMID: 39868184 PMCID: PMC11761087 DOI: 10.1101/2025.01.14.633005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
SARS-CoV-2 nonstructural protein 1 (nsp1) promotes innate immune evasion by inhibiting host translation in human cells. However, the role of nsp1 in other host species remains elusive, especially in bats which are natural reservoirs of sarbecoviruses and possess a markedly different innate immune system than humans. Here, we reveal that SARS-CoV-2 nsp1 potently inhibits translation in bat cells from Rhinolophus lepidus, belonging to the same genus as known sarbecovirus reservoirs hosts. We determined a cryo-electron microscopy structure of SARS-CoV-2 nsp1 bound to the Rhinolophus lepidus 40S ribosome and show that it blocks the mRNA entry channel via targeting a highly conserved site among mammals. Accordingly, we found that nsp1 blocked protein translation in mammalian cell lines from several species, underscoring its broadly inhibitory activity and conserved role in numerous SARS-CoV-2 hosts. Our findings illuminate the arms race between coronaviruses and mammalian host immunity (including bats), providing a foundation for understanding the determinants of viral maintenance in bat hosts and spillovers.
Collapse
Affiliation(s)
- Risako Gen
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Amin Addetia
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Daniel Asarnow
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Young-Jun Park
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Joel Quispe
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Matthew C Chan
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jack T Brown
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Jimin Lee
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Melody G Campbell
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - David Veesler
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington; Seattle, WA 98195, USA
| |
Collapse
|
155
|
Chaddock NJM, Crossfield SSR, Pujades-Rodriguez M, Iles MM, Morgan AW. Genetic proxies for clinical traits are associated with increased risk of severe COVID-19. Sci Rep 2025; 15:2083. [PMID: 39814935 PMCID: PMC11736077 DOI: 10.1038/s41598-025-86260-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 01/09/2025] [Indexed: 01/18/2025] Open
Abstract
Routine use of genetic data in healthcare is much-discussed, yet little is known about its performance in epidemiological models including traditional risk factors. Using severe COVID-19 as an exemplar, we explore the integration of polygenic risk scores (PRS) into disease models alongside sociodemographic and clinical variables. PRS were optimized for 23 clinical variables and related traits previously-associated with severe COVID-19 in up to 450,449 UK Biobank participants, and tested in 9,560 individuals diagnosed in the pre-vaccination era. Associations were further adjusted for (i) sociodemographic and (ii) clinical variables. Pathway analyses of PRS were performed to improve biological understanding of disease. In univariate analyses, 17 PRS were associated with increased risk of severe COVID-19 and, of these, four remained associated with COVID-19 outcomes following adjustment for sociodemographic/clinical variables: hypertension PRS (OR = 1.1, 95%CI 1.03-1.18), atrial fibrillation PRS (OR = 1.12, 95%CI 1.03-1.22), peripheral vascular disease PRS (OR = 0.9, 95%CI 0.82-0.99), and Alzheimer's disease PRS (OR = 1.14, 95%CI 1.05-1.25). Pathway analyses revealed enrichment of genetic variants in pathways for cardiac muscle contraction (genes N = 5; beta[SE] = 3.48[0.60]; adjusted-P = 1.86 × 10-5). These findings underscore the potential for integrating genetic data into epidemiological models and highlight the advantages of utilizing multiple trait PRS rather than a single PRS for a specific outcome of interest.
Collapse
Affiliation(s)
- N J M Chaddock
- University of Leeds (School of Medicine and Leeds Institute for Data Analytics), Leeds, UK
| | - S S R Crossfield
- University of Leeds (School of Medicine and Leeds Institute for Data Analytics), Leeds, UK
| | - M Pujades-Rodriguez
- University of Leeds (School of Medicine and Leeds Institute for Data Analytics), Leeds, UK
| | - M M Iles
- University of Leeds (School of Medicine and Leeds Institute for Data Analytics), Leeds, UK
- Leeds Teaching Hospitals NHS Trust (NIHR Leeds Biomedical Research Centre and NIHR Leeds Medtech and In vitro Diagnostics Co-operative), Leeds, UK
| | - A W Morgan
- University of Leeds (School of Medicine and Leeds Institute for Data Analytics), Leeds, UK.
- Leeds Teaching Hospitals NHS Trust (NIHR Leeds Biomedical Research Centre and NIHR Leeds Medtech and In vitro Diagnostics Co-operative), Leeds, UK.
| |
Collapse
|
156
|
Jarras H, Blais I, Goyer B, Bazié WW, Rabezanahary H, Thériault M, Santerre K, Langlois MA, Masson JF, Pelletier JN, Brousseau N, Boudreau D, Trottier S, Baz M, Gilbert C. Impact of SARS-CoV-2 vaccination and of seasonal variations on the innate immune inflammatory response. Front Immunol 2025; 15:1513717. [PMID: 39877354 PMCID: PMC11772892 DOI: 10.3389/fimmu.2024.1513717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction The innate immune response is an important first checkpoint in the evolution of an infection. Although adaptive immunity is generally considered the immune component that retains antigenic memory, innate immune responses can also be affected by previous stimulations. This study evaluated the impact of vaccination on innate cell activation by TLR7/8 agonist R848, as well as seasonal variations. Methods To this end, blood samples from a cohort of 304 food and retail workers from the Quebec City region were collected during three visits at 12-week intervals. Peripheral blood mononuclear cells and polymorphonuclear neutrophils were isolated during the first and third visits and were stimulated with R848 to assess the innate immune response. Results Our results show that IL-8 production after stimulation decreased after vaccination. In addition, the IL-8 response was significantly different depending on the season when the visit occurred, for both COVID-19 vaccinated and unvaccinated individuals. Discussion This study highlights that innate immune responses can be affected by SARS-CoV-2 vaccination and fluctuate seasonally.
Collapse
Affiliation(s)
- Hend Jarras
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Isalie Blais
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Benjamin Goyer
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Wilfried W. Bazié
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Programme de Recherche sur les Maladies Infectieuses, Centre Muraz, Institut National de Santé Publique, Bobo-Dioulasso, Houet, Burkina Faso
| | - Henintsoa Rabezanahary
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Mathieu Thériault
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Kim Santerre
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jean-François Masson
- Department of Chemistry, Institut Courtois, Quebec Center for Advanced Materials, Regroupement québécois sur les matériaux de pointe, and Centre Interdisciplinaire de Recherche sur le Cerveau et l’Apprentissage, Université de Montréal, Montreal, QC, Canada
| | - Joelle N. Pelletier
- Department of Chemistry, Department of Biochemistry, Université de Montréal, Montreal, QC, Canada
- PROTEO — The Québec Network for Research on Protein Function, Engineering, and Applications, Quebec City, QC, Canada
| | - Nicholas Brousseau
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Biological Risks Department, Institut national de santé publique du Québec, Quebec City, QC, Canada
| | - Denis Boudreau
- Département de Chimie et Center for Optics, Photonics and Lasers (COPL), Université Laval, Quebec City, QC, Canada
| | - Sylvie Trottier
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Département de Microbiologie-Infectiologie et d’Immunologie, Faculté de Médecine, Université Laval, Quebec City, QC, Canada
| | - Mariana Baz
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Département de Microbiologie-Infectiologie et d’Immunologie, Faculté de Médecine, Université Laval, Quebec City, QC, Canada
| | - Caroline Gilbert
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Département de Microbiologie-Infectiologie et d’Immunologie, Faculté de Médecine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
157
|
Palla S, Palla SR, Liu JJ, Chao TL, Lee TH, Kavala V, Liu IC, Wang LHC, Chang SY, Yao CF, Liang PH. Green Synthesis of Tetrahydropyrazino[2,1-a:5,4-a']diisoquinolines as SARS-CoV-2 Entry Inhibitors. ACS OMEGA 2025; 10:1164-1176. [PMID: 39829498 PMCID: PMC11740144 DOI: 10.1021/acsomega.4c08640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025]
Abstract
A class of tetrahydropyrazino[2,1-a:5,4-a']diisoquinoline derivatives were synthesized under environmentally friendly conditions using water as the solvent. The 3-D structures of some synthesized compounds were determined by X-ray diffraction. Since naturally occurring isoquinoline alkaloids have significant antiviral activities against a wide range of viruses, including coronaviruses, the synthesized compounds were assayed for their inhibitory activities against SARS-CoV-2. Our results showed that the active compounds 50 and 96 blocked the delta SARS-CoV-2 entry into VeroE6 cells to display EC50 of 26.5 ± 6.9 and 17.0 ± 3.7 μM, respectively, by inhibiting the interaction between SARS-CoV-2 Spike's receptor binding domain (RBD) and human receptor angiotensin-converting enzyme 2 (ACE2), and CC50 greater than 100 μM. This study provides a green synthesis method of tetrahydropyrazinodiisoquinoline for antiviral or other applications.
Collapse
Affiliation(s)
- Sowndarya Palla
- Department
of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Srinivasa Rao Palla
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
- Taiwan
International Graduate Program, Academia
Sinica, Taipei 11529, Taiwan
| | - Jia-Jin Liu
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
| | - Tai-Ling Chao
- Department
of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei 10048, Taiwan
| | - Ting-Hui Lee
- Department
of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Veerababurao Kavala
- Department
of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - I-Chen Liu
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
| | - Lily Hui-Ching Wang
- Institute
of Molecular and Cellular Biology, National
Tsing Hua University, Hsinchu 30013, Taiwan
| | - Sui-Yuan Chang
- Department
of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei 10048, Taiwan
- Department
of Laboratory Medicine, National Taiwan
University Hospital, Taipei 10002, Taiwan
| | - Ching-Fa Yao
- Department
of Chemistry, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Po-Huang Liang
- Institute
of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
- Institute
of Biochemical Sciences, National Taiwan
University, Taipei 10617, Taiwan
- Taiwan
International Graduate Program, Academia
Sinica, Taipei 11529, Taiwan
| |
Collapse
|
158
|
Al-Momani H, Nelson A, Al Balawi H, Al Balawi D, Aolymat I, Khasawneh AI, Tabl H, Alsheikh A, Zueter AM, Pearson J, Ward C. Assessment of upper respiratory and gut bacterial microbiomes during COVID-19 infection in adults: potential aerodigestive transmission. Sci Rep 2025; 15:1811. [PMID: 39805887 PMCID: PMC11730684 DOI: 10.1038/s41598-025-85806-5] [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: 07/16/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
SARS-CoV-2 is the viral pathogen responsible for COVID-19. Although morbidity and mortality frequently occur as a result of lung disease, the gastrointestinal (GI) tract is recognized as a primary location for SARS-CoV-2. Connections and interactions between the microbiome of the gut and respiratory system have been linked with viral infections via what has been referred to as the 'gut-lung axis' with potential aerodigestive communication in health and disease. This research explored the relationship between the microbiomes of the upper respiratory and GI tracts in patients with COVID-19 and examined Extraesophageal reflux (EOR), a mechanism which could contribute to dysregulated communication between the GI and respiratory tract (as identified in COVID-19). 97 patients with a laboratory diagnosis of COVID-19 infection, and 50 age-matched controls were recruited and stool, saliva and sputum were obtained from each participant. ELISA Pepsin tests and Reflux Symptom Index scores (RSI) were conducted for EOR assessment. DNA sequencing of the V4 region of the 16 S rRNA gene was performed for microbiome analysis. No differences were observed between the fecal microbiome's alpha and Shannon diversity indices; however, a distinct microbial composition was observed in COVID-19 patients (when compared to the controls). The respiratory microbiota from individuals with COVID-19 demonstrated a statistically significant reduction in Shannon diversity and bacterial richness alongside an overall reduction in the prevalence of organisms from a typical healthy respiratory microbiome. Furthermore, the bacterial richness of the stool and sputum samples was significantly lower among COVID-19 patients admitted to ICU. A significantly higher RSI score and salivary pepsin level were detected among those with COVID-19. The data indicates that COVID-19 is associated with a dysregulation of both the gut and lung microbiome with a more marked perturbation in the lung, particularly among COVID-19 patients who had been admitted to the ICU. The presence of increased RSI scores, combined with elevated levels of Pepsin, suggests that increased micro-aspiration may occur, which is consistent with of under-recognized interactions between the GI and lung microbiomes in COVID-19 patients and requires additional study. Such studies would benefit from the insights provided by biological samples which reflect the continuum of the aerodigestive tract.
Collapse
Affiliation(s)
- Hafez Al-Momani
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, P.O box 330127, Zarqa, 13133, Jordan.
| | - Andrew Nelson
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Northumberland Building, Newcastle Upon Tyne, NE1 8ST, United Kingdom
| | - Hadeel Al Balawi
- Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Dua'a Al Balawi
- Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, 13133, Jordan
| | - Iman Aolymat
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, P.O box 330127, Zarqa, 13133, Jordan
| | - Ashraf I Khasawneh
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, P.O box 330127, Zarqa, 13133, Jordan
| | - Hala Tabl
- Department of Microbiology, Pathology and Forensic Medicine, Faculty of Medicine, The Hashemite University, P.O box 330127, Zarqa, 13133, Jordan
| | - Ayman Alsheikh
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Zarqa University, P.O.box 2000, Zarqa, 13110, Jordan
| | - AbdelRahman M Zueter
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O box 330127, Zarqa, 13133, Jordan
| | - Jeffrey Pearson
- Biosciences Institute, Newcastle University Medical School, Newcastle upon Tyne, NE2 4HH, GB, United Kingdom
| | - Christopher Ward
- Translational and Clinical Research Institute, Newcastle University Medical School, Newcastle upon Tyne, NE2 4HH, GB, United Kingdom
| |
Collapse
|
159
|
Yskak A, Sokharev Y, Zhumalynov K, Koneva E, Afanasyeva N, Borodulin D, Babaskin D, Nugmanov A, Nurushev M, Chashkov V. Hormonal Implications of SARS-CoV-2: A Review of Endocrine Disruptions. SCIENTIFICA 2025; 2025:7305185. [PMID: 39830837 PMCID: PMC11742418 DOI: 10.1155/sci5/7305185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/27/2024] [Accepted: 11/22/2024] [Indexed: 01/22/2025]
Abstract
To improve medical care and rehabilitation algorithms for patients affected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), it is important to evaluate and summarize the available data on the effect of coronavirus infection (COVID-19) on the endocrine system. The purpose of this review was to study the effect of COVID-19 on the endocrine system. The scientific novelty of this study is the evaluation of the effect of coronavirus infection on the endocrine system and the potential effect of hormones on susceptibility to COVID-19. The results of this review show that the endocrine system is vulnerable to disorders caused by COVID-19, mainly thyroid dysfunction and hyperglycemia. The information in the published literature mentioned here contains some unclear aspects and contradictory data, but much remains to be studied and clarified regarding the impact of COVID-19 on the endocrine system. In particular, this concerns the study of the hyperglycemic status of patients who have had coronavirus infection, which is extremely important for the future metabolic health of COVID-19 survivors. This review contributes to the scientific discourse by systematically synthesizing disparate studies to identify patterns, gaps, and emerging trends in the literature concerning the effects of COVID-19 on the endocrine system. By integrating these findings, this study offers a novel perspective on potential hormonal interactions influencing COVID-19 susceptibility and outcomes, proposing new hypotheses and frameworks for future research.
Collapse
Affiliation(s)
- Aliya Yskak
- Research Institute of Applied Biotechnology, Akhmet Baitursynuly Kostanay Regional University, Kostanay, Kazakhstan
- Faculty of Soil Science, Lomonosov Moscow State University, Moscow, Russia
| | - Yevgeniy Sokharev
- Pathological Anatomy Department, Municipal State Company “Kostanay Regional Pathoanatomical Bureau” of the Health Department of the Akimat of the Kostanay Region, Kostanay, Kazakhstan
| | - Kuanysh Zhumalynov
- Department of Natural Sciences, Akhmet Baitursynuly Kostanay Regional University, Kostanay, Kazakhstan
| | - Elizaveta Koneva
- Department of Sports Medicine and Medical Rehabilitation, Sechenov University, Moscow, Russia
| | - Natalia Afanasyeva
- Resource Center “Medical Sechenov Pre-University”, Sechenov University, Moscow, Russia
| | - Dmitri Borodulin
- Department of Technology of Storage and Processing of Fruits, Vegetables and Plant Growing Products, Russian State Agrarian University-Moscow Timiryazev Agricultural Academy, Moscow, Russia
| | | | - Almabek Nugmanov
- Department of Natural Sciences, Akhmet Baitursynuly Kostanay Regional University, Kostanay, Kazakhstan
| | - Murat Nurushev
- Higher School of Natural Sciences, Astana International University, Astana, Kazakhstan
| | - Vadim Chashkov
- Department of Natural Sciences, Akhmet Baitursynuly Kostanay Regional University, Kostanay, Kazakhstan
| |
Collapse
|
160
|
Khosravi S, IzadPanah M, Mirzaei S, Rahmani H, Oroomiei N. Challenges faced by the Iranian health system in containing COVID-19: insights from a conventional content analysis. BMC Health Serv Res 2025; 25:60. [PMID: 39799331 PMCID: PMC11725184 DOI: 10.1186/s12913-025-12223-9] [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: 09/22/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND The COVID-19 pandemic is a global crisis, and health systems worldwide have faced numerous challenges in containing it. This study aimed to identify the challenges faced by the Iranian health system in controlling the COVID-19 pandemic. METHODS A conventional content analysis approach was employed in this qualitative study. The research sample included experts responsible for managing, making decisions about preventing COVID-19, and caring for and treating patients. The purposive sampling technique was used to select the participants. Data saturation was achieved after 29 interviews. Additionally, conventional content analysis was applied for data analysis. RESULTS The participants in this study identified the most significant challenges faced by Iran's health system in controlling the COVID-19 pandemic, including issues related to human resources, physical and organizational structures, financial resources, inter-sectoral collaboration, personal protective equipment (PPE), social responsibility, and information services. CONCLUSIONS Identifying these challenges can help health system managers prioritize the most critical issues. Addressing these challenges requires more than just the efforts of health systems; cooperation at the macro governance level is essential. Governments should start planning to formulate appropriate strategies.
Collapse
Grants
- 40100024 Bam University of Medical Sciences, Bam, Iran
- 40100024 Bam University of Medical Sciences, Bam, Iran
- 40100024 Bam University of Medical Sciences, Bam, Iran
- 40100024 Bam University of Medical Sciences, Bam, Iran
- 40100024 Bam University of Medical Sciences, Bam, Iran
Collapse
Affiliation(s)
- Sajad Khosravi
- Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Mahboobeh IzadPanah
- Student Research Committee, School of Public Health, Bam University of Medical Sciences, Bam, Iran
| | - Saeid Mirzaei
- Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Hosein Rahmani
- Master of sciences, Kerman branch, Islamic Azad University, Kerman, Iran
| | - Nadia Oroomiei
- Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran.
- Bam University of Medical Sciences, Sardaran Shahid Square- Shahid Rajaei Boulevard, Bam, 7616913555, Iran.
| |
Collapse
|
161
|
HAMDAN M, KULABAŞ N, KÜÇÜKGÜZEL İ. In silico Evaluation of H1-Antihistamine as Potential Inhibitors of SARS-CoV-2 RNA-dependent RNA Polymerase: Repurposing Study of COVID-19 Therapy. Turk J Pharm Sci 2025; 21:566-576. [PMID: 39801109 PMCID: PMC11730003 DOI: 10.4274/tjps.galenos.2024.49768] [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: 05/31/2024] [Accepted: 06/23/2024] [Indexed: 01/16/2025]
Abstract
Introduction Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), from the family Coronaviridae, is the seventh known coronavirus to infect humans and cause acute respiratory syndrome. Although vaccination efforts have been conducted against this virus, which emerged in Wuhan, China, in December 2019 and has spread rapidly around the world, the lack of an Food and Drug Administration-approved antiviral agent has made drug repurposing an important approach for emergency response during the COVID-19 pandemic. The aim of this study was to investigate the potential of H1-antihistamines as antiviral agents against SARS-CoV-2 RNA-dependent RNA polymerase enzyme. Materials and Methods Using molecular docking techniques, we explored the interactions between H1-antihistamines and RNA-dependent RNA polymerase (RdRp), a key enzyme involved in viral replication. The three-dimensional structure of 37 H1-antihistamine molecules was drawn and their energies were minimized using Spartan 0.4. Subsequently, we conducted a docking study with Autodock Vina to assess the binding affinity of these molecules to the target site. The docking scores and conformations were then visualized using Discovery Studio. Results The results examined showed that the docking scores of the H1-antihistamines were between 5.0 and 8.3 kcal/mol. These findings suggested that among all the analyzed drugs, bilastine, fexofenadine, montelukast, zafirlukast, mizolastine, and rupatadine might bind with the best binding energy (< -7.0 kcal/mol) and inhibit RdRp, potentially halting the replication of the virus. Conclusion This study highlights the potential of H1-antihistamines in combating COVID-19 and underscores the value of computational approaches in rapid drug discovery and repurposing efforts. Finally, experimental studies are required to measure the potency of H1-antihistamines before their clinical use against COVID-19 as RdRp inhibitors.
Collapse
Affiliation(s)
- Mazin HAMDAN
- Marmara University Institute of Health Sciences, Department of Pharmaceutical Chemistry, İstanbul, Türkiye
| | - Necla KULABAŞ
- Marmara University Faculty of Pharmacy, Department of Pharmaceutical Chemistry, İstanbul, Türkiye
| | - İlkay KÜÇÜKGÜZEL
- Fenerbahçe University Faculty of Pharmacy, Department of Pharmaceutical Chemistry, İstanbul, Türkiye
| |
Collapse
|
162
|
Licata A, Seidita A, Como S, de Carlo G, Cammilleri M, Bonica R, Soresi M, Veronese N, Chianetta R, Citarrella R, Giannitrapani L, Barbagallo M. Herbal and Dietary Supplements as Adjunctive Treatment for Mild SARS-CoV-2 Infection in Italy. Nutrients 2025; 17:230. [PMID: 39861359 PMCID: PMC11767322 DOI: 10.3390/nu17020230] [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: 11/25/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
During the COVID-19 pandemic, several observational studies proved a certain efficacy of nutraceuticals, herbal products, and other dietary supplements as adjuvant therapies used alongside antiviral drugs. Although their use has not been widespread in Italy, according to preliminary evidence, many supplements with demonstrated immunomodulatory effects, such as vitamins C and D, herbal medicines and essential oils, might relieve the respiratory symptoms of COVID-19, since SARS-CoV-2 can activate inflammasome-mediated inflammatory signaling pathways. Other observational studies have shown that herbal treatments, such as Echinacea purpurea and ginseng, help alleviate respiratory symptoms and reduce serum levels of inflammatory cytokines, which are typically overexpressed in both adult and pediatric SARS-CoV-2 patients. Further, vitamins C and D can attenuate the immune response thanks to their cytokine suppression ability and to their known antimicrobial activity and potential to modulate T helper cell response. The strong immune response triggered by SARS-CoV-2 infection is responsible for the severity of the disease. Preliminary data have also shown that L-arginine, an endothelial-derived relaxing factor, is able to modulate endothelial damage, which appears to be one of the main targets of this systemic disease. Finally, some essential oils and their isolated compounds, such as eucalyptol, may be helpful in reducing many of the respiratory symptoms of COVID-19, although others, such as menthol, are not recommended, since it can lead to an undervaluation of the clinical status of a patient. In this narrative review, despite the lack of strong evidence in this field, we aimed to give an overview of the current available literature (mainly observational and cross-sectional studies) regarding herbal products and dietary supplements and their use in the treatment of mild disease from SARS-CoV-2 infection. Obviously, dietary supplements and herbal products do not constitute a standardized treatment for COVID-19 disease, but they could represent an adjunctive and useful treatment when used together with antivirals.
Collapse
Affiliation(s)
- Anna Licata
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Aurelio Seidita
- Unit of Internal Medicine, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90146 Palermo, Italy
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Silvia Como
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Gabriele de Carlo
- Unit of Internal Medicine, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, 90146 Palermo, Italy; (A.S.)
| | - Marcella Cammilleri
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Roberta Bonica
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Maurizio Soresi
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Nicola Veronese
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Roberta Chianetta
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Roberto Citarrella
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Lydia Giannitrapani
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Mario Barbagallo
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| |
Collapse
|
163
|
Liu J, Luo S, Xu X, Zhang E, Liang H, Zhang JZH, Duan L. Evaluating the Synergistic Effects of Multi-Epitope Nanobodies on BA.2.86 Variant Immune Escape. J Phys Chem Lett 2025; 16:396-405. [PMID: 39780712 DOI: 10.1021/acs.jpclett.4c03028] [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: 01/11/2025]
Abstract
Addressing the frequent emergence of SARS-CoV-2 mutant strains requires therapeutic approaches with innovative neutralization mechanisms. The targeting of multivalent nanobodies can enhance potency and reduce the risk of viral escape, positioning them as promising drug candidates. Here, the synergistic mechanisms of the two types of nanobodies are investigated deeply. Our research revealed that the Fu2-1-Fu2-2 system exhibited significant synergy, whereas the Sb#15-Sb#68 system demonstrated antagonism, in which entropy was the dominant contributor to antagonism. Conformational analysis further demonstrated that the presence of a monomeric nanobody influenced the flexibility of residues near other epitopes, thereby affecting the overall synergy of the systems. Moreover, we identified that changes in the hydrogen bond network and the charge of residues played a critical role in the binding between nanobodies and spike. We hope this study will provide novel insights into the development of multivalent nanobody combinations.
Collapse
Affiliation(s)
- Jinxin Liu
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Song Luo
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Xiaole Xu
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Enhao Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - Houde Liang
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| | - John Z H Zhang
- Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen 518055, China
- NYU-ECNU Center for Computational Chemistry and Shanghai Frontiers Science Center of AI and DL, NYU Shanghai, Shanghai 200124, China
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Lili Duan
- School of Physics and Electronics, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
164
|
Le NP, Le BT, Le VP, Park JE. Molecular characterization of swine acute diarrhea syndrome coronavirus detected in Vietnamese pigs. Vet Res 2025; 56:4. [PMID: 39789654 PMCID: PMC11720870 DOI: 10.1186/s13567-024-01445-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/25/2024] [Indexed: 01/12/2025] Open
Abstract
Swine acute diarrhea syndrome coronavirus (SADS-CoV) is a novel porcine coronavirus that was recently identified in southern China in 2017. At present, there is a lack of nationwide epidemiological investigations of the newly emerged SADS-CoV. Because Vietnam is geographically adjacent to southern China, many diseases have spread from China to Vietnam. To assess SADS-CoV transmission to Vietnam, we conducted a retrospective study to detect SADS-CoV in samples collected from pig farms in northern Vietnam. Among the 69 fecal samples tested, 5 were positive for SADS-CoV. The spike gene sequence showed high genetic homology with strains circulating in China. Our study is the first to show that SADS-CoV has spread from China to Vietnam and highlights the need for global epidemiological investigations of SADS-CoV.
Collapse
Affiliation(s)
- Nam Phuong Le
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Bac Tran Le
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Van Phan Le
- College of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi, Vietnam.
| | - Jung-Eun Park
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|
165
|
Bian H, Zhu S, Xing W, Qi L, Xue J, Peng X, Jin Z, Zhao H. Research Status and Direction of Chronic Obstructive Pulmonary Disease Complicated with Coronary Heart Disease: A Bibliometric Analysis from 2005 to 2024. Int J Chron Obstruct Pulmon Dis 2025; 20:23-41. [PMID: 39802036 PMCID: PMC11724669 DOI: 10.2147/copd.s495326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025] Open
Abstract
Objective There is increasing evidence that chronic obstructive pulmonary disease (COPD) is associated with coronary heart disease (CHD). In this study, we provide valuable insights in the field by examining the evolution of the relationship between COPD and CHD over the past 20 years. Methods A comprehensive computer search was conducted in the Web of Science (WOS) core dataset, covering literature on COPD combined with CHD from January 1, 2005, to August 20, 2024. Visual analyses were performed using VOSviewer, CiteSpace, and Bibliometrix to assess countries, institutions, the centrality of institutional intermediaries, authorship patterns, including co-cited authors and references, and keywords; Excel (version 2021) software was utilized for generating relevant descriptive analysis tables. Results A total of 2420 publications sourced from WOS were included in this study. Since 2005, there has been a continuous increase in the literature about COPD combined with CHD; polynomial fitting yielded an R² value of 0.7758. The volume of literature in this domain is projected to continue growing steadily. The United States emerged as the leading country by publication count; Lin Cheng-li ranked first among authors, while China Medical University topped institutional contributions. Notably, Sin dd, Mannino dm, and Helvaci Mr were identified as the top three authors based on citation frequency. The Journal of Vascular Surgery recorded the highest number of publications, whereas The Lancet was recognized as the most influential among the top ten co-cited journals. The most frequently cited reference pertains to systemic inflammation's role in increasing cardiovascular risk among patients with COPD. Through keyword clustering analysis, we categorized all keywords into three distinct groups: management strategies for COPD and CHD; diseases associated with both conditions; and epidemiological characteristics concerning their burden-current hotspots include multimorbidity factors such as hypertension and obesity alongside outcomes like diagnosis during COVID-19 pandemic implications within societal contexts are highlighted here too. Conclusion Presently focused research on COPD coupled with CHD primarily revolves around five key areas: pathogenesis exploration, early diagnostic techniques, COVID-19 infection, dynamics intervention, methodologies, and treatment protocol development efforts. To improve the early detection rate of COPD complicated with CHD, the main development direction in the future is to extract computed tomography (CT) features using imaging omics and establish an early prediction model. The results of this study will provide new ideas and directions for subsequent related research.
Collapse
Affiliation(s)
- Hupo Bian
- Department of Radiology, The First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, People’s Republic of China
| | - Shaoqi Zhu
- Department of Endocrinology, The First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, People’s Republic of China
| | - Wenjian Xing
- Department of Radiology, The Linghu People’s Hospital, Huzhou, Zhejiang, People’s Republic of China
| | - Luying Qi
- Department of Radiology, The First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, People’s Republic of China
| | - Jingnan Xue
- Department of Radiology, The First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, People’s Republic of China
| | - Xiuhua Peng
- Department of Radiology, The First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, People’s Republic of China
| | - Zanhui Jin
- Department of Radiology, The First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, People’s Republic of China
| | - Hongxing Zhao
- Department of Radiology, The First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, People’s Republic of China
- Huzhou Key Laboratory of Precise Diagnosis and Treatment of Urinary Tumors, Huzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
166
|
Lee E, Rauscher S. The Conformational Space of the SARS-CoV-2 Main Protease Active Site Loops Is Determined by Ligand Binding and Interprotomer Allostery. Biochemistry 2025; 64:32-46. [PMID: 39513739 DOI: 10.1021/acs.biochem.4c00575] [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: 11/15/2024]
Abstract
The main protease (Mpro) of SARS-CoV-2 is essential for viral replication and is, therefore, an important drug target. Here, we investigate two flexible loops in Mpro that play a role in catalysis. Using all-atom molecular dynamics simulations, we analyze the structural ensemble of Mpro in an apo state and substrate-bound state. We find that the flexible loops can adopt open, intermediate (partly open), and closed conformations in solution, which differs from the partially closed state observed in crystal structures of Mpro. When the loops are in closed or intermediate states, the catalytic residues are more likely to be in close proximity, which is crucial for catalysis. Additionally, we find that substrate binding to one protomer of the homodimer increases the frequency of intermediate states in the bound protomer while also affecting the structural propensity of the apo protomer's flexible loops. Using dynamic network analysis, we identify multiple allosteric pathways connecting the two active sites of the homodimer. Common to these pathways is an allosteric hotspot involving the N-terminus, a critical region that comprises part of the binding pocket. Taken together, the results of our simulation study provide detailed insight into the relationships between the flexible loops and substrate binding in a prime drug target for COVID-19.
Collapse
Affiliation(s)
- Ethan Lee
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H8, Canada
| | - Sarah Rauscher
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H8, Canada
- Department of Physics, University of Toronto, Toronto, ON M5S 1A7, Canada
| |
Collapse
|
167
|
Wang D, Li H, Liu Y, Li H, Liu Y, Hou L. The impact of inflammatory response on psychological status of medical staff during COVID-19 pandemic. PSICOLOGIA-REFLEXAO E CRITICA 2025; 38:3. [PMID: 39757257 DOI: 10.1186/s41155-024-00335-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Limited research has been conducted on the relationship between inflammatory markers and psychological status in medical staff fighting COVID-19. OBJECTIVE This article examines the psychological and inflammatory conditions of medical personnel working on the front lines of the battle against COVID-19. METHODS A total of 102 clinical staff members were included in this study. All subjects received the Symptom Checklist-90 questionnaire (SCL-90) and Posttraumatic Stress Disorder Checklist-Civilian questionnaires for assessing different mental symptoms. The levels of various inflammatory markers, including IL-1β, IL-2, IL-6, IL-8, TNF-a, and IFN-γ, along with GDNF, were evaluated. RESULTS Spearman correlation analysis showed that the levels of IL-6 were positively associated with the anxiety score (Spearman's rho = .230, p = .021), obsessive-compulsive symptoms (Spearman's rho = .201, p = .042). The levels of IL-8 were negatively associated with the anxiety score (Spearman's rho = -.223, p = .028), obsessive-compulsive symptoms (Spearman's rho = -.252, p = .012), hyperarousal (Spearman's rho = -.221, p = .028). The levels of TNF-α were positively associated with the anxiety score (Spearman's rho = .201, p = .045), obsessive-compulsive symptoms (Spearman's rho = .222, p = .035). CONCLUSION Generally, our results suggested that IL-6, IL-8 and TNF-α might play a role in the development of psychological symptoms among medical staff.
Collapse
Affiliation(s)
- Dong Wang
- Department of Geriatric Psychiatry, Suzhou Mental Health Center, Suzhou Guangji Hospital, the Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Haijin Li
- Department of Psychiatry, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yansong Liu
- Department of Geriatric Psychiatry, Suzhou Mental Health Center, Suzhou Guangji Hospital, the Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Hong Li
- Department of Psychiatry, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yangyang Liu
- First People's Hospital of Guannan County, Lianyungang, China
| | - Lijun Hou
- The Affiliated Infectious Hospital of Soochow University, 10 Guangqian Road, Suzhou, Jiangsu, 215131, China.
- The Fifth People's Hospital of Suzhou, Suzhou, China.
| |
Collapse
|
168
|
Zeng J, Zhang X, Huang C, Tian S, Zhao H. Dampened TLR2-mediated Inflammatory Signaling in Bats. Mol Biol Evol 2025; 42:msae253. [PMID: 39663845 PMCID: PMC11702297 DOI: 10.1093/molbev/msae253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024] Open
Abstract
Bats are considered natural hosts for numerous viruses. Their ability to carry viruses that cause severe diseases or even death in other mammals without falling ill themselves has attracted widespread research attention. Toll-like receptor 2 forms heterodimers with Toll-like receptor 1 or Toll-like receptor 6 on cell membranes, recognizing specific pathogen-associated molecular patterns and playing a key role in innate immune responses. Previous studies have shown that moderate Toll-like receptor 2-mediated immune signals aid in pathogen clearance, while excessive or inappropriate Toll-like receptor 2-mediated immune signals can cause self-damage. In this study, we observed that TLR2, unlike TLR1 or TLR6, has undergone relaxed selection in bats compared with other mammals, indicating a reduced functional constraint on TLR2 specifically in bats. Indeed, our cell-based functional assays demonstrated that the ability of Toll-like receptor 2 to bind with Toll-like receptor 1 or Toll-like receptor 6 was significantly reduced in bats, leading to dampened inflammatory signaling. We identified mutations unique to bats that were responsible for this observation. Additionally, we found that mutations at residues 375 and 376 of Toll-like receptor 2 in the common ancestor of bats also resulted in reduced inflammatory response, suggesting that this reduction occurred early in bat evolution. Together, our study reveals that the Toll-like receptor 2-mediated inflammatory response has been specifically dampened in bats, which may be one of the reasons why they could harbor many viruses without falling ill.
Collapse
Affiliation(s)
- Jiaming Zeng
- Key Laboratory of Biodiversity and Environment on the Qinghai–Tibetan Plateau, Ministry of Education, State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan 430072 Hubei, China
| | - Xiangyi Zhang
- Key Laboratory of Biodiversity and Environment on the Qinghai–Tibetan Plateau, Ministry of Education, State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan 430072 Hubei, China
| | - Chen Huang
- Key Laboratory of Biodiversity and Environment on the Qinghai–Tibetan Plateau, Ministry of Education, State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan 430072 Hubei, China
| | - Shilin Tian
- Key Laboratory of Biodiversity and Environment on the Qinghai–Tibetan Plateau, Ministry of Education, State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan 430072 Hubei, China
| | - Huabin Zhao
- Key Laboratory of Biodiversity and Environment on the Qinghai–Tibetan Plateau, Ministry of Education, State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan 430072 Hubei, China
| |
Collapse
|
169
|
Hills FR, Geoghegan JL, Bostina M. Architects of infection: A structural overview of SARS-related coronavirus spike glycoproteins. Virology 2025; 604:110383. [PMID: 39983449 DOI: 10.1016/j.virol.2024.110383] [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: 10/20/2024] [Revised: 12/22/2024] [Accepted: 12/29/2024] [Indexed: 02/23/2025]
Abstract
The frequency of zoonotic viral emergence within the Coronaviridae family highlights the critical need to understand the structural features of spike proteins that govern viral entry and host adaptation. Investigating the structural conservation and variation in key regions of the spike protein-those involved in host range, binding affinity, viral entry, and immune evasion-is essential for predicting the evolutionary pathways of coronaviruses, assessing the risk of future host-jumping events, and discovering pan-neutralising antibodies. Here we summarise our current structural understanding of the spike proteins similar to SARS-CoV-2 from the Coronaviridae family and compare key functional similarities and differences. Our aim is to demonstrate the significant structural and sequence conservation between spike proteins from a range of host species and to outline the importance of animal coronavirus surveillance and structural investigation in our endeavour for pandemic preparedness against emerging viruses.
Collapse
Affiliation(s)
- Francesca R Hills
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Jemma L Geoghegan
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Mihnea Bostina
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
170
|
Nguyen QT, Mai TT, Tuong LT, Nguyen TTN, Vo TP, Nguyen DN, Phan-Van CT, Trinh DTT, Tran VT, Thai KM. Binding ability of Delta and Omicron towards the angiotensin-converting enzyme 2 receptor and antibodies: a computational study. J Biomol Struct Dyn 2025:1-13. [PMID: 39755961 DOI: 10.1080/07391102.2024.2446659] [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: 11/24/2023] [Accepted: 08/31/2024] [Indexed: 01/07/2025]
Abstract
The COVID-19 pandemic posed a threat to global society. Delta and Omicron are concerning variants due to the risk of increasing human-to-human transmissibility and immune evasion. This study aims to evaluate the binding ability of these variants toward the angiotensin-converting enzyme 2 receptor and antibodies using a computational approach. The receptor-binding domain (RBD) of the two variants was created by CHARMM-GUI and then docked to the hACE2 receptor and two antibodies (REGN10933 and REGN10987). These complexes were also subjected to molecular dynamics simulation within 100 ns. As a result, the two variants, Omicron and Delta, exhibited stronger interaction with the hACE2 receptor than the wild type. The mutations in the RBD region also facilitated the virus's escape from antibody neutralization.
Collapse
Affiliation(s)
- Quoc-Thai Nguyen
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Tan Thanh Mai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Lam-Truong Tuong
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Thi-Thao-Nhung Nguyen
- University of Health Sciences, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Thanh-Phuong Vo
- University of Health Sciences, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Dac-Nhan Nguyen
- University of Health Sciences, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Cong-Thanh Phan-Van
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | | | - Van-Thanh Tran
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Khac-Minh Thai
- University of Health Sciences, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam
| |
Collapse
|
171
|
Yegnaswamy S, C SK, Aldaais E. Conformational dynamics of the membrane protein of MERS-CoV in comparison with SARS-CoV-2 in ERGIC complex. J Biomol Struct Dyn 2025:1-15. [PMID: 39755960 DOI: 10.1080/07391102.2024.2437529] [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: 01/28/2024] [Accepted: 05/21/2024] [Indexed: 01/07/2025]
Abstract
The present study explores the conformational dynamics of the membrane protein of Middle East Respiratory Syndrome Coronavirus (MERS-CoV) within the Endoplasmic Reticulum-Golgi Intermediate Compartment (ERGIC) complex using an all-atomistic molecular dynamics simulation approach. Significant structural changes were observed in the N-terminal, C-terminal, transmembrane, and beta-sheet sandwich domains of the MERS-CoV membrane protein. This study also highlights the structural similarities between the MERS-CoV and the SARS-CoV-2 membrane proteins, particularly in how both exhibit a distinct kink in the transmembrane helix caused by aromatic residue-lipid interactions. A structural expansion below the transmembrane and above the beta-sheet sandwich domain within the dimer was observed in all the M-proteins. This site on the beta-sheet sandwich domains near the C-terminal end could serve as a potential drug-binding site. Notably, a stable helical structure was identified in the C-terminal domain of the MERS-CoV membrane protein, whereas a proper secondary structural conformation was not observed in the SARS-CoV-2 membrane protein. Further, the SARS-CoV-2 membrane protein exhibited stronger binding to the lipid bilayer than the MERS-CoV, indicating its greater structural stability within the ERGIC complex. The structural similarity between the membrane protein of MERS-CoV and SARS-CoV-2 suggests the feasibility of employing a common inhibitor against these beta-coronaviruses. Furthermore, this analysis enhances our understanding of the membrane protein's interactions with proteins and lipids, paving the way for therapeutic developments against these viruses.
Collapse
Affiliation(s)
- Subha Yegnaswamy
- School of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, Navi Mumbai, Maharashtra, India
| | - Selvaa Kumar C
- School of Biotechnology and Bioinformatics, D.Y. Patil Deemed to be University, Navi Mumbai, Maharashtra, India
| | - Ebtisam Aldaais
- College of Applied Medical Sciences, lmam Abdulrahman Bin Faisal University (lAU), Dammam, Saudi Arabia
| |
Collapse
|
172
|
de Souza UJB, Spilki FR, Tanuri A, Roehe PM, Campos FS. Two Years of SARS-CoV-2 Omicron Genomic Evolution in Brazil (2022-2024): Subvariant Tracking and Assessment of Regional Sequencing Efforts. Viruses 2025; 17:64. [PMID: 39861853 PMCID: PMC11768930 DOI: 10.3390/v17010064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
SARS-CoV-2, the virus responsible for COVID-19, has undergone significant genetic evolution since its emergence in 2019. This study examines the genomic diversity of SARS-CoV-2 in Brazil after the worst phase of the pandemic, the wider adoption of routine vaccination, and the abolishment of other non-pharmacological preventive measures from July 2022 to July 2024 using 55,951 sequences retrieved from the GISAID database. The analysis focuses on the correlation between confirmed COVID-19 cases, sequencing efforts across Brazilian states, and the distribution and evolution of viral lineages. Our findings reveal significant regional disparities in genomic surveillance, with São Paulo and Rio de Janeiro recovering the largest number of genomes, while Tocantins and Amazonas showed higher sequencing rates relative to their reported case numbers, indicating proactive surveillance efforts. We identified 626 distinct SARS-CoV-2 lineages circulating in Brazil, with dominant subvariants shifting over time from BA.5 in 2022 to XBB and JN.1 in 2023-2024. The emergence of new subvariants in this new epidemiological scenario underscores the importance of ongoing genomic surveillance to track viral evolution and inform public health strategies, providing valuable information to update vaccines and implement other measures, such as lockdowns, mask usage, social distancing, health education, and self-testing.
Collapse
Affiliation(s)
- Ueric José Borges de Souza
- Bioinformatics and Biotechnology Laboratory, Campus of Gurupi, Federal University of Tocantins, Gurupi 77410-570, Brazil
| | | | - Amilcar Tanuri
- Laboratory of Genetics and Immunology of Viral Infections, Department of Virology, Paulo de Góes Institute of Microbiology, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Paulo Michel Roehe
- Virology Laboratory, Department of Microbiology, Immunology, and Parasitology, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90050-170, Brazil;
| | - Fabrício Souza Campos
- Bioinformatics and Biotechnology Laboratory, Campus of Gurupi, Federal University of Tocantins, Gurupi 77410-570, Brazil
- Virology Laboratory, Department of Microbiology, Immunology, and Parasitology, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90050-170, Brazil;
| |
Collapse
|
173
|
Gawande MS, Zade N, Kumar P, Gundewar S, Weerarathna IN, Verma P. The role of artificial intelligence in pandemic responses: from epidemiological modeling to vaccine development. MOLECULAR BIOMEDICINE 2025; 6:1. [PMID: 39747786 PMCID: PMC11695538 DOI: 10.1186/s43556-024-00238-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
Integrating Artificial Intelligence (AI) across numerous disciplines has transformed the worldwide landscape of pandemic response. This review investigates the multidimensional role of AI in the pandemic, which arises as a global health crisis, and its role in preparedness and responses, ranging from enhanced epidemiological modelling to the acceleration of vaccine development. The confluence of AI technologies has guided us in a new era of data-driven decision-making, revolutionizing our ability to anticipate, mitigate, and treat infectious illnesses. The review begins by discussing the impact of a pandemic on emerging countries worldwide, elaborating on the critical significance of AI in epidemiological modelling, bringing data-driven decision-making, and enabling forecasting, mitigation and response to the pandemic. In epidemiology, AI-driven epidemiological models like SIR (Susceptible-Infectious-Recovered) and SIS (Susceptible-Infectious-Susceptible) are applied to predict the spread of disease, preventing outbreaks and optimising vaccine distribution. The review also demonstrates how Machine Learning (ML) algorithms and predictive analytics improve our knowledge of disease propagation patterns. The collaborative aspect of AI in vaccine discovery and clinical trials of various vaccines is emphasised, focusing on constructing AI-powered surveillance networks. Conclusively, the review presents a comprehensive assessment of how AI impacts epidemiological modelling, builds AI-enabled dynamic models by collaborating ML and Deep Learning (DL) techniques, and develops and implements vaccines and clinical trials. The review also focuses on screening, forecasting, contact tracing and monitoring the virus-causing pandemic. It advocates for sustained research, real-world implications, ethical application and strategic integration of AI technologies to strengthen our collective ability to face and alleviate the effects of global health issues.
Collapse
Affiliation(s)
- Mayur Suresh Gawande
- Department of Artificial Intelligence and Data Science, Faculty of Engineering and Technology, Datta Meghe Institute of Higher Education and Research (Deemed to Be University), Sawangi (Meghe), Wardha, Maharashtra, 442001, India
| | - Nikita Zade
- Department of Artificial Intelligence and Data Science, Faculty of Engineering and Technology, Datta Meghe Institute of Higher Education and Research (Deemed to Be University), Sawangi (Meghe), Wardha, Maharashtra, 442001, India
| | - Praveen Kumar
- Department of Computer Science and Medical Engineering, Faculty of Engineering and Technology, Datta Meghe Institute of Higher Education and Research (Deemed to Be University), Sawangi (Meghe), Wardha, Maharashtra, 442001, India.
| | - Swapnil Gundewar
- Department of Artificial Intelligence and Machine Learning, Faculty of Engineering and Technology, Datta Meghe Institute of Higher Education and Research (Deemed to Be University), Wardha, Maharashtra, 442001, India
| | - Induni Nayodhara Weerarathna
- Department of Biomedical Sciences, School of Allied Health Sciences, Datta Meghe Institute of Higher Education and Research (Deemed to Be University), Wardha, Maharashtra, 442001, India
| | - Prateek Verma
- Department of Artificial Intelligence and Machine Learning, Faculty of Engineering and Technology, Datta Meghe Institute of Higher Education and Research (Deemed to Be University), Wardha, Maharashtra, 442001, India
| |
Collapse
|
174
|
Gasmi A, Kassym L, Menzel A, Anzar W, Dadar M, Semenova Y, Arshad M, Bihunyak T, Meguid NA, Peana M, Bekbergenova Z, Bjørklund G. Genetic and Epigenetic Determinants of COVID-19 Susceptibility: A Systematic Review. Curr Med Chem 2025; 32:753-770. [PMID: 38251695 DOI: 10.2174/0109298673267890231221100659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/04/2023] [Accepted: 11/14/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND The molecular mechanisms regulating coronavirus pathogenesis are complex, including virus-host interactions associated with replication and innate immune control. However, some genetic and epigenetic conditions associated with comorbidities increase the risk of hospitalization and can prove fatal in infected patients. This systematic review will provide insight into host genetic and epigenetic factors that interfere with COVID-19 expression in light of available evidence. METHODS This study conducted a systematic review to examine the genetic and epigenetic susceptibility to COVID-19 using a comprehensive approach. Through systematic searches and applying relevant keywords across prominent online databases, including Scopus, PubMed, Web of Science, and Science Direct, we compiled all pertinent papers and reports published in English between December 2019 and June 2023. RESULTS The findings reveal that the host's HLA genotype plays a substantial role in determining how viral protein antigens are showcased and the subsequent immune system reaction to these antigens. Within females, genes responsible for immune system regulation are found on the X chromosome, resulting in reduced viral load and inflammation levels when contrasted with males. Possessing blood group A may contribute to an increased susceptibility to contracting COVID-19 as well as a heightened risk of mortality associated with the disease. The capacity of SARS-CoV-2 involves inhibiting the antiviral interferon (IFN) reactions, resulting in uncontrolled viral multiplication. CONCLUSION There is a notable absence of research into the gender-related predisposition to infection, necessitating a thorough examination. According to the available literature, a significant portion of individuals affected by the ailment or displaying severe ramifications already had suppressed immune systems, categorizing them as a group with elevated risk.
Collapse
Affiliation(s)
- Amin Gasmi
- Department of Research, Société Francophone de Nutrithérapie et de Nutrigénétique Appliquée, Villeurbanne, France
| | - Laura Kassym
- Department of Research, Astana Medical University, Astana, Kazakhstan
| | - Alain Menzel
- Department of Research, Laboratoires Réunis, Junglinster, Luxembourg
| | - Wajiha Anzar
- Department of Research, Dow University of Health Sciences, Karachi, Pakistan
| | - Maryam Dadar
- Department of Research, CONEM Iran Microbiology Research Group, Tehran, Iran
| | - Yuliya Semenova
- Department of Research, Nazarbayev University School of Medicine, Astana, Kazakhstan
| | - Mehreen Arshad
- Department of Research, National University of Sciences and Technology, Islamabad, Pakistan
| | - Tetyana Bihunyak
- Department of Research, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Nagwa Abdel Meguid
- Research on Children with Special Needs Department, National Research Centre, Giza, Egypt
- CONEM Egypt Child Brain Research Group, National Research Center, Giza, Egypt
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Sassari, Italy
| | | | - Geir Bjørklund
- Department of Research, Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| |
Collapse
|
175
|
Garcia-Segura P, Llop-Peiró A, Novau-Ferré N, Mestres-Truyol J, Saldivar-Espinoza B, Pujadas G, Garcia-Vallvé S. SARS-CoV-2 main protease (M-pro) mutational profiling: An insight into mutation coldspots. Comput Biol Med 2025; 184:109344. [PMID: 39531923 DOI: 10.1016/j.compbiomed.2024.109344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/20/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
SARS-CoV-2 and the COVID-19 pandemic have marked a milestone in the history of scientific research worldwide. To ensure that treatments are successful in the mid-long term, it is crucial to characterize SARS-CoV-2 mutations, as they might lead to viral resistance. Data from >5,700,000 SARS-CoV-2 genomes available at GISAID was used to report SARS-CoV-2 mutations. Given the pivotal role of its main protease (M-pro) in virus replication, a detailed analysis of SARS-CoV-2 M-pro mutations was conducted, with particular attention to mutation-resistant residues or mutation coldspots, defined as those residues that have mutated in five or fewer genomes. 32 mutation coldspots were identified, most of which mediate interprotomer interactions or funneling interaction networks from the substrate-binding site towards the dimerization surface and vice versa. Besides, mutation coldspots were virtually conserved in all main proteases from other CoVs. Our results provide valuable information about key residues to M-pro structure that could be useful in rational target-directed drug design and establish a solid groundwork based on mutation analyses for the inhibition of M-pro dimerization, with a potential applicability to future coronavirus outbreaks.
Collapse
Affiliation(s)
- Pol Garcia-Segura
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain.
| | - Ariadna Llop-Peiró
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain.
| | - Nil Novau-Ferré
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain.
| | - Júlia Mestres-Truyol
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain.
| | - Bryan Saldivar-Espinoza
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain.
| | - Gerard Pujadas
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain
| | - Santiago Garcia-Vallvé
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Research group in Cheminformatics & Nutrition, Campus de Sescelades, 43007, Tarragona, Spain.
| |
Collapse
|
176
|
Wu Y, He L, Li R, Li J, Zhao Q, Shao B. A20 as a Potential Therapeutic Target for COVID-19. Immun Inflamm Dis 2025; 13:e70127. [PMID: 39853876 PMCID: PMC11760982 DOI: 10.1002/iid3.70127] [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: 04/12/2024] [Revised: 11/29/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major concern due to its astonishing prevalence and high fatality rate, especially among elderly people. Patients suffering from COVID-19 may exhibit immunosuppression in the initial stage of infection, while a cytokine storm can occur when the disease progresses to a severe stage. This inopportune immune rhythm not only makes patients more susceptible to the virus but also leads to numerous complications resulting from the excessive production of inflammatory factors. A20, which is widely accepted as a pivotal regulator of inflammation, has been shown to be implicated in the processes of antiviral responses and immunosuppression. Thus, A20 may participate in regulating the pathological processes of COVID-19. METHODS This narrative literature review summarizes recent evidence on the mechanisms of A20 in regulating the pathological processes of COVID-19. We also downloaded single-cell RNA-seq data sets from healthy individuals and patients with varying severities of COVID-19 from the NCBI GEO database to further dissect A20's regulatory mechanisms of these intricate cytokine pathways that are closely associated with SARS-CoV-2 infection. RESULTS A20 might be one of the most critical anti-infectious and anti-inflammatory factors involved in the pathogenesis of COVID-19. It effectively suppresses the immune damage and inflammatory storm caused by viral infection. CONCLUSIONS Understanding the relationship between A20-regulated signaling pathways and pathological processes of COVID-19 can provide insight into potential targets for intervention. Precise regulation of A20 to induce antiviral activity and an anti-inflammatory response could mediate the pathogenesis of COVID-19 and could become an effective treatment.
Collapse
Affiliation(s)
- Yongyao Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Lilan He
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Rong Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Jiuxuan Li
- Laboratory of Radiation Biology, Laboratory Medicine Centre, Department of Blood TransfusionThe Second Affiliated HospitalArmy Military Medical UniversityChongqingChina
| | - Qing Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Bin Shao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
177
|
Alosaimi B, Awadalla M, Alturaiki W, Chen Z, Zhang Z, Zhu A, Rebh F, Alshukairi AN, Zhao J, Alkadi HS. A 10 Year Long-Lived Cellular and Humoral MERS-CoV Immunity Cross-Recognizing the Wild-Type and Variants of SARS-CoV-2: A Potential One-Way MERS-CoV Cross-Protection Toward a Pan-Coronavirus Vaccine. J Med Virol 2025; 97:e70071. [PMID: 39822038 PMCID: PMC11740004 DOI: 10.1002/jmv.70071] [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: 05/13/2024] [Revised: 10/09/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025]
Abstract
MERS is a respiratory disease caused by MERS-CoV. Multiple outbreaks have been reported, and the virus co-circulates with SARS-CoV-2. The long-term (> 6 years) cellular and humoral immune responses to MERS-CoV and their potential cross-reactivity to SARS-CoV-2 and its variants are unknown. We comprehensively investigated long-lasting MERS-CoV-specific cellular and humoral immunity, and its cross-reactivity against SARS-CoV-2 and its variants, in individuals recovered from MERS-CoV infection 1-10 years prior. Two cohorts of MERS-CoV survivors (31 unvaccinated, 38 COVID-19 vaccinated) were assessed for MERS-CoV IgG, memory CD4+/CD8+ T cells, and neutralizing antibodies against MERS-CoV and SARS-CoV-2 variants. MERS-CoV IgG levels and T cell responses were higher in the 1-5 vs 6-10 year postinfection groups. Vaccinated MERS-CoV survivors had significantly elevated MERS-CoV IgG and neutralization compared to unvaccinated. Both groups demonstrated cross-reactive neutralization of SARS-CoV-2 variants. MERS-CoV survivors vaccinated against SARS-CoV-2 had higher anti-MERS IgG, cellular immunity, and neutralization than unvaccinated survivors. MERS-CoV immune responses can persist for a decade. COVID-19 vaccination boosted humoral and cellular immunity in MERS-CoV survivors, suggesting the benefits of vaccination for this population. These findings have implications for pan-coronavirus vaccine development.
Collapse
Affiliation(s)
- Bandar Alosaimi
- Research Center, King Fahad Medical City, Riyadh Second Health ClusterRiyadhSaudi Arabia
| | - Maaweya Awadalla
- Research Center, King Fahad Medical City, Riyadh Second Health ClusterRiyadhSaudi Arabia
| | - Wael Alturaiki
- Department of Medical Laboratory SciencesCollege of Applied Medical Sciences, Majmaah University, MajmaahRiyadh RegionSaudi Arabia
| | - Zhao Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Zhaoyong Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Fatimah Rebh
- Department of Internal MedicineSection of Infectious Diseases, Prince Mohammed Bin Abdulaziz HospitalRiyadhSaudi Arabia
| | - Abeer N. Alshukairi
- Department of MedicineKing Faisal Specialist Hospital and Research CenterJeddahSaudi Arabia
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
- Guangzhou laboratory, Bio‐islandGuangzhouGuangdongChina
| | - Haitham S. Alkadi
- Research Center, King Fahad Medical City, Riyadh Second Health ClusterRiyadhSaudi Arabia
| |
Collapse
|
178
|
Okada K, Kin C, Yamashita Y, Kawamura S, Sato K, Chiba K, Miyake H. Possible mechanisms of spermatogenic dysfunction induced by viral infections: Insights from COVID-19. Reprod Med Biol 2025; 24:e12625. [PMID: 39845480 PMCID: PMC11751869 DOI: 10.1002/rmb2.12625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Background As the COVID-19 pandemic nears resolution in 2024, the mechanisms by which SARS-CoV-2 and other viral infections induce spermatogenic dysfunction remain poorly understood. This review examines the mechanisms by which viral infections, particularly COVID-19, disrupt spermatogenesis and highlights the implications for male reproductive health. While reports suggest that spermatogenic dysfunction caused by COVID-19 is mild and transient, these findings may have broader applications in understanding and treating spermatogenic dysfunction caused by future viral infections. Methods The PubMed database was searched to identify original and review articles investigating the mechanisms by which viral infections, particularly SARS-CoV-2, contribute to spermatogenic dysfunction. Main Findings SARS-CoV-2 affects the testis through multiple mechanisms, including ACE2 receptor-mediated entry, direct viral damage, inflammatory response, blood-testis barrier disruption, hormonal imbalance, oxidative stress, and impaired spermatogenesis. The combination of these factors can disrupt testicular function and highlights the complexity of the effects of COVID-19 on male reproductive health. Conclusion COVID-19 may disrupt spermatogenesis through direct testicular infection, systemic inflammation, hormonal disruption, and oxidative stress. Ongoing research, vaccination efforts, and clinical vigilance are essential to address these challenges and develop effective treatment and prevention strategies.
Collapse
Affiliation(s)
- Keisuke Okada
- Department of UrologyKobe City Medical Center West HospitalKobeJapan
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| | - Chanhyon Kin
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| | - Yosuke Yamashita
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| | - Shun Kawamura
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| | - Katsuya Sato
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| | - Koji Chiba
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| | - Hideaki Miyake
- Division of Urology, Department of Organs TherapeuticsKobe University Graduate School of MedicineKobeJapan
| |
Collapse
|
179
|
Abbasifard M, Ostad Ebrahimi H, Taghipur Khajeh Sharifi G, Bahrehmand F, Bagheri-Hosseinabadi Z. Investigation of the circulatory microRNAs and their involvement in regulation of inflammation in patients with COVID-19. Hum Immunol 2025; 86:111208. [PMID: 39667207 DOI: 10.1016/j.humimm.2024.111208] [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: 11/09/2024] [Accepted: 11/26/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Dysregulated levels of cytokines may lead to cytokine storm, which has been implicated in the immunopathogenesis of coronavirus disease 2019 (COVID-19). Here in the current study, the role of microRNA (miR)-155-5p, miR-146a, and miR-221-3p in the regulation of the immune responses and inflammatory state in patients with COVID-19 was investigated. METHODS In this case-control study, peripheral blood samples were obtained from 75 COVID-19 subjects and 100 healthy controls. From the plasma samples, RNA was extracted and cDNA was synthesized, and subsequently the transcript level of miRNAs was measured by Real-time PCR. The plasma levels of interleukin (IL)-4 and interferon (IFN)-γ were determined using ELISA. RESULTS miR-155-5p (fold change = 1.87, P = 0.020) and miR-221-3p (fold change = 2.26, P = 0.008), but not miR-146a, was upregulated in the plasma sample of COVID-19 cases compared to controls. The level of IFN-γ (but not IL-4) was significantly higher in the plasma samples of COVID-19 patients compared to control group. The expression level of miR-155-5p (r = 0.35, corrected P = 0.066) and miR-221-3p (r = 0.25, corrected P = 0.066) had positive correlation with the plasma levels of IFN-γ. CONCLUSIONS IFN-γ pathway in involved in the pathogenesis of COVID-19 that is regulated through miR-155-5p and miR-221-3p. These miRNAs showed potential utility as biomarkers for predicting the severity of COVID-19.
Collapse
Affiliation(s)
- Mitra Abbasifard
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hamid Ostad Ebrahimi
- Department of Paediatrics, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | | - Fatemeh Bahrehmand
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Zahra Bagheri-Hosseinabadi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Clinical Biochemistry, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
180
|
Fathi P, Alfonso AL, Yek C, Putman Z, Drew M, Esposito D, Zaidi I, Chea S, Ly S, Sath R, Lon C, Chea H, Leang R, Huy R, Ly S, Seng H, Tan CW, Zhu F, Wang L, Oliveira F, Sadtler K, Manning J. Humoral Immunity Profiling to Pandemic and Bat-Derived Coronavirus Variants: A Geographical Comparison. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2403503. [PMID: 39471070 PMCID: PMC11714182 DOI: 10.1002/advs.202403503] [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: 04/03/2024] [Revised: 09/05/2024] [Indexed: 11/01/2024]
Abstract
Dynamic pathogen exposure may impact the immunological response to SARS-CoV-2 (SCV2). One potential explanation for the lack of severe SCV2-related morbidity and mortality in Southeast Asia is prior exposure to related betacoronaviruses. Recent discoveries of SCV2-related betacoronaviruses from horseshoe bats (Rhinolophus sinicus) in Thailand, Laos, and Cambodia suggest the potential for bat-to-human spillover exposures in the region. In this work, serum antibodies to protein constructs from SCV2 and a representative bat coronavirus isolated in Cambodia (RshSTT182) are measured in pre-pandemic Cambodian human sera using ELISA assays. Of 293 Cambodian samples tested (N = 131 with acute malaria, n = 162 with acute undifferentiated febrile illness), 32 (10.9%) are seropositive for SCV2 based on established Spike and receptor-binding domain (RBD) cutoffs. Within SCV2 seropositive samples, 16 (50%) have higher antibody levels to antigens from the representative virus RshSTT182 versus SCV2 antigens; competitive binding ELISA assays demonstrate inhibition of reactivity to SCV2 Spike after pre-incubation with RshSTT182 Spike. Surrogate virus neutralization tests demonstrate that 8/30 (26.7%) SCV2 ELISA positive pre-pandemic Cambodian samples have neutralizing activity against SCV2, while 14/30 (46.7%) have activity against other SCV2-related betacoronaviruses. These data suggest that exposure to related betacoronaviruses may elicit cross-reactive immunity to SCV2 prior to the global pandemic.
Collapse
Affiliation(s)
- Parinaz Fathi
- Section on ImmunoengineeringBiomedical Engineering and Technology Acceleration CenterNational Institute of Biomedical Imaging and BioengineeringBethesdaMD20892USA
| | - Andrea Lucia Alfonso
- Section on ImmunoengineeringBiomedical Engineering and Technology Acceleration CenterNational Institute of Biomedical Imaging and BioengineeringBethesdaMD20892USA
| | - Christina Yek
- Laboratory of Malaria and Vector ResearchNational Institute of Allergy and Infectious DiseasesRockvilleMD20892USA
| | - Zoe Putman
- Protein Expression LaboratoryNCI RAS InitiativeFrederick National Laboratory for Cancer ResearchFrederickMD21701USA
| | - Matthew Drew
- Protein Expression LaboratoryNCI RAS InitiativeFrederick National Laboratory for Cancer ResearchFrederickMD21701USA
| | - Dominic Esposito
- Protein Expression LaboratoryNCI RAS InitiativeFrederick National Laboratory for Cancer ResearchFrederickMD21701USA
| | - Irfan Zaidi
- Laboratory of Malaria Immunology and VaccinologyNational Institute of Allergy and Infectious DiseasesBethesdaMD20892USA
| | - Sophana Chea
- International Center of Excellence in Research CambodiaNational Institute of Allergy and Infectious DiseasesPhnom Penh120801Cambodia
| | - Sokna Ly
- International Center of Excellence in Research CambodiaNational Institute of Allergy and Infectious DiseasesPhnom Penh120801Cambodia
| | - Rathanak Sath
- International Center of Excellence in Research CambodiaNational Institute of Allergy and Infectious DiseasesPhnom Penh120801Cambodia
| | - Chanthap Lon
- International Center of Excellence in Research CambodiaNational Institute of Allergy and Infectious DiseasesPhnom Penh120801Cambodia
| | - Huch Chea
- National Center for Parasitology, Entomology, and Malaria ControlMinistry of HealthPhnom Penh120801Cambodia
| | - Rithea Leang
- National Center for Parasitology, Entomology, and Malaria ControlMinistry of HealthPhnom Penh120801Cambodia
| | - Rekol Huy
- National Center for Parasitology, Entomology, and Malaria ControlMinistry of HealthPhnom Penh120801Cambodia
| | - Sovann Ly
- Cambodian Center for Disease ControlMinistry of HealthPhnom Penh120407Cambodia
| | - Heng Seng
- Cambodian Center for Disease ControlMinistry of HealthPhnom Penh120407Cambodia
| | - Chee Wah Tan
- Programme for Emerging Infectious DiseasesDuke‐National University of Singapore Medical School169857SingaporeSingapore
- Infectious Diseases Translational Research ProgrammeDepartment of Microbiology and ImmunologyYong Loo Lin School of MedicineNational University of Singapore117597SingaporeSingapore
| | - Feng Zhu
- Programme for Emerging Infectious DiseasesDuke‐National University of Singapore Medical School169857SingaporeSingapore
| | - Lin‐Fa Wang
- Programme for Emerging Infectious DiseasesDuke‐National University of Singapore Medical School169857SingaporeSingapore
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector ResearchNational Institute of Allergy and Infectious DiseasesRockvilleMD20892USA
| | - Kaitlyn Sadtler
- Section on ImmunoengineeringBiomedical Engineering and Technology Acceleration CenterNational Institute of Biomedical Imaging and BioengineeringBethesdaMD20892USA
| | - Jessica Manning
- Laboratory of Malaria and Vector ResearchNational Institute of Allergy and Infectious DiseasesRockvilleMD20892USA
- International Center of Excellence in Research CambodiaNational Institute of Allergy and Infectious DiseasesPhnom Penh120801Cambodia
- Present address:
SanofiWashingtonDC20004USA
| |
Collapse
|
181
|
Zhou X, Liu H, Yang S, Dong X, Xie C, Ou W, Chen J, Yang Z, Ye Y, Ivanov KI, Liu L, Zou J, Li C, Guo D. Rapid Generation of Reverse Genetics Systems for Coronavirus Research and High-Throughput Antiviral Screening Using Gibson DNA Assembly. J Med Virol 2025; 97:e70171. [PMID: 39821287 DOI: 10.1002/jmv.70171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/01/2024] [Accepted: 12/03/2024] [Indexed: 01/19/2025]
Abstract
Coronaviruses (CoVs) pose a significant threat to human health, as demonstrated by the COVID-19 pandemic. The large size of the CoV genome (around 30 kb) represents a major obstacle to the development of reverse genetics systems, which are invaluable for basic research and antiviral drug screening. In this study, we established a rapid and convenient method for generating reverse genetic systems for various CoVs using a bacterial artificial chromosome (BAC) vector and Gibson DNA assembly. Using this system, we constructed infectious cDNA clones of coronaviruses from three genera: human coronavirus 229E (HCoV-229E) of the genus Alphacoronavirus, mouse hepatitis virus A59 (MHV-59) of Betacoronavirus, and porcine deltacoronavirus (PDCoV-Haiti) of Deltacoronavirus. Since beta coronaviruses including severe acute respiratory syndrome coronavirus (SARS-CoV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and Middle East respiratory syndrome coronavirus (MERS-CoV) represent major human pathogens, we modified the infectious clone of the beta coronavirus MHV-A59 by replacing its NS5a gene with a fluorescent reporter gene to create a system suitable for high-throughput drug screening. Thus, this study provides a practical and cost-effective approach to developing reverse genetics platforms for CoV research and antiviral drug screening.
Collapse
Affiliation(s)
- Xinrong Zhou
- 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
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Hongchao Liu
- 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
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Sidi Yang
- 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
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Xiaoran Dong
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Cailing Xie
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Weixin Ou
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Jinwei Chen
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Zixiao Yang
- MOE Key Laboratory of Tropical Disease Control, Center for Infection and Immunity Studies (CIIS), School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yuzhen Ye
- 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
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Konstantin I Ivanov
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Lihong Liu
- 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
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
| | - Jie Zou
- MOE Key Laboratory of Tropical Disease Control, Center for Infection and Immunity Studies (CIIS), School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Chunmei Li
- MOE Key Laboratory of Tropical Disease Control, Center for Infection and Immunity Studies (CIIS), School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Deyin Guo
- 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
- Department of Basic Research, Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, Guangdong, China
- Department of Pathogen Biology and Biosecurity, Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
182
|
Kadoya SS, Li Y, Wang Y, Katayama H, Sano D. State-space modelling using wastewater virus and epidemiological data to estimate reported COVID-19 cases and the potential infection numbers. J R Soc Interface 2025; 22:20240456. [PMID: 39772733 PMCID: PMC11706650 DOI: 10.1098/rsif.2024.0456] [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: 07/03/2024] [Revised: 08/25/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
The current situation of COVID-19 measures makes it difficult to accurately assess the prevalence of SARS-CoV-2 due to a decrease in reporting rates, leading to missed initial transmission events and subsequent outbreaks. There is growing recognition that wastewater virus data assist in estimating potential infections, including asymptomatic and unreported infections. Understanding the COVID-19 situation hidden behind the reported cases is critical for decision-making when choosing appropriate social intervention measures. However, current models implicitly assume homogeneity in human behaviour, such as virus shedding patterns within the population, making it challenging to predict the emergence of new variants due to variant-specific transmission or shedding parameters. This can result in predictions with considerable uncertainty. In this study, we established a state-space model based on wastewater viral load to predict both reported cases and potential infection numbers. Our model using wastewater virus data showed high goodness-of-fit to COVID-19 case numbers despite the dataset including waves of two distinct variants. Furthermore, the model successfully provided estimates of potential infection, reflecting the superspreading nature of SARS-CoV-2 transmission. This study supports the notion that wastewater surveillance and state-space modelling have the potential to effectively predict both reported cases and potential infections.
Collapse
Affiliation(s)
- Syun-suke Kadoya
- Department of Urban Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo113-8656, Japan
| | - Yubing Li
- Department of Frontier Science for Advanced Environment, Graduate School of Environmental Studies, Tohoku University, Aoba 6-6-06, Aramaki, Aoba-ku, Sendai, Miyagi980-8579, Japan
| | - Yilei Wang
- Department of Frontier Science for Advanced Environment, Graduate School of Environmental Studies, Tohoku University, Aoba 6-6-06, Aramaki, Aoba-ku, Sendai, Miyagi980-8579, Japan
| | - Hiroyuki Katayama
- Department of Urban Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo113-8656, Japan
| | - Daisuke Sano
- Department of Frontier Science for Advanced Environment, Graduate School of Environmental Studies, Tohoku University, Aoba 6-6-06, Aramaki, Aoba-ku, Sendai, Miyagi980-8579, Japan
- Department of Civil and Environmental Engineering, Graduate School of Engineering, Tohoku University, Aoba 6-6-06, Aramaki, Aoba-ku, Sendai, Miyagi980-8579, Japan
| |
Collapse
|
183
|
Silvério BS, Guilardi MD, Martins JO, Duro RLS, de Sousa LLF, Cabral-Miranda G, Janini LMR, Poon LLM, Durães-Carvalho R. Coronavirus Cryptic Landscape and Draft Genome of a Novel CoV Clade Related to MERS From Bats Circulating in Northeastern Brazil. J Med Virol 2025; 97:e70173. [PMID: 39825725 DOI: 10.1002/jmv.70173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/03/2024] [Accepted: 01/05/2025] [Indexed: 01/20/2025]
Abstract
We identified seven distinct coronaviruses (CoVs) in bats from Brazil, classified into 229E-related (Alpha-CoV), Nobecovirus, Sarbecovirus, and Merbecovirus (Beta-CoV), including one closely related to MERS-like CoV with 82.8% genome coverage. To accomplish this, we screened 423 oral and rectal swabs from 16 different bat species using molecular assays, RNA sequencing, and evolutionary analysis. Notably, gaps in the spike-encoding gene led us to design new primers and perform Sanger sequencing, which revealed high similarities to MERS-related (MERSr) CoV strains found in humans and camels. Additionally, we identified key residues in the receptor-binding domain (RBD) of the spike protein, suggesting potential interactions with DPP4, the receptor for MERSr-CoV. Our analyses also revealed evidence of recombination involving our laboratory-produced sequences. These findings highlight the extensive genetic diversity of CoVs, the presence of novel viral lineages, and the occurrence of recombination events among bat CoVs circulating in Brazil, underscoring the critical role bats play as reservoirs for emerging viruses and emphasizing the necessity of ongoing surveillance to monitor the public health risks associated with CoV spillover events.
Collapse
Affiliation(s)
- Bruna Stefanie Silvério
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo-SP, Brazil
| | - Mariana Dias Guilardi
- Interunit Bioinformatics Graduate Program, Institute of Chemistry, University of São Paulo, São Paulo-SP, Brazil
| | - Junior Olímpio Martins
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo-SP, Brazil
| | - Rodrigo Lopes Sanz Duro
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Larissa Leão F de Sousa
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
- Rabies Diagnosis Laboratory, Central Laboratory of Public Health - LACEN, Fortaleza-CE, Brazil
| | | | - Luiz Mário Ramos Janini
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Leo L M Poon
- School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Ricardo Durães-Carvalho
- Department of Morphology and Genetics, Federal University of São Paulo, São Paulo-SP, Brazil
- Interunit Bioinformatics Graduate Program, Institute of Chemistry, University of São Paulo, São Paulo-SP, Brazil
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
184
|
Kaondera-Shava RF, Galanti M, Perini M, Suh J, Farley SM, Chicumbe S, Jani I, Cassy A, Macicame I, Manafe N, El-Sadr W, Shaman J. SARS-CoV-2 transmission dynamics in Mozambique and Zimbabwe during the first 3 years of the pandemic. ROYAL SOCIETY OPEN SCIENCE 2025; 12:241275. [PMID: 39845719 PMCID: PMC11750399 DOI: 10.1098/rsos.241275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 01/24/2025]
Abstract
The 2019 emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its rapid spread created a public health emergency of international concern. However, the impact of the pandemic in Sub-Saharan Africa, as documented in cases, hospitalizations and deaths, appears far lower than in the Americas, Europe and Asia. Characterization of the transmission dynamics is critical for understanding how SARS-CoV-2 spreads and the true scale of the pandemic. Here, to better understand SARS-CoV-2 transmission dynamics in two southern African countries, Mozambique and Zimbabwe, we developed a dynamic model-Bayesian inference system to estimate key epidemiological parameters, namely the transmission and ascertainment rates. Total infection burdens (reported and unreported) during the first 3 years of the pandemic were reconstructed using a model-inference approach. Transmission rates rose with each successive wave, which aligns with observations in other continents. Ascertainment rates were found to be low and consistent with other African countries. Overall, the estimated disease burden was higher than the documented cases, indicating a need for improved reporting and surveillance. These findings aid understanding of COVID-19 disease and respiratory virus transmission dynamics in two African countries little investigated to date and can help guide future public health planning and control strategies.
Collapse
Affiliation(s)
- Roselyn F. Kaondera-Shava
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY10032, USA
| | - Marta Galanti
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY10032, USA
| | - Matteo Perini
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY10032, USA
| | - Jiyeon Suh
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY10032, USA
| | - Shannon M. Farley
- Department of Population and Family Health, Mailman School of Public Health, Columbia University, New York, NY10032, USA
| | - Sergio Chicumbe
- Instituto Nacional de Saúde (INS) EN1, Bairro da Vila-Parcela n 3943, Distrito de Marracuene, Maputo, C.P.264, Mozambique
| | - Ilesh Jani
- Instituto Nacional de Saúde (INS) EN1, Bairro da Vila-Parcela n 3943, Distrito de Marracuene, Maputo, C.P.264, Mozambique
| | - Annette Cassy
- Instituto Nacional de Saúde (INS) EN1, Bairro da Vila-Parcela n 3943, Distrito de Marracuene, Maputo, C.P.264, Mozambique
| | - Ivalda Macicame
- Instituto Nacional de Saúde (INS) EN1, Bairro da Vila-Parcela n 3943, Distrito de Marracuene, Maputo, C.P.264, Mozambique
| | - Naisa Manafe
- Instituto Nacional de Saúde (INS) EN1, Bairro da Vila-Parcela n 3943, Distrito de Marracuene, Maputo, C.P.264, Mozambique
| | - Wafaa El-Sadr
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY10032, USA
| | - Jeffrey Shaman
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY10032, USA
- Columbia Climate School, Columbia University, New York, NY10025, USA
| |
Collapse
|
185
|
Mallick S, Duttaroy AK, Bose B. A Snapshot of Cytokine Dynamics: A Fine Balance Between Health and Disease. J Cell Biochem 2025; 126:e30680. [PMID: 39668456 DOI: 10.1002/jcb.30680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/23/2024] [Accepted: 11/08/2024] [Indexed: 12/14/2024]
Abstract
Health and disease are intricately intertwined and often determined by the delicate balance of biological processes. Cytokines, a family of small signalling molecules, are pivotal in maintaining this balance, ensuring the body's immune system functions optimally. In a healthy condition, cytokines act as potent mediators of immune responses. They orchestrate the activities of immune cells, coordinating their proliferation, differentiation, and migration. This intricate role of cytokine signalling enables the body to effectively combat infections, repair damaged tissues, and regulate inflammation. However, the delicate equilibrium of cytokine production is susceptible to disruption. Excessive or abnormal cytokine levels can lead to a cascade of pathological conditions, including autoimmune diseases, chronic inflammation, infections, allergies, and even cancer. Interestingly, from the bunch of cytokines, few cytokines play an essential role in maintaining the balance between normal physiological status and diseases. In this review, we have appraised key cytokines' potential role and feedback loops in augmenting the imbalances in the body's biological functions, presenting a critical link between inflammation and disease pathology. Moreover, we have also highlighted the significance of cytokines and their molecular interplay, particularly in the recent viral pandemic COVID-19 disease. Hence, understandings regarding the interplay between viral infection and cytokine responses are essential and fascinating for developing effective therapeutic strategies.
Collapse
Affiliation(s)
- Sumit Mallick
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka, India
| |
Collapse
|
186
|
Mazzotti L, Borges de Souza P, Azzali I, Angeli D, Nanni O, Sambri V, Semprini S, Bravaccini S, Cerchione C, Gaimari A, Nicolini F, Ancarani V, Martinelli G, Pasetto A, Calderon H, Juan M, Mazza M. Exploring the Relationship Between Humoral and Cellular T Cell Responses Against SARS-CoV-2 in Exposed Individuals From Emilia Romagna Region and COVID-19 Severity. HLA 2025; 105:e70011. [PMID: 39807702 PMCID: PMC11731316 DOI: 10.1111/tan.70011] [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: 07/09/2024] [Revised: 11/03/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025]
Abstract
COVID-19 remains a significant global health problem with uncertain long-term consequences for convalescents. We investigated the relationships between anti-N protein antibody levels, severe acute respiratory syndrome (SARS)-CoV-2-associated TCR repertoire parameters, HLA type and epidemiological information from three cohorts of 524 SARS-CoV-2-infected subjects subgrouped in acute phase, seronegative and seropositive convalescents from the Emilia Romagna region. Epidemiological information and anti-N antibody index were associated with TCR repertoire data. HLA type was inferred from TCR repertoire using the HLA3 tool and its association with clonal breadth (CB) and clonal depth (CD) was assessed. Age above 58 years, male and COVID-19 hospitalisation were significantly and independently associated with seropositivity (p = 0.004; p = 0.004; p = 0.04), suggesting an association between high antibody titres and symptoms' severity. As for the TCR repertoire analysis, we found no difference in CB among the cohorts, while CD was higher in seronegative than acute (p = 0.04). However, clustering analysis supported that seronegative patients are endowed with broader CB and deeper CD indicating a compensatory mechanism without effective seroconversion. The CD calculated on the TCRs associated with the single SARS-CoV-2 ORFs in convalescents is higher when compared to the acute. Lastly, we identified and reported on novel HLAs significantly associated with increased risk of hospitalisation such as HLA-C*07:02 carriers (OR = 3.9, CI = 1.1-13.4, p = 0.03) and on HLAs that associate significantly with lower or higher TCR repertoire parameters in a population exposed for the first time to SARS-CoV-2.
Collapse
Affiliation(s)
- Lucia Mazzotti
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | | | - Irene Azzali
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Davide Angeli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Oriana Nanni
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Vittorio Sambri
- Microbiology UnitThe Great Romagna Area Hub LaboratoryPievesestinaItaly
- DIMECBologna UniversityBolognaItaly
| | - Simona Semprini
- Microbiology UnitThe Great Romagna Area Hub LaboratoryPievesestinaItaly
| | - Sara Bravaccini
- Department of Medicine and SurgeryUniversity of Enna “Kore”EnnaItaly
| | - Claudio Cerchione
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Anna Gaimari
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Fabio Nicolini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Valentina Ancarani
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| | - Giovanni Martinelli
- Department of Hematology and Sciences OncologyInstitute of Haematology “L. and A. Seràgnoli” S. Orsola, University Hospital in BolognaBolognaItaly
| | - Anna Pasetto
- Section for Cell TherapyRadiumhospitalet, Oslo University HospitalOsloNorway
- Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
| | - Hugo Calderon
- Department of ImmunologyCentre de Diagnòstic Biomèdic, Hospital Clínic of BarcelonaBarcelonaSpain
| | - Manel Juan
- Department of ImmunologyCentre de Diagnòstic Biomèdic, Hospital Clínic of BarcelonaBarcelonaSpain
| | - Massimiliano Mazza
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori"MeldolaItaly
| |
Collapse
|
187
|
Ko C, Cheng CC, Mistretta D, Ambike S, Sacherl J, Velkov S, Liao BH, Bester R, Gültan M, Polezhaeva O, Herrmann A, Jakwerth CA, Schmidt-Weber CB, Bugert JJ, Wölfel R, Grass V, Essbauer S, Schnepf D, Keppler OT, Vondran FWR, Pichlmair A, Mogler C, Ebert G, Protzer U. SARS-CoV-2 Productively Infects Human Hepatocytes and Induces Cell Death. J Med Virol 2025; 97:e70156. [PMID: 39760326 DOI: 10.1002/jmv.70156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 11/25/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025]
Abstract
SARS-CoV-2 infection is accompanied by elevated liver enzymes, and patients with pre-existing liver conditions experience more severe disease. While it was known that SARS-CoV-2 infects human hepatocytes, our study determines the mechanism of infection, demonstrates viral replication and spread, and highlights direct hepatocyte damage. Viral replication was readily detectable upon infection of primary human hepatocytes and hepatoma cells with the ancestral SARS-CoV-2, Delta, and Omicron variants. Hepatocytes express the SARS-CoV-2 receptor ACE2 and the host cell protease TMPRSS2, and knocking down ACE2 and TMPRSS2 impaired SARS-CoV-2 infection. Progeny viruses released from infected hepatocytes showed the typical coronavirus morphology by electron microscopy and proved infectious when transferred to fresh cells, indicating that hepatocytes can contribute to virus spread. Importantly, SARS-CoV-2 infection rapidly induced hepatocyte death in a replication-dependent fashion, with the Omicron variant showing faster onset but less extensive cell death. C57BL/6 wild-type mice infected with a mouse-adapted SARS-CoV-2 strain showed high levels of viral RNA in liver and lung tissues. ALT peaked when viral RNA was cleared from the liver. Liver histology revealed profound tissue damage and immune cell infiltration, indicating that direct cytopathic effects of SARS-CoV-2 and immune-mediated killing of infected hepatocytes contribute to liver pathology.
Collapse
Grants
- This study was supported by the German Research Foundation (DFG) via SFB-TRR179 (project 272983813 to U.P.), TRR22 (project 398577603 to C.S.W.) and TRR353 (project 471011418 to G.E.), by the State of Bavaria via research network FOR-COVID and Bay-VOC, by the project "Virological and immunological determinants of COVID-19 pathogenesis-lessons to get prepared for future pandemics" (KA1-Co-02 "COVIPA" to U.P.) and "Airborne Transmission of SARS Coronavirus - From Fundamental Science to Efficient Air Cleaning Systems" (KA1-Co-06 "CORAERO" to G.E.), grants from the Helmholtz Association's Initiative and Networking Fund, by the European Commission FET Open Grant VIROFIGHT (grant no. 899619), by the State of Bavaria and the European Union via a grant for regional infrastructure development (EFRE - REACT, to U.P. and G.E.), by the State of Bavaria via research networks FOR-COVID and Bay-VOC (to U.P. and O.T.K.) by the Federal Ministry of Education and Research (project ESCAPE; 01KI20169A to C.S.W.), and by the Medical Biological Defense Research Program of the Bundeswehr Medical Service (to J.J.B.). In addition, this research was supported by intramural funds from KRICT (project KK2432-10 and BSF24-111 to C.K.).
Collapse
Affiliation(s)
- Chunkyu Ko
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, South Korea
| | - Cho-Chin Cheng
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Daniele Mistretta
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Shubhankar Ambike
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Julia Sacherl
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Stoyan Velkov
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Bo-Hung Liao
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Romina Bester
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Merve Gültan
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Olga Polezhaeva
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Alexander Herrmann
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Constanze A Jakwerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich/Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technical University of Munich/Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
- German Center for Lung Research (DZL), Munich Partner Site, Munich, Germany
| | - Joachim J Bugert
- Department of Viruses and Intracellular Pathogens, Bundeswehr Institute of Microbiology, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| | - Roman Wölfel
- Department of Viruses and Intracellular Pathogens, Bundeswehr Institute of Microbiology, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| | - Vincent Grass
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
| | - Sandra Essbauer
- Department of Viruses and Intracellular Pathogens, Bundeswehr Institute of Microbiology, Munich, Germany
| | - Daniel Schnepf
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK
| | - Oliver T Keppler
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
- Max von Pettenkofer Institute & Gene Center, Faculty of Medicine, University of Munich, Munich, Germany
| | - Florian W R Vondran
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
- ReMediES, Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Andreas Pichlmair
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| | - Carolin Mogler
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Gregor Ebert
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| | - Ulrike Protzer
- Institute of Virology, Technical University of Munich/Helmholtz Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner Sites Munich and Hannover-Braunschweig, Munich, Germany
| |
Collapse
|
188
|
Sultana R, Stahelin RV. Strengths and limitations of SARS-CoV-2 virus-like particle systems. Virology 2025; 601:110285. [PMID: 39536645 PMCID: PMC11624109 DOI: 10.1016/j.virol.2024.110285] [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: 08/05/2024] [Revised: 10/19/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Virus-like particles (VLPs) resemble the parent virus but lack the viral genome, providing a safe and efficient platform for the analysis of virus assembly and budding as well as the development of vaccines and drugs. During the COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the formation of SARS-CoV-2 VLPs was investigated as an alternative to authentic virions because the latter requires biosafety level 3 (BSL-3) facilities. This allowed researchers to model its assembly and budding processes, examine the role of mutations in variants of concern, and determine how the structural proteins interact with each other. Also, the absence of viral genome in VLPs circumvents worries of gains in infectivity via mutagenesis. This review summarizes the strengths and limitations of several SARS-CoV-2 VLP systems and details some of the strides that have been made in using these systems to study virus assembly and budding, viral entry, and antibody and vaccine development.
Collapse
Affiliation(s)
- Rokaia Sultana
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, 47907, West Lafayette, IN, USA
| | - Robert V Stahelin
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, 47907, West Lafayette, IN, USA.
| |
Collapse
|
189
|
Zhang Y, Song SJ, He J, Zhao ZH, Zhang K, Zhang Y, Li X. Targeted Drug Delivery to ACE2 + Cells Using Engineered Extracellular Vesicles: A Potential Therapeutic Approach for COVID-19. Curr Pharm Biotechnol 2025; 26:443-454. [PMID: 38551053 DOI: 10.2174/0113892010282251240324123038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/29/2024] [Accepted: 02/12/2024] [Indexed: 03/25/2025]
Abstract
BACKGROUND Extracellular vesicles (EVs) are emerging as potential drug carriers in the fight against COVID-19. This study investigates the ability of EVs as drug carriers to target SARS-CoV-2-infected cells. METHODS EVs were modified using Xstamp technology to carry the virus's RBD, enhancing targeting ability to hACE2+ cells and improving drug delivery efficiency. Characterization confirmed EVs' suitability as drug carriers. For in vitro tests, A549, Caco-2, and 4T1 cells were used to assess the targeting specificity of EVRs (EVs with membrane-surface enriched RBD). Moreover, we utilized an ex vivo lung tissue model overexpressing hACE2 as an ex vivo model to confirm the targeting capability of EVRs toward lung tissue. The study also evaluated drug loading efficiency and assessed the potential of the anti-inflammatory activity on A549 lung cancer cells exposed to lipopolysaccharide. RESULTS The results demonstrate the successful construction of RBD-fused EVRs on the membrane- surface. In both in vitro and ex vivo models, EVRs significantly enhance their targeting ability towards hACE2+ cells, rendering them a safe and efficient drug carrier. Furthermore, ultrasound loading efficiently incorporates IL-10 into EVRs, establishing an effective drug delivery system that ameliorates the pro-inflammatory response induced by LPS-stimulated A549 cells. CONCLUSION These findings indicate promising opportunities for engineered EVs as a novel nanomedicine carrier, offering valuable insights for therapeutic strategies against COVID-19 and other diseases.
Collapse
Affiliation(s)
- Yao Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Sheng-Jiao Song
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Jin He
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Zhuo-Hua Zhao
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Ke Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Yuan Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Xing Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| |
Collapse
|
190
|
Greene KS, Choi A, Yang N, Chen M, Li R, Qiu Y, Ezzatpour S, Rojas KS, Shen J, Wilson KF, Katt WP, Aguilar HC, Lukey MJ, Whittaker GR, Cerione RA. Glutamine metabolism is essential for coronavirus replication in host cells and in mice. J Biol Chem 2025; 301:108063. [PMID: 39662828 PMCID: PMC11750454 DOI: 10.1016/j.jbc.2024.108063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/21/2024] [Accepted: 11/30/2024] [Indexed: 12/13/2024] Open
Abstract
Understanding the fundamental biochemical and metabolic requirements for the replication of coronaviruses within infected cells is of notable interest for the development of broad-based therapeutic strategies, given the likelihood of the emergence of new pandemic-potential virus species, as well as future variants of SARS-CoV-2. Here we demonstrate members of the glutaminase family of enzymes (GLS and GLS2), which catalyze the hydrolysis of glutamine to glutamate (i.e., the first step in glutamine metabolism), play key roles in coronavirus replication in host cells. Using a range of human seasonal and zoonotic coronaviruses, we show three examples where GLS expression increases during coronavirus infection of host cells, and another where GLS2 is upregulated. The viruses hijack the metabolic machinery responsible for glutamine metabolism to generate the building blocks for biosynthetic processes and satisfy the bioenergetic requirements demanded by the "glutamine addiction" of virus-infected cells. We demonstrate that genetic silencing of glutaminase enzymes reduces coronavirus infection and that newer members of two classes of allosteric inhibitors targeting these enzymes, designated as SU1, a pan-GLS/GLS2 inhibitor, and UP4, a specific GLS inhibitor, block viral replication in epithelial cells. Moreover, treatment of SARS-CoV-2 infected K18-human ACE2 transgenic mice with SU1 resulted in their complete survival compared to untreated control animals, which succumbed within 10 days post-infection. Overall, these findings highlight the importance of glutamine metabolism for coronavirus replication in human cells and mice and show that glutaminase inhibitors can block coronavirus infection and thereby may represent a novel class of broad-based anti-viral drug candidates.
Collapse
Affiliation(s)
- Kai Su Greene
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Annette Choi
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA
| | - Nianhui Yang
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Matthew Chen
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Ruizhi Li
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Yijian Qiu
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Shahrzad Ezzatpour
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA
| | - Katherine S Rojas
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Jonathan Shen
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Kristin F Wilson
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - William P Katt
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Hector C Aguilar
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA
| | - Michael J Lukey
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Gary R Whittaker
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA; Public & Ecosystem Health, Cornell University, Ithaca, New York, USA
| | - Richard A Cerione
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, USA.
| |
Collapse
|
191
|
Guerra D, Radić L, Brinkkemper M, Poniman M, van der Maas L, Torres JL, Ward AB, Sliepen K, Schinkel J, Sanders RW, van Gils MJ, Beaumont T. Broadening sarbecovirus neutralization with bispecific antibodies combining distinct conserved targets on the receptor binding domain. Hum Vaccin Immunother 2024; 20:2388344. [PMID: 39165108 PMCID: PMC11340772 DOI: 10.1080/21645515.2024.2388344] [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: 05/02/2024] [Revised: 07/05/2024] [Accepted: 08/01/2024] [Indexed: 08/22/2024] Open
Abstract
Monoclonal neutralizing antibodies (mAbs) are considered an important prophylactic against SARS-CoV-2 infection in at-risk populations and a strategy to counteract future sarbecovirus-induced disease. However, most mAbs isolated so far neutralize only a few sarbecovirus strains. Therefore, there is a growing interest in bispecific antibodies (bsAbs) which can simultaneously target different spike epitopes and thereby increase neutralizing breadth and prevent viral escape. Here, we generate and characterize a panel of 30 novel broadly reactive bsAbs using an efficient controlled Fab-arm exchange protocol. We specifically combine some of the broadest mAbs described so far, which target conserved epitopes on the receptor binding domain (RBD). Several bsAbs show superior cross-binding and neutralization compared to the parental mAbs and cocktails against sarbecoviruses from diverse clades, including recent SARS-CoV-2 variants. BsAbs which include mAb COVA2-02 are among the most potent and broad combinations. As a result, we study the unknown epitope of COVA2-02 and show that this mAb targets a distinct conserved region at the base of the RBD, which could be of interest when designing next-generation bsAb constructs to contribute to a better pandemic preparedness.
Collapse
Affiliation(s)
- Denise Guerra
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Laura Radić
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Mitch Brinkkemper
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Meliawati Poniman
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Lara van der Maas
- Department of Structural Biology and Computational Biology, The Scripps Research Institute, La Jolla, USA
| | - Jonathan L. Torres
- Department of Structural Biology and Computational Biology, The Scripps Research Institute, La Jolla, USA
| | - Andrew B. Ward
- Department of Structural Biology and Computational Biology, The Scripps Research Institute, La Jolla, USA
| | - Kwinten Sliepen
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Janke Schinkel
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Rogier W. Sanders
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, USA
| | - Marit J. van Gils
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Tim Beaumont
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
192
|
Seo E, Shin EC, Jung MK. SARS-CoV-2 vaccine-elicited immune responses in solid organ transplant recipients. CLINICAL TRANSPLANTATION AND RESEARCH 2024; 38:247-256. [PMID: 39743229 PMCID: PMC11732761 DOI: 10.4285/ctr.24.0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Solid organ transplant recipients (SOTRs) are considered a high-risk group for coronavirus disease 2019 (COVID-19). The adaptive immune responses generated by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination include humoral and cellular immune responses. Most studies on the SARS-CoV-2 vaccine have focused primarily on humoral immunity, but cellular immunity is vital for effectively controlling progression to severe COVID-19. In SOTRs, the vaccine-induced adaptive immune response is significantly attenuated compared to the response in healthy individuals. Nevertheless, vaccinated SOTRs exhibit a reduced rate and severity of SARS-CoV-2 infection. This review aims to provide a concise overview of the current understanding of SARS-CoV-2 vaccine-induced immune responses in SOTRs.
Collapse
Affiliation(s)
- Euri Seo
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Korea
| | - Eui-Cheol Shin
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Min Kyung Jung
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon, Korea
| |
Collapse
|
193
|
Sangeda RZ, James D, Mariki H, Mbwambo ME, Mwenesi ME, Nyaki H, Tinuga F, Manyanga DP. Childhood vaccination trends during 2019 to 2022 in Tanzania and the impact of the COVID-19 pandemic. Hum Vaccin Immunother 2024; 20:2356342. [PMID: 38780570 PMCID: PMC11123454 DOI: 10.1080/21645515.2024.2356342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
The COVID-19 pandemic has significantly disrupted healthcare systems at all levels globally, notably affecting routine healthcare services, such as childhood vaccination. This study examined the impact of these disruptions on routine childhood vaccination programmes in Tanzania. We conducted a longitudinal study over four years in five Tanzanian regions: Mwanza, Dar es Salaam, Mtwara, Arusha, and Dodoma. This study analyzed the trends in the use of six essential vaccines: Bacille Calmette-Guérin (BCG), bivalent Oral Polio Vaccine (bOPV), Diphtheria Tetanus Pertussis, Hepatitis-B and Hib (DTP-HepB-Hib), measles-rubella (MR), Pneumococcal Conjugate Vaccine (PCV), and Rota vaccines. We evaluated annual and monthly vaccination trends using time-series and regression analyses. Predictive modeling was performed using an autoregressive integrated moving average (ARIMA) model. A total of 32,602,734 vaccination events were recorded across the regions from 2019 to 2022. Despite declining vaccination rates in 2020, there was a notable rebound in 2021, indicating the resilience of Tanzania's immunization program. The analysis also highlighted regional differences in vaccination rates when standardized per 1000 people. Seasonal fluctuations were observed in monthly vaccination rates, with BCG showing the most stable trend. Predictive modeling of BCG indicated stable and increasing vaccination coverage by 2023. These findings underscore the robustness of Tanzania's childhood immunization infrastructure in overcoming the challenges posed by the COVID-19 pandemic, as indicated by the strong recovery of vaccination rates post-2020. We provide valuable insights into the dynamics of vaccination during a global health crisis and highlight the importance of sustained immunization efforts to maintain public health.
Collapse
Affiliation(s)
- Raphael Zozimus Sangeda
- Department of Pharmaceutical Microbiology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Daniel James
- Department of Pharmaceutical Microbiology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Harrison Mariki
- Department of Research and Development, Afya Intelligence, Dar es Salaam, Tanzania
| | - Mbonea Erick Mbwambo
- Department of Research and Development, Afya Intelligence, Dar es Salaam, Tanzania
| | - Mwendwa E. Mwenesi
- Immunization and Vaccine Development, Ministry of Health Tanzania, Dodoma, Tanzania
| | - Honesti Nyaki
- Immunization and Vaccine Development, Ministry of Health Tanzania, Dodoma, Tanzania
| | - Florian Tinuga
- Immunization and Vaccine Development, Ministry of Health Tanzania, Dodoma, Tanzania
| | - Daudi Peter Manyanga
- Department of Universal Health Coverage, Communicable & Non-Communicable Diseases, World Health Organization Inter-Country Support Team for East and Southern African Countries, Harare, Zimbabwe
| |
Collapse
|
194
|
Jia X, Jing X, Li M, Gao M, Zhong Y, Li E, Liu Y, Li R, Yao G, Liu Q, Zhou M, Hou Y, An L, Hong Y, Li S, Zhang J, Wang W, Zhang K, Gong P, Chiu S. An adenosine analog shows high antiviral potency against coronavirus and arenavirus mainly through an unusual base pairing mode. Nat Commun 2024; 15:10750. [PMID: 39737930 DOI: 10.1038/s41467-024-54918-3] [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: 02/21/2024] [Accepted: 11/22/2024] [Indexed: 01/01/2025] Open
Abstract
By targeting the essential viral RNA-dependent RNA polymerase (RdRP), nucleoside analogs (NAs) have exhibited great potential in antiviral therapy for RNA virus-related diseases. However, most ribose-modified NAs do not present broad-spectrum features, likely due to differences in ribose-RdRP interactions across virus families. Here, we show that HNC-1664, an adenosine analog with modifications both in ribose and base, has broad-spectrum antiviral activity against positive-strand coronaviruses and negative-strand arenaviruses. Importantly, treatment with HNC-1664 demonstrate anti-SARS-CoV-2 efficacy in infected K18-human ACE2 mice, with reduced viral titer and mortality, as well as improved lung injury. Enzymology data demonstrate that HNC-1664 inhibits RNA synthesis mainly at the pre-catalysis stage. The cryo-EM structures of HNC-1664-bound RdRP-RNA complexes from both SARS-CoV-2 and LASV reveal an unusual base pairing mode of HNC-1664 in part due to its base modification, thus revealing its great potency in binding but not catalysis. Under certain circumstances, 1664-TP can be slowly incorporated by RdRP through regular Watson-Crick base pairing, as evidenced by enzymology data and an HNC-1664-incorporated crystal structure of the RdRP-RNA complex. Overall, HNC-1664 achieves broad-spectrum characteristics by favoring an alternative base pairing strategy to non-catalytically block RNA synthesis, providing a novel concept for the rational development of NA drugs.
Collapse
Affiliation(s)
- Xiaoying Jia
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, No. 262 Jin Long Street, Wuhan, Hubei, China
| | - Xuping Jing
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, No. 262 Jin Long Street, Wuhan, Hubei, China
| | - Ming Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Minli Gao
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yao Zhong
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, No. 262 Jin Long Street, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yang Liu
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, No. 262 Jin Long Street, Wuhan, Hubei, China
| | - Rui Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, No. 262 Jin Long Street, Wuhan, Hubei, China
| | - Guoqiang Yao
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qiaojie Liu
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, No. 262 Jin Long Street, Wuhan, Hubei, China
| | - Minmin Zhou
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, No. 262 Jin Long Street, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuxia Hou
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, No. 262 Jin Long Street, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Linfeng An
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yibao Hong
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shanshan Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jiancun Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
| | - Wei Wang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, No. 262 Jin Long Street, Wuhan, Hubei, China.
| | - Kaiming Zhang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- MOE Key Laboratory for Cellular Dynamics and Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Hefei, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui, China.
| | - Peng Gong
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, No. 262 Jin Long Street, Wuhan, Hubei, China.
- Drug Discovery Center for Infectious Diseases, Nankai University, Tianjin, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, Anhui, China.
| |
Collapse
|
195
|
Aynaud MM, Caldwell L, Al-Zahrani KN, Barutcu S, Chan K, Obersterescu A, Ogunjimi AA, Jin M, Zakoor KR, Patel S, Padilla R, Jen MCC, Veniegas PM, Dewsi N, Yonathan F, Zhang L, Ayson-Fortunato A, Aquino A, Krzyzanowski P, Simpson J, Bartlett J, Lungu I, Wouters BG, Rini JM, Gekas M, Poutanen S, Pelletier L, Mazzulli T, Wrana JL. Systematic surveillance of SARS-CoV-2 reveals dynamics of variant mutagenesis and transmission in a large urban population. Nat Commun 2024; 15:10795. [PMID: 39738001 DOI: 10.1038/s41467-024-55031-1] [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: 07/24/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
Highly mutable pathogens generate viral diversity that impacts virulence, transmissibility, treatment, and thwarts acquired immunity. We previously described C19-SPAR-Seq, a high-throughput, next-generation sequencing platform to detect SARS-CoV-2 that we here deployed to systematically profile variant dynamics of SARS-CoV-2 for over 3 years in a large, North American urban environment (Toronto, Canada). Sequencing of the ACE2 receptor binding motif and polybasic furin cleavage site of the Spike gene in over 70,000 patients revealed that population sweeps of canonical variants of concern (VOCs) occurred in repeating wavelets. Furthermore, we found that VOC mutant derivatives and putative quasispecies that targeted functionally important residues and were found in future VOCs arose frequently, but were always extinguished. Systematic screening of functionally relevant domains in pathogens could thus provide a powerful tool for monitoring spread and mutational trajectories, particularly those with zoonotic potential.
Collapse
Affiliation(s)
- Marie-Ming Aynaud
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Lauren Caldwell
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Khalid N Al-Zahrani
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Seda Barutcu
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Kin Chan
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
- The Network Biology Collaborative Centre (NBCC), Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Andreea Obersterescu
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Abiodun A Ogunjimi
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Min Jin
- Departments of Molecular Genetics and Biochemistry, University of Toronto, MaRS Center, Toronto, Ontario, M5G 1M1, Canada
| | - Kathleen-Rose Zakoor
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Shyam Patel
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
- Department of Molecular Genetics, Donnelly Centre, University of Toronto, Toronto, Ontario, M5S 3E1, Canada
| | - Ron Padilla
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Mark C C Jen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
- The Network Biology Collaborative Centre (NBCC), Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Princess Mae Veniegas
- Department of Microbiology, Mount Sinai Hospital/University Health Network, Toronto, Ontario, M5G 1X5, Canada
| | - Nursrin Dewsi
- Department of Microbiology, Mount Sinai Hospital/University Health Network, Toronto, Ontario, M5G 1X5, Canada
| | - Filiam Yonathan
- Department of Microbiology, Mount Sinai Hospital/University Health Network, Toronto, Ontario, M5G 1X5, Canada
| | - Lucy Zhang
- Department of Microbiology, Mount Sinai Hospital/University Health Network, Toronto, Ontario, M5G 1X5, Canada
| | - Amelia Ayson-Fortunato
- Department of Microbiology, Mount Sinai Hospital/University Health Network, Toronto, Ontario, M5G 1X5, Canada
| | - Analiza Aquino
- Department of Microbiology, Mount Sinai Hospital/University Health Network, Toronto, Ontario, M5G 1X5, Canada
| | - Paul Krzyzanowski
- Ontario Institute for Cancer Research, Toronto General Hospital, Toronto, Ontario, M5G 0A3, Canada
| | - Jared Simpson
- Ontario Institute for Cancer Research, Toronto General Hospital, Toronto, Ontario, M5G 0A3, Canada
| | - John Bartlett
- Ontario Institute for Cancer Research, Toronto General Hospital, Toronto, Ontario, M5G 0A3, Canada
| | - Ilinca Lungu
- Ontario Institute for Cancer Research, Toronto General Hospital, Toronto, Ontario, M5G 0A3, Canada
| | - Bradly G Wouters
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Ontario, M5G 2C4, Canada
| | - James M Rini
- Departments of Molecular Genetics and Biochemistry, University of Toronto, MaRS Center, Toronto, Ontario, M5G 1M1, Canada
| | - Michael Gekas
- Department of Microbiology, Mount Sinai Hospital/University Health Network, Toronto, Ontario, M5G 1X5, Canada
| | - Susan Poutanen
- Department of Microbiology, Mount Sinai Hospital/University Health Network, Toronto, Ontario, M5G 1X5, Canada
| | - Laurence Pelletier
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
- The Network Biology Collaborative Centre (NBCC), Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
- Departments of Molecular Genetics and Biochemistry, University of Toronto, MaRS Center, Toronto, Ontario, M5G 1M1, Canada
| | - Tony Mazzulli
- Department of Microbiology, Mount Sinai Hospital/University Health Network, Toronto, Ontario, M5G 1X5, Canada
- Princess Margaret Cancer Centre and Campbell Family Institute for Cancer Research, University Health Network, Toronto, Ontario, M5G 2C4, Canada
| | - Jeffrey L Wrana
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada.
- The Network Biology Collaborative Centre (NBCC), Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada.
- Departments of Molecular Genetics and Biochemistry, University of Toronto, MaRS Center, Toronto, Ontario, M5G 1M1, Canada.
| |
Collapse
|
196
|
Latifi A, Flegr J. Persistent Health and Cognitive Impairments up to Four Years Post-COVID-19 in Young Students: The Impact of Virus Variants and Vaccination Timing. Biomedicines 2024; 13:69. [PMID: 39857653 PMCID: PMC11760454 DOI: 10.3390/biomedicines13010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025] Open
Abstract
Background: The long-term consequences of COVID-19 infection are becoming increasingly evident in recent studies. This repeated cross-sectional study aimed to explore the long-term health and cognitive effects of COVID-19, focusing on how virus variants, vaccination, illness severity, and time since infection impact post-COVID-19 outcomes. Methods: We examined three cohorts of university students (N = 584) and used non-parametric methods to assess correlations of various health and cognitive variables with SARS-CoV-2 infection, COVID-19 severity, vaccination status, time since infection, time since vaccination, and virus variants. Results: Our results suggest that some health and cognitive impairments may persist, with some even appearing to progressively worsen-particularly fatigue in women and memory in men-up to four years post-infection. The data further indicate that the ancestral SARS-CoV-2 variant may have the most significant long-term impact, while the Omicron variant appears to have the least. Interestingly, the severity of the acute illness was not correlated with the variant of SARS-CoV-2. The analysis also revealed that individuals who contracted COVID-19 after vaccination had better health and cognitive outcomes compared to those infected before vaccination. Conclusions: Overall, our results indicate that even in young individuals who predominantly experienced only mild forms of the infection, a gradual decline in health and fitness can occur over a span of four years post-infection. Notably, some negative trends-at least in men-only began to stabilize or even reverse during the fourth year, whereas in women, these trends showed no such improvement. These findings suggest that the long-term public health impacts of COVID-19 may be more severe and affect a much broader population than is commonly assumed.
Collapse
Affiliation(s)
| | - Jaroslav Flegr
- Laboratory of Evolutionary Biology, Department of Philosophy and History of Sciences, Faculty of Science, Charles University, Viničná 7, 128 00 Prague, Czech Republic;
| |
Collapse
|
197
|
Yang QW, Yue CL, Chen M, Ling YY, Dong Q, Zhou YX, Cao Y, Ding YX, Zhao X, Huang H, Zhang ZH, Hu L, Xu XH. Daphnetin may protect from SARS-CoV-2 infection by reducing ACE2. Sci Rep 2024; 14:30682. [PMID: 39730426 DOI: 10.1038/s41598-024-79734-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/12/2024] [Indexed: 12/29/2024] Open
Abstract
To combat the SARS-CoV-2 pandemic, innovative prevention strategies are needed, including reducing ACE2 expression on respiratory cells. This study screened approved drugs in China for their ability to downregulate ACE2. Daphnetin (DAP) was found to significantly reduce ACE2 mRNA and protein levels in PC9 cells. DAP exerts its inhibitory effects on ACE2 expression by targeting HIF-1α and JAK2, thereby impeding the transcription of the ACE2 gene. The SARS-CoV-2 pseudovirus infection assay confirmed that DAP-treated PC9 cells exhibited decreased susceptibility to viral infection. At therapeutic doses, DAP effectively lowers ACE2 expression in the respiratory systems of mice and humans. This suggests that DAP, already approved for other conditions, could be a new preventive measure against SARS-CoV-2, offering a cost-effective and accessible way to reduce SARS-CoV-2 spread.
Collapse
Affiliation(s)
- Qian-Wen Yang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Chang-Ling Yue
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Meng Chen
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China
| | - Yun-Yun Ling
- Department of Chemistry, Wannan Medical College, Wuhu, 241002, China
| | - Qi Dong
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Ying-Xin Zhou
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China
| | - Yin Cao
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China
| | - Yan-Xia Ding
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Xu Zhao
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China
| | - Hai Huang
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Zhao-Huan Zhang
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China.
| | - Lei Hu
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China.
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China.
| | - Xiao-Hui Xu
- School of Preclinical Medicine, Wannan Medical College, Wuhu, 241002, China.
- Anhui Province Key Laboratory of Basic Research and Transformation of Age- related Diseases, Wannan Medical College, Wuhu, 241002, China.
| |
Collapse
|
198
|
Mohammadshahi J, Ghobadi H, Shargi A, Moradkhani H, Rezaei H, Kazemy M, Aslani MR. Neutrophil-to-Lymphocyte and Platelet Ratio (N/LP Ratio), a Reliable Criterion for Predicting In-Hospital Mortality in Both Genders Infected With SARS-CoV-2. Mediators Inflamm 2024; 2024:5720709. [PMID: 39759900 PMCID: PMC11699982 DOI: 10.1155/mi/5720709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Background: The neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), platelet-to-lymphocyte ratio (PLR), derived neutrophil-to-lymphocyte ratio (dNLR), neutrophil-to-lymphocyte and platelet ratio (N/LP ratio), aggregate index of systemic inflammation (AISI), systemic inflammation response index (SIRI), and systemic inflammation index (SII) have emerged as noteworthy determinants in evaluating the severity and mortality prognosis of inflammatory diseases. In order to predict mortality rate, this study aimed to assess the impact of systemic inflammatory markers on both men and women who were admitted to the hospital due to SARS-CoV-2 infection. Methods: The laboratory parameters of the 2007 COVID-19 patients were analyzed in a retrospective study (men = 1145 and women = 862). Receiver operating characteristic (ROC) analysis was used to determine the capability of inflammatory markers to differentiate the severity of COVID-19, while survival probability was determined using Kaplan-Meier curves, with the endpoint being death. To prevent any linear bias, the inflammatory indices were assessed separately using univariate analysis for Charlson comorbidity index (CCI), and adjustments were made for confounding factors if p < 0.2. Results: Adjusted-NLR, adjusted-MLR, N/LP ratio, adjusted-dNLR, adjusted-AISI, adjusted-SII, and adjusted-SIRI exhibited remarkably higher values in patients who did not survive as compared to those who did. The multivariate Cox regression models demonstrated significant association between survival and N/LP ratio (HR = 1.564, 95% CI = 1.161 to 2.107, p < 0.01) in men and N/LP ratio (HR = 1.745, 95% CI = 1.230 to 2.477, p < 0.01) and adjusted-SII (HR = 6.855, 95% CI = 1.454 to 32.321, p < 0.05) in women. Conclusion: A reliable predictor in the current study of men and women with COVID-19 was N/LP ratio.
Collapse
Affiliation(s)
- Jafar Mohammadshahi
- Department of Infectious Diseases and Tropical Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hassan Ghobadi
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Afshan Shargi
- Department of Community Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hossein Moradkhani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamed Rezaei
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mahur Kazemy
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Reza Aslani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
199
|
Angius F, Puxeddu S, Zaimi S, Canton S, Nematollahzadeh S, Pibiri A, Delogu I, Alvisi G, Moi ML, Manzin A. SARS-CoV-2 Evolution: Implications for Diagnosis, Treatment, Vaccine Effectiveness and Development. Vaccines (Basel) 2024; 13:17. [PMID: 39852796 PMCID: PMC11769326 DOI: 10.3390/vaccines13010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
The COVID-19 pandemic, driven by the rapid evolution of the SARS-CoV-2 virus, presents ongoing challenges to global public health. SARS-CoV-2 is characterized by rapidly evolving mutations, especially in (but not limited to) the spike protein, complicating predictions about its evolutionary trajectory. These mutations have significantly affected transmissibility, immune evasion, and vaccine efficacy, leading to multiple pandemic waves with over half a billion cases and seven million deaths globally. Despite several strategies, from rapid vaccine development and administration to the design and availability of antivirals, including monoclonal antibodies, already having been employed, the persistent circulation of the virus and the emergence of new variants continue to result in high case numbers and fatalities. In the past four years, immense research efforts have contributed much to our understanding of the viral pathogenesis mechanism, the COVID-19 syndrome, and the host-microbe interactions, leading to the development of effective vaccines, diagnostic tools, and treatments. The focus of this review is to provide a comprehensive analysis of the functional impact of mutations on diagnosis, treatments, and vaccine effectiveness. We further discuss vaccine safety in pregnancy and the implications of hybrid immunity on long-term protection against infection, as well as the latest developments on a pan-coronavirus vaccine and nasal formulations, emphasizing the need for continued surveillance, research, and adaptive public health strategies in response to the ongoing SARS-CoV-2 evolution race.
Collapse
Affiliation(s)
- Fabrizio Angius
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvia Puxeddu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Silvio Zaimi
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Serena Canton
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Sepehr Nematollahzadeh
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Andrea Pibiri
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Ilenia Delogu
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| | - Gualtiero Alvisi
- Department of Molecular Medicine, University of Padova, 35121 Padova, Italy; (S.N.); (G.A.)
| | - Meng Ling Moi
- School of International Health, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Aldo Manzin
- Microbiology and Virology Unit, Department of Biomedical Sciences, University of Cagliari, University Campus, 09042 Monserrato, Italy (A.P.); (I.D.); (A.M.)
| |
Collapse
|
200
|
Qu H, Wang S, He M, Wu Y, Yan F, Liu T, Zhang M. Is it feasible to use AI-based drug design methods in the process of generating effective COVID-19 inhibitors? A validation study using molecular docking, molecular simulation, and pharmacophore methods. J Biomol Struct Dyn 2024:1-14. [PMID: 39727340 DOI: 10.1080/07391102.2024.2445169] [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: 03/05/2024] [Accepted: 07/21/2024] [Indexed: 12/28/2024]
Abstract
Although the COVID-19 pandemic has been brought under control to some extent globally, there is still debate in the industry about the feasibility of using artificial intelligence (AI) to generate COVID small-molecule inhibitors. In this study, we explored the feasibility of using AI to design effective inhibitors of COVID-19. By combining a generative model with reinforcement learning and molecular docking, we designed small-molecule inhibitors targeting the COVID-19 3CLpro enzyme. After screening based on molecular docking scores and physicochemical properties, we obtained five candidate inhibitors. Furthermore, theoretical calculations confirmed that these candidate inhibitors have significant binding stability with COVID-19 3CLpro, comparable to or better than existing COVID-19 inhibitors. Additionally, through ligand-based pharmacophore model screening, we validated the effectiveness of the generative model, demonstrating the potential value of AI in drug design.
Collapse
Affiliation(s)
- Hanyang Qu
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Shengpeng Wang
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Mingyang He
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Yuhui Wu
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Fei Yan
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Tiaotiao Liu
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Meiling Zhang
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| |
Collapse
|