1
|
Zhou S, Hui X, Wang W, Zhao C, Jin M, Qin Y, Chen M. SARS-CoV-2 and HCoV-OC43 regulate host m6A modification via activation of the mTORC1 signalling pathway to facilitate viral replication. Emerg Microbes Infect 2025; 14:2447620. [PMID: 39745173 PMCID: PMC11852242 DOI: 10.1080/22221751.2024.2447620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/08/2024] [Accepted: 12/22/2024] [Indexed: 02/25/2025]
Abstract
N6-methyladenosine (m6A) is the most prevalent post-transcriptional modification in eukaryotic RNA and is also present in various viral RNAs, where it plays a crucial role in regulating the viral life cycle. However, the molecular mechanisms through which viruses regulate host RNA m6A methylation are not fully understood. In this study, we reveal that SARS-CoV-2 and HCoV-OC43 infection enhance host m6A modification by activating the mTORC1 signalling pathway. Specifically, the viral non-structural protein nsp14 upregulates the expression of S-adenosylmethionine synthase MAT2A in an mTORC1-dependent manner. This mTORC1-MAT2A axis subsequently stimulates the synthesis of S-adenosylmethionine (SAM). The increase of SAM then enhances the m6A methylation of host RNA and facilitates viral replication. Our findings uncover a molecular mechanism by which viruses regulate host m6A methylation and provide insights into how SARS-CoV-2 hijacks host cellular epitranscriptomic modifications to promote its replication.
Collapse
Affiliation(s)
- Shixiong Zhou
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
| | - Xianfeng Hui
- National key laboratory of agricultural microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Weiwei Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
| | - Chunbei Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
| | - Meilin Jin
- National key laboratory of agricultural microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Yali Qin
- School of Life Sciences, Hubei University, Wuhan, People’s Republic of China
| | - Mingzhou Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
- School of Life Sciences, Hubei University, Wuhan, People’s Republic of China
| |
Collapse
|
2
|
Maziec K, Baliga-Gil A, Kierzek E. Delivery strategies for RNA-targeting therapeutic nucleic acids and RNA-based vaccines against respiratory RNA viruses: IAV, SARS-CoV-2, RSV. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102572. [PMID: 40529300 PMCID: PMC12173128 DOI: 10.1016/j.omtn.2025.102572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/20/2025]
Abstract
Therapeutic nucleic acids, including small interfering RNA (siRNA), and antisense oligonucleotides (ASOs), targeting RNA viruses such as influenza A virus (IAV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and respiratory syncytial virus (RSV), play a crucial role in contemporary medicine. The primary goal of short oligonucleotide-based antivirals is to precisely inhibit viral mechanisms by interacting with viral RNA, thereby opening new avenues for infection treatment. RNA recently was also used to invent mRNA vaccine for different illness prevention. Therapeutic nucleic acids and mRNA vaccine attracted considerable attention during the COVID-19 pandemic due to the pressing necessity to develop an effective strategy to address this global threat. In addition to the advancement of therapeutic nucleic acids aimed at targeting respiratory viruses, the effective delivery of these molecules to infected cells is of paramount importance. Similarly, mRNA vaccine's effectiveness also depends on effective delivery. This article offers a comprehensive summary and analysis of various delivery strategies, along with the challenges encountered in their development. Representative studies conducted in cellular models, model organisms, and human are presented for examination. Furthermore, the article explores future perspectives regarding the delivery of therapeutic nucleic acids and mRNA vaccines aimed at combating IAV, SARS-CoV-2, and RSV.
Collapse
Affiliation(s)
- Kinga Maziec
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Agnieszka Baliga-Gil
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Elzbieta Kierzek
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| |
Collapse
|
3
|
Chung J, Pierce J, Franklin C, Olson RM, Morrison AR, Amos-Landgraf J. Translating animal models of SARS-CoV-2 infection to vascular, neurological and gastrointestinal manifestations of COVID-19. Dis Model Mech 2025; 18:dmm052086. [PMID: 40195851 PMCID: PMC12010913 DOI: 10.1242/dmm.052086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
Since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) initiated a global pandemic resulting in an estimated 775 million infections with over 7 million deaths, it has become evident that COVID-19 is not solely a pulmonary disease. Emerging evidence has shown that, in a subset of patients, certain symptoms - including chest pain, stroke, anosmia, dysgeusia, diarrhea and abdominal pain - all indicate a role of vascular, neurological and gastrointestinal (GI) pathology in the disease process. Many of these disease processes persist long after the acute disease has been resolved, resulting in 'long COVID' or post-acute sequelae of COVID-19 (PASC). The molecular mechanisms underlying the acute and systemic conditions associated with COVID-19 remain incompletely defined. Appropriate animal models provide a method of understanding underlying disease mechanisms at the system level through the study of disease progression, tissue pathology, immune system response to the pathogen and behavioral responses. However, very few studies have addressed PASC and whether existing models hold promise for studying this challenging problem. Here, we review the current literature on cardiovascular, neurological and GI pathobiology caused by COVID-19 in patients, along with established animal models of the acute disease manifestations and their prospects for use in PASC studies. Our aim is to provide guidance for the selection of appropriate models in order to recapitulate certain aspects of the disease to enhance the translatability of mechanistic studies.
Collapse
Affiliation(s)
- James Chung
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Julia Pierce
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Department of Research, Ocean State Research Institute, Inc., Providence, RI 02908-4734, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02908, USA
| | - Craig Franklin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| | - Rachel M. Olson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
- Laboratory for Infectious Disease Research, University of Missouri, Columbia, MO 65211, USA
| | - Alan R. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, USA
- Department of Research, Ocean State Research Institute, Inc., Providence, RI 02908-4734, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, RI 02908, USA
| | - James Amos-Landgraf
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
4
|
Zhi W, Wang L, Dai L, Xu J, He T, Zong X, Xu J, Cai H, Pi J, Sun P, Chen S, Huang X, Zhou H. SERS-based lateral flow immunoassay for rapid and sensitive sensing of nucleocapsid protein toward SARS-CoV-2 screening in clinical samples. Anal Chim Acta 2025; 1360:344149. [PMID: 40409906 DOI: 10.1016/j.aca.2025.344149] [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: 12/30/2024] [Revised: 05/01/2025] [Accepted: 05/02/2025] [Indexed: 05/25/2025]
Abstract
Early and accurate identification of SARS-CoV-2 infection is crucial for epidemic prevention and control. Lateral flow immunoassay (LFIA) has become the mainstream method for screening SARS-CoV-2 infection due to its rapid, simple and amenable for point-of-care detection (POCT), but still suffered from the poor sensitivity and accuracy. In this study, Au nanoparticles (NPs) with controllable Ag shell (Au@Ag) were manufactured via a seed-mediated growth method. The Au@Ag-based LFIA exhibited superb colorimetric (CM) signal and intense surface-enhanced Raman scattering (SERS) signal for dual-mode sensing of nucleocapsid protein (N protein), a naturally protein expression in vivo during SARS-CoV-2 infection. The limit of detection (LOD) of the SERS-LFIA mode was 2.16 pg/mL, which was around 150-time more sensitive than conventional visual CM-LFIA mode (300 pg/mL). More importantly, the proposed LFIA is capable of quantitatively detecting N protein-spiked real samples with satisfactory recoveries from 83 % to 91.4 %. Clinical pharyngeal swab samples of the infected patients (n = 20) and healthy subjects (n = 20) were effectively discriminated in the developed SERS-LFIA, where the negative accuracy rate was 100 % and the positive accuracy rate was 85 %, among which samples from P1, P18, and P19 were false-negative results. The results obtained from the LFIA immunoassay were in good agreement with the standard PCR method in clinic, and superior to those of the commercially colloidal gold strip by using the same antibodies. In conclusion, the LFIA proposed here can perform specific, rapid, and ultrasensitive analysis of N protein toward early warning of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Weixia Zhi
- The Second Clinical Medical College (Shenzhen People's Hospital), The Fifth Affiliated Hospital, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Lingwei Wang
- The Second Clinical Medical College (Shenzhen People's Hospital), The Fifth Affiliated Hospital, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Li Dai
- The Second Clinical Medical College (Shenzhen People's Hospital), The Fifth Affiliated Hospital, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Jing Xu
- The Second Clinical Medical College (Shenzhen People's Hospital), The Fifth Affiliated Hospital, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Tingting He
- The Second Clinical Medical College (Shenzhen People's Hospital), The Fifth Affiliated Hospital, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Xiangxin Zong
- The Second Clinical Medical College (Shenzhen People's Hospital), The Fifth Affiliated Hospital, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Jun Xu
- The Second Clinical Medical College (Shenzhen People's Hospital), The Fifth Affiliated Hospital, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Huaihong Cai
- College of Chemistry and Materials Science, Jinan University, Guangzhou, 510632, China
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, 523000, China
| | - Pinghua Sun
- The Second Clinical Medical College (Shenzhen People's Hospital), The Fifth Affiliated Hospital, College of Pharmacy, Jinan University, Guangzhou, 510632, China; Institute for Safflower Industry Research, Key Laboratory of Xinjiang Phytomedicine Resource and Utilization (Ministry of Education), School of Pharmacy, Shihezi University, Shihezi, 832003, China
| | - Shanze Chen
- The Second Clinical Medical College (Shenzhen People's Hospital), The Fifth Affiliated Hospital, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Xueqin Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, 523000, China.
| | - Haibo Zhou
- The Second Clinical Medical College (Shenzhen People's Hospital), The Fifth Affiliated Hospital, College of Pharmacy, Jinan University, Guangzhou, 510632, China; Institute for Safflower Industry Research, Key Laboratory of Xinjiang Phytomedicine Resource and Utilization (Ministry of Education), School of Pharmacy, Shihezi University, Shihezi, 832003, China.
| |
Collapse
|
5
|
Karhan C, Sake SM, Gunesch AP, Grethe C, Hellwinkel B, Köhler NM, Kiefer AF, Hapko U, Kany AM, Pietschmann T, Hirsch AKH. Unlocking the antiviral arsenal: Structure-guided optimization of small-molecule inhibitors against RSV and hCoV-229E. Eur J Med Chem 2025; 291:117282. [PMID: 40199027 DOI: 10.1016/j.ejmech.2025.117282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/20/2024] [Accepted: 01/13/2025] [Indexed: 04/10/2025]
Abstract
Acute respiratory diseases in humans can be caused by various viral pathogens such as respiratory syncytial virus (RSV), human coronavirus 229E (hCoV-229E), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). To prevent severe cases by an early treatment, one effective strategy is to inhibit viral infection at the entry stage of the replication cycle. However, there is a lack of efficient, FDA-approved small-molecule drugs targeting these pathogens. Previously, we identified two dual RSV/hCoV-229E small-molecule inhibitors with activity in the single-digit micromolar range. In this study, we focused on a structure-guided optimization approach of the more promising prototype addressing activity, cell viability, selectivity, solubility and metabolic stability. We present valuable insights into the structure-activity relationship (SAR), and report the discovery of a sub-micromolar RSV entry inhibitor, a dual RSV/CoV-229E inhibitor and a highly potent compound against hCoV-229E.
Collapse
Affiliation(s)
- Christina Karhan
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany; Saarland University, Department of Pharmacy, Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Svenja M Sake
- TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Helmholtz Centre for Infection Research and the Hannover Medical School, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Antonia P Gunesch
- TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Helmholtz Centre for Infection Research and the Hannover Medical School, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Christina Grethe
- TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Helmholtz Centre for Infection Research and the Hannover Medical School, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Benedikt Hellwinkel
- TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Helmholtz Centre for Infection Research and the Hannover Medical School, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Natalie M Köhler
- TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Helmholtz Centre for Infection Research and the Hannover Medical School, Feodor-Lynen-Str. 7, 30625, Hannover, Germany
| | - Alexander F Kiefer
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Uladzislau Hapko
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany; Saarland University, Department of Pharmacy, Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Andreas M Kany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Thomas Pietschmann
- TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Helmholtz Centre for Infection Research and the Hannover Medical School, Feodor-Lynen-Str. 7, 30625, Hannover, Germany; German Centre for Infection Research (DZIF), Inhoffenstr. 7, 38124, Braunschweig, Germany; Helmholtz International Lab for Anti-Infectives, Campus E8.1, 66123, Saarbrücken, Germany; Cluster of Excellence RESIST (EXC 2155), Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus Building E8.1, 66123, Saarbrücken, Germany; Saarland University, Department of Pharmacy, Campus Building E8.1, 66123, Saarbrücken, Germany; Helmholtz International Lab for Anti-Infectives, Campus E8.1, 66123, Saarbrücken, Germany; Cluster of Excellence RESIST (EXC 2155), Hanover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| |
Collapse
|
6
|
Puglia I, Caporale M, Di Teodoro G, Spedicato M, Profeta F, Marcacci M, Di Pancrazio C, Valleriani F, Rossi E, Auerswald H, Lorusso A. Optimization of an infectious subgenomic amplicons reverse genetics protocol for the rescue of synthetic coronaviruses. J Virol Methods 2025; 336:115152. [PMID: 40188879 DOI: 10.1016/j.jviromet.2025.115152] [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: 07/24/2023] [Revised: 03/18/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025]
Abstract
Reverse genetics (rg) systems are indispensable tools for investigating the pathogenesis of RNA viruses, facilitating vaccine design, and advancing antiviral therapeutic strategies. In this study, we optimized the Infectious Subgenomic Amplicons (ISA) method for generating synthetic r-wt SARS-CoV-2 Wuhan-Hu-1. This system was validated by demonstrating the successful rescue of infectious viral particles from overlapping DNA fragments and their propagation in vitro. Sequencing confirmed 100 % identity of the recovered virus with the Wuhan-Hu-1 reference genome. Importantly, in vivo experiments using K18-hACE2 mice revealed that the r-wt SARS-CoV-2 Wuhan-Hu-1 strain caused clinical symptoms, weight loss, and mortality comparable to those induced by a virulent SARS-CoV-2 field variant. This ISA rg method offers a rapid and reproducible approach to generating synthetic coronaviruses, with potential applications in pathogenesis studies, antiviral testing, and vaccine development.
Collapse
Affiliation(s)
- Ilaria Puglia
- PhD National Programme in One Health approaches to infectious diseases and life science research, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Italy; Istituto Zooprofilattico Sperimentale dell' Abruzzo e Molise, Teramo, Italy
| | - Marialuigia Caporale
- PhD National Programme in One Health approaches to infectious diseases and life science research, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Italy; Istituto Zooprofilattico Sperimentale dell' Abruzzo e Molise, Teramo, Italy
| | | | - Massimo Spedicato
- Istituto Zooprofilattico Sperimentale dell' Abruzzo e Molise, Teramo, Italy
| | - Francesca Profeta
- Istituto Zooprofilattico Sperimentale dell' Abruzzo e Molise, Teramo, Italy
| | - Maurilia Marcacci
- Istituto Zooprofilattico Sperimentale dell' Abruzzo e Molise, Teramo, Italy
| | | | | | - Emanuela Rossi
- Istituto Zooprofilattico Sperimentale dell' Abruzzo e Molise, Teramo, Italy
| | - Heidi Auerswald
- Istituto Zooprofilattico Sperimentale dell' Abruzzo e Molise, Teramo, Italy
| | - Alessio Lorusso
- Istituto Zooprofilattico Sperimentale dell' Abruzzo e Molise, Teramo, Italy.
| |
Collapse
|
7
|
Alves MCS, da Silva RCC, de Leitão-Júnior SSP, de Balbino VQ. Therapeutic Approaches for COVID-19: A Review of Antiviral Treatments, Immunotherapies, and Emerging Interventions. Adv Ther 2025; 42:3045-3058. [PMID: 40338485 DOI: 10.1007/s12325-025-03218-3] [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: 03/19/2025] [Accepted: 04/22/2025] [Indexed: 05/09/2025]
Abstract
The coronavirus disease 2019 (COVID-19) global health crisis, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has presented unprecedented challenges to global healthcare systems, leading to rapid advances in treatment development. This review comprehensively examines the current therapeutic approaches for managing COVID-19, including direct-acting antivirals, immunomodulators, anticoagulants, and adjuvant therapies, as well as emerging and experimental approaches. Direct-acting antivirals target various stages of the viral life cycle, offering specific intervention points, while immunomodulators aim to modulate the host's immune response, reducing disease severity. Anticoagulant therapies address the coagulopathy frequently observed in severe cases, and adjuvant treatments provide supportive care to improve overall outcomes. We also explore the challenges and limitations of implementing these treatments, such as drug resistance, variable patient responses, and access to therapies, especially in resource-limited settings. The review also discusses future perspectives, including the potential of next-generation vaccines, personalized medicine, and global collaboration in shaping future COVID-19 treatment paradigms. Continuous innovation, combined with an integrated and adaptable approach, will be crucial to effectively managing COVID-19 and mitigating the impact of future pandemics.
Collapse
Affiliation(s)
- Maria C S Alves
- Laboratory of Bioinformatics and Evolutionary Biology, Center for Biosciences, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, 50670-423, Brazil.
| | - Ruana C C da Silva
- Laboratory of Health Sciences Research, Federal University of Grande Dourados, Dourados, Mato Grosso do Sul, 79825-070, Brazil
| | - Sérgio S P de Leitão-Júnior
- Laboratory of Bioinformatics and Evolutionary Biology, Center for Biosciences, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, 50670-423, Brazil
- Serra Talhada Academic Unit, Federal Rural University of Pernambuco, Serra Talhada, Pernambuco, 56909-535, Brazil
| | - Valdir Q de Balbino
- Laboratory of Bioinformatics and Evolutionary Biology, Center for Biosciences, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, 50670-423, Brazil.
| |
Collapse
|
8
|
Kim M, Lee MK, Jo I, Jang Y, Han SB, Lee J, Yang A, Jarhad DB, Choi H, Nogi Y, Saito-Tarashima N, Minakawa N, Kim M, Jeong LS. Identification of 4'-Thiouridine as an Orally Available Antiviral Agent Targeting Both RdRp and NiRAN Functions of SARS-CoV-2 Nsp12. J Med Chem 2025; 68:12414-12433. [PMID: 40512093 DOI: 10.1021/acs.jmedchem.4c02874] [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: 06/28/2025]
Abstract
Through screening of a nucleos(t)ide-focused chemical library, we identified 4'-thiouridine (1) as a potent antiviral agent against SARS-CoV-2 in Vero cells, with an EC50 value of 1.71 μM and a CC50 value exceeding 100 μM. Its triphosphate metabolite, compound 8, inhibited the RNA-dependent RNA polymerase activity of the SARS-CoV-2 Nsp12-Nsp7-Nsp82 complex, terminating nascent RNA synthesis through misincorporation. Additionally, compound 8 suppressed the function of the NiRAN domain of Nsp12, effectively blocking both RNAylation and NMPylation of Nsp9. Pharmacokinetic analysis in mice showed excellent oral bioavailability of compound 1. Oral administration at 100 mg/kg/day, twice daily for 5 days, protected mice from lethal SARS-CoV-2 infection, resulting in 40% survival and near-complete recovery of body weight by day 14 postinfection. Compound 1 also exhibited broad-spectrum activity against various coronaviruses and other RNA viruses. These findings highlight that compound 1 is a promising orally available antiviral candidate.
Collapse
Affiliation(s)
- Minjae Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Myoung Kyu Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Inseong Jo
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Yejin Jang
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Soo Bong Han
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
- Department of Medicinal Chemistry and Pharmacology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Juyeon Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Ayeon Yang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Dnyandev B Jarhad
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Hongseok Choi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Yuhei Nogi
- Graduate School of Pharmaceutical Science, Tokushima University, Tokushima 770-8505, Japan
| | - Noriko Saito-Tarashima
- Graduate School of Pharmaceutical Science, Tokushima University, Tokushima 770-8505, Japan
| | - Noriaki Minakawa
- Graduate School of Pharmaceutical Science, Tokushima University, Tokushima 770-8505, Japan
| | - Meehyein Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Lak Shin Jeong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- Future Medicine Co., Ltd, 54 Changup-ro, Sujeong-gu, Seongnam, Gyeonggi-do 13449, Republic of Korea
| |
Collapse
|
9
|
Mehyar N, Samman N, Al Gheribi S, Mashhour A, Chan P, Al-Kaysi RO, Perlman S, Boudjelal M, Islam I. First-in-class inhibitors of Nsp15 endoribonuclease of SARS-CoV-2: modeling, synthesis, and enzymatic assay of thiazolidinedione and rhodanine analogs. J Biol Chem 2025:110409. [PMID: 40562099 DOI: 10.1016/j.jbc.2025.110409] [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: 01/16/2025] [Revised: 06/06/2025] [Accepted: 06/18/2025] [Indexed: 06/28/2025] Open
Abstract
During infection, the coronavirus Nsp15, a uridine-specific endoribonuclease, suppresses the host cell's antiviral response. Recently, researchers have paid more attention to this relatively underexplored yet potentially viable drug target. In this study, we employed fluorescence resonance energy transfer-based screening assays to identify potent Nsp15 inhibitors. Subsequently, we used active-site in silico docking methods to design new molecules with enhanced inhibitory properties. Solution assays were used to measure the potency and determine the mechanism of these inhibitors. We identified a novel class of thiazolidinedione and rhodanine analogs that inhibit SARS-CoV-2 Nsp15. Docking these compounds into the uridine-binding site shows that most analogs form two hydrogen bonds with Ser294. The most potent inhibitors are compounds KCO237 and KCO251 (half-maximal inhibitory concentration: 0.304 μM, 0.931 μM respectively). The inhibition kinetics of KCO237 and KCO251 best align with a reversible mixed inhibition model. Mutating Ser294 did not completely abolish Nsp15 activity or the inhibitory effect of KCO237 or KCO251. These findings suggest that thiazolidinedione and rhodanine analogs likely inhibit Nsp15 by binding to the uridine active site while also implicating a possible secondary allosteric binding site. The ability of these compounds to inhibit VERO 6 cell infection with SARS-CoV-2 at subtoxic levels highlights their potential for development as novel antiviral treatments for SARS-CoV-2 and other coronavirus-related diseases.
Collapse
Affiliation(s)
- Nimer Mehyar
- King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs, Riyadh, 11481, Saudi Arabia; King Abdullah International Medical Research Center, Ministry of National Guards-Health Affairs, Riyadh, 11481, Saudi Arabia; King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, 11481, Saudi Arabia.
| | - Nosaibah Samman
- King Abdullah International Medical Research Center, Ministry of National Guards-Health Affairs, Riyadh, 11481, Saudi Arabia; King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, 11481, Saudi Arabia
| | - Shatha Al Gheribi
- King Abdullah International Medical Research Center, Ministry of National Guards-Health Affairs, Riyadh, 11481, Saudi Arabia; King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, 11481, Saudi Arabia
| | - Abdullah Mashhour
- King Abdullah International Medical Research Center, Ministry of National Guards-Health Affairs, Riyadh, 11481, Saudi Arabia; King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, 11481, Saudi Arabia
| | - Pearl Chan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, 52242, IA, USA
| | - Rabih O Al-Kaysi
- King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard-Health Affairs, Riyadh, 11481, Saudi Arabia; King Abdullah International Medical Research Center, Ministry of National Guards-Health Affairs, Riyadh, 11481, Saudi Arabia; King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, 11481, Saudi Arabia
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, 52242, IA, USA
| | - Mohamed Boudjelal
- King Abdullah International Medical Research Center, Ministry of National Guards-Health Affairs, Riyadh, 11481, Saudi Arabia; King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, 11481, Saudi Arabia
| | - Imadul Islam
- King Abdullah International Medical Research Center, Ministry of National Guards-Health Affairs, Riyadh, 11481, Saudi Arabia; King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, 11481, Saudi Arabia
| |
Collapse
|
10
|
Argyrou M, Pitsillou E, Hung A, El-Osta A, Karagiannis TC. Insights into the pathogenic mechanisms associated with the SARS-CoV-2 spike protein. J Struct Biol 2025; 217:108229. [PMID: 40562255 DOI: 10.1016/j.jsb.2025.108229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/25/2025] [Accepted: 06/23/2025] [Indexed: 06/28/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pathogenic agent responsible for the coronavirus disease 2019 (COVID-19) pandemic, uses the trimeric spike protein to gain entry into the host cell. Structural studies have revealed that the spike protein is comprised of the S1 and S2 subunits. The S1 subunit of the spike protein contains the receptor-binding domain (RBD), which binds to the human angiotensin-converting enzyme 2 (ACE2) receptor. The interaction between the RBD and ACE2 facilitates membrane fusion and host cell infection. The SARS-CoV-2 spike protein also contains a unique insertion of four amino acids that results in the 682-RRAR↓S-686 polybasic furin cleavage motif at the boundary of the S1 and S2 subunits. The furin cleavage motif contributes to the high infectivity and transmissibility of SARS-CoV-2. This review provides a comprehensive analysis of the molecular interactions of the spike protein, with a specific focus on the RBD and furin cleavage site. In addition to examining the binding characteristics with ACE2, the interactions with alternative receptors, such as neuropilin-1 (NRP1) and the nicotinic acetylcholine receptors (nAChRs) are highlighted. The ability of the spike protein to bind alternative receptors and host factors has been linked to the pathophysiology of COVID-19 and the persistence of symptoms in the post COVID-19 condition. Furthermore, we examine the impact of spike protein mutations on receptor affinity and disease severity. SARS-CoV-2 continues to evolve, with variants remaining an ongoing threat to public health. Understanding these molecular interactions is critical for the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Mia Argyrou
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC 3004, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Eleni Pitsillou
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC 3004, Australia; Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC 3053, Australia; School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Andrew Hung
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC 3004, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China; Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, 3/F Lui Che Woo Clinical Sciences Building, 30-32 Ngan Shing Street, Sha Tin, Hong Kong SAR, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, China; Biomedical Laboratory Science, Department of Technology, Faculty of Health, University College Copenhagen 2200 Copenhagen, Denmark
| | - Tom C Karagiannis
- Epigenetics in Human Health and Disease Program, Baker Heart and Diabetes Institute, 75 Commercial Road, Prahran, VIC 3004, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, VIC 3010, Australia; Epigenomic Medicine Laboratory at prospED Polytechnic, Carlton, VIC 3053, Australia; Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
11
|
Zhang J, Kennedy A, de Melo Jorge DM, Xing L, Reid W, Bui S, Joppich J, Rose M, Ercan S, Tang Q, Ginsburg D, Tai AW, Wang Y. SARS-CoV-2 remodels the Golgi apparatus to facilitate viral assembly and secretion. PLoS Pathog 2025; 21:e1013295. [PMID: 40540531 DOI: 10.1371/journal.ppat.1013295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Accepted: 06/12/2025] [Indexed: 06/22/2025] Open
Abstract
The COVID-19 pandemic is caused by the enveloped virus SARS-CoV-2. Despite extensive investigation, the molecular mechanisms for its assembly and secretion remain largely elusive. Here, we show that SARS-CoV-2 infection induces global alterations of the host endomembrane system, including dramatic Golgi fragmentation. SARS-CoV-2 virions are enriched in the fragmented Golgi. Blocking endoplasmic reticulum (ER) to Golgi trafficking dramatically inhibits SARS-CoV-2 assembly and secretion without reducing viral genome replication. Significantly, SARS-CoV-2 infection down-regulates GRASP55 but up-regulates TGN46 protein levels. Surprisingly, GRASP55 expression reduces both viral secretion and spike number on each virion without affecting viral entry, while GRASP55 depletion displays opposite effects. In contrast, TGN46 depletion only inhibits viral secretion without affecting spike incorporation into virions. Taken together, we show that SARS-CoV-2 alters Golgi structure and function to modulate viral assembly and secretion, highlighting the Golgi as a potential therapeutic target for blocking SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jianchao Zhang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Andrew Kennedy
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Daniel Macedo de Melo Jorge
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Lijuan Xing
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Whitney Reid
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Sarah Bui
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Joseph Joppich
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Molly Rose
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Sevval Ercan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, United States of America
| | - David Ginsburg
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Departmentof Internal Medicine, Human Genetics, and Pediatrics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Andrew W Tai
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| |
Collapse
|
12
|
Mótyán JA, Nagy T, Nagyné Veres Á, Golda M, Mahdi M, Tőzsér J. Identification of SARS-CoV-2 Main Protease Cleavage Sites in Bovine β-Casein. Int J Mol Sci 2025; 26:5829. [PMID: 40565292 DOI: 10.3390/ijms26125829] [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: 04/30/2025] [Revised: 06/10/2025] [Accepted: 06/16/2025] [Indexed: 06/28/2025] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the coronavirus disease of 2019 (COVID-19) and has persistently caused infections since its emergence in late 2019. The main protease (Mpro) of SARS-CoV-2 plays a crucial role in its life-cycle; thus, it is an important target for drug development. One of the first virus-specific drugs that has been approved for the treatment of COVID-19 patients is Paxlovid, which contains nirmatrelvir, a covalent inhibitor of Mpro. Screening of inhibitor candidates and specificity studies also rely on efficient substrates and activity assays. Casein is one of the most commonly applied universal substrates that can be used to study a wide range of proteases, including SARS-CoV-2 Mpro. Casein is a known substrate for Mpro in vitro, but the specific casein isoform cleaved by Mpro remained unidentified, and the cleavage sites have yet to be determined. This work studied cleavage of α-, β- and κ-isoforms of bovine casein by SARS-CoV-2 Mpro, using in vitro and in silico approaches. The candidate cleavage sites were predicted in silico based on the protein sequences, and the cleavage positions were identified based on mass spectrometric analysis of cleavage fragments. Based on our results, only β-casein contains cleavage sites for Mpro and thus can be used as its substrate in vitro. The newly identified cleavage site sequences further widen the knowledge about the specificity of SARS-CoV-2 Mpro.
Collapse
Affiliation(s)
- János András Mótyán
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tibor Nagy
- Department of Applied Chemistry, Faculty of Sciences and Technology, University of Debrecen, 4032 Debrecen, Hungary
| | - Ágota Nagyné Veres
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Mária Golda
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Mohamed Mahdi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - József Tőzsér
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
13
|
Amodeo D, Marchi S, Fiaschi L, Raucci L, Biba C, Salvestroni V, Trombetta CM, Manini I, Zazzi M, Montomoli E, Vicenti I, Cevenini G, Messina G. Analysis of the SARS-CoV-2 inactivation mechanism using violet-blue light (405 nm). Appl Environ Microbiol 2025; 91:e0040325. [PMID: 40366184 DOI: 10.1128/aem.00403-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
The study evaluated the effects of violet-blue light (VBL) on cell viability and replication, carbonylation of three structural proteins (S, E, and N) and one non-structural protein (NSP13), and direct damage to the RNA of SARS-CoV-2. The virus was exposed to increasing doses of VBL along with influenza A and B viruses to compare their susceptibility. At the highest dose (21.6 J/cm2), SARS-CoV-2 was significantly more susceptible to VBL than the influenza viruses, with a reduction in viral titer of 2.33 log10. Viral RNA did not show significant changes after exposure to VBL, as demonstrated by next-generation sequencing and real-time PCR quantification, suggesting that the inactivation process does not involve direct nucleic acid damage. To exclude the role of the culture suspension in the inactivation process, virus viability experiments were performed using different dilutions of Dulbecco's modified Eagle's medium (DMEM) in phosphate-buffered saline (PBS). The results indicated that the suspension medium played a secondary role in virus inactivation, as viability did not increase with increasing DMEM dilution. Subsequent tests with three different antioxidants (NAC, AsA, and SOD) at different concentrations prevented viral inactivation, from 99.99% to 85.43% (with SOD 0.003 mM). Carbonylation of S and E proteins was more pronounced when viruses were suspended in DMEM rather than PBS, although the tests demonstrated that the intrinsic properties of the viral membrane were a crucial element to consider in relation to its susceptibility to VBL.IMPORTANCELight-based disinfection methods are often used in combination with other cleaning methods due to their non-invasive nature, versatility, and environmental benefits. VBL is an effective approach as it induces the production of reactive oxygen species that reduce microbial viability. In this study, lipid peroxidation was identified as an important factor affecting the structural integrity and function of the viral envelope, reducing its ability to interact with host cells and consequently its ability to be infectious. The lipid envelope of SARS-CoV-2, composed mainly of glycerophospholipids and lacking cholesterol and sphingolipids, appears to be the critical factor in its susceptibility, distinguishing it from influenza viruses, which have a lipid profile richer in components that protect against oxidative stress.
Collapse
Affiliation(s)
- Davide Amodeo
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Serena Marchi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Lia Fiaschi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Luisa Raucci
- Department of Biotechnology, chemistry and pharmacy, University of Siena, Siena, Italy
| | - Camilla Biba
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Valentina Salvestroni
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | - Ilaria Manini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Maurizio Zazzi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VisMederi srl, Siena, Italy
| | - Ilaria Vicenti
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gabriele Cevenini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gabriele Messina
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
14
|
Lugano D, Mwangi K, Mware B, Kibet G, Osiany S, Kiritu E, Dobi P, Muli C, Njeru R, de Oliveira T, Njenga MK, Routh A, Oyola SO. Characterization of SARS-CoV-2 intrahost genetic evolution in vaccinated and non-vaccinated patients from the Kenyan population. J Virol 2025; 99:e0048225. [PMID: 40326760 DOI: 10.1128/jvi.00482-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 05/07/2025] Open
Abstract
Vaccination is a key control measure of coronavirus disease 2019 by preventing severe effects of disease outcomes, reducing hospitalization rates and death, and increasing immunity. However, vaccination can affect the evolution and adaptation of SARS-CoV-2 largely through vaccine-induced immune pressure. Here, we investigated intrahost recombination and single nucleotide variations (iSNVs) on the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome in non-vaccinated and vaccinated sequences from the Kenyan population to profile intrahost viral genetic evolution and adaptations driven by vaccine-induced immune pressure. We identified recombination hotspots in the S, N, and ORF1a/b genes and showed the genetic evolution landscape of SARS-CoV-2 by comparing within- and inter-wave recombination events from the beginning of the pandemic (June 2020 to December 2022) in Kenya. We further reveal differential expression of recombinant RNA species between vaccinated and non-vaccinated individuals and perform an in-depth analysis of iSNVs to identify and characterize the functional properties of non-synonymous mutations found in ORF-1 a/b, S, and N genes. Lastly, we detected a minority variant in non-vaccinated patients in Kenya, with an immune escape mutation S255F of the spike gene, and showed differential recombinant RNA species. Overall, this work identified unique in vivo mutations and intrahost recombination patterns in SARS-CoV-2, which could have significant implications for virus evolution, virulence, and immune escape.IMPORTANCEThe impact of vaccination on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genetic diversity in Kenya and much of Africa remains unknown. This can be attributed to lower sequencing rates; however, this information is relevant to improvement in vaccine and antiviral research. In this study, we investigated how vaccination and SARS-CoV-2 transmission waves affect intrahost non-homologous recombination and single nucleotide variations (iSNVs). We identified unique in vivo mutations and intrahost recombination patterns in SARS-CoV-2, which could have significant implications for virus evolution, virulence, and immune escape. We also demonstrate a methodology for studying genetic changes in a pathogen by a simultaneous analysis of both intrahost single nucleotide variations and recombination events. The study reveals the diversity of SARS-CoV-2 in Kenya and highlights the need for sustained genomic surveillance in Kenya and Africa to better understand how the virus evolves. Such surveillance ensures detection of drifts in evolution, allowing information for updates in vaccines, policy making, and containment of future variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Doreen Lugano
- International Livestock Research Institute, Nairobi, Kenya
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, USA
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kennedy Mwangi
- International Livestock Research Institute, Nairobi, Kenya
| | - Bernard Mware
- International Livestock Research Institute, Nairobi, Kenya
| | - Gilbert Kibet
- International Livestock Research Institute, Nairobi, Kenya
| | - Shebbar Osiany
- International Livestock Research Institute, Nairobi, Kenya
| | - Edward Kiritu
- International Livestock Research Institute, Nairobi, Kenya
| | - Paul Dobi
- International Livestock Research Institute, Nairobi, Kenya
| | - Collins Muli
- International Livestock Research Institute, Nairobi, Kenya
| | - Regina Njeru
- International Livestock Research Institute, Nairobi, Kenya
| | - Tulio de Oliveira
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - M Kariuki Njenga
- Washington State Global Health Program-Kenya, Washington State University, Pullman, Washington, USA
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
| | - Andrew Routh
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, USA
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, USA
| | - Samuel O Oyola
- International Livestock Research Institute, Nairobi, Kenya
| |
Collapse
|
15
|
Jones CP, Ferré-D'Amaré AR. Crystallographic and cryoEM analyses reveal SARS-CoV-2 SL5 is a mobile T-shaped four-way junction with deep pockets. RNA (NEW YORK, N.Y.) 2025; 31:949-960. [PMID: 40527531 DOI: 10.1261/rna.080413.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 03/25/2025] [Indexed: 06/19/2025]
Abstract
Stem-loop 5 (SL5) is a structural element that is conserved across coronavirus genomic RNAs. It spans the start codon from which the long ORF1 is translated in full-length viral RNA. Phylogenetic conservation indicates that it is comprised of four paired elements, but the specific 3D arrangement of these helices has remained unknown. Now, we have solved the crystal structure of SL5 from SARS-CoV-2 at 3.3 Å resolution, finding that the RNA adopts a T-shaped four-way junction fold in which two coaxial stacks of two helices each pack orthogonally. This arrangement results in deep pockets at the helical junction, where cations bind. Except for limited interactions in this region, the structure is remarkable for the paucity of tertiary contacts. We confirmed the stability of this fold in solution by FRET and carried out single-particle cryogenic-sample electron microscopy (cryoEM). The resulting ∼5 Å resolution cryoEM map, and 3D variability analysis, suggest conformational flexibility at the junction. In vitro translation of structure-guided mutants demonstrated that SL5 inhibits protein synthesis. Thus, it is likely that SL5 recruits additional factors in vivo. This, and its characteristic clefts at the four-way junction, make SL5 an attractive target for the discovery of RNA-targeted antiviral small molecules.
Collapse
Affiliation(s)
- Christopher P Jones
- Laboratory of Nucleic Acids, National Heart, Lung and Blood Institute, Bethesda, Maryland 20892-8012, USA
| | - Adrian R Ferré-D'Amaré
- Laboratory of Nucleic Acids, National Heart, Lung and Blood Institute, Bethesda, Maryland 20892-8012, USA
| |
Collapse
|
16
|
Yin X, Pu Y, Yuan S, Pache L, Churas C, Weston S, Riva L, Simons LM, Cisneros WJ, Clausen T, Biddle G, Doss-Gollin S, Deming M, De Jesus PD, Kim HN, Fuentes D, Whitelock JM, Esko JD, Lord MS, Mena I, García-Sastre A, Hultquist JF, Frieman MB, Ideker T, Pratt D, Martin-Sancho L, Chanda SK. Global siRNA screen identifies human host factors critical for SARS-CoV-2 replication and late stages of infection. PLoS Biol 2025; 23:e3002738. [PMID: 40504864 DOI: 10.1371/journal.pbio.3002738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 05/12/2025] [Indexed: 06/18/2025] Open
Abstract
Defining the subset of cellular factors governing SARS-CoV-2 replication can provide critical insights into viral pathogenesis and identify targets for host-directed antiviral therapies. While a number of genetic screens have previously reported SARS-CoV-2 host dependency factors, most of these approaches relied on utilizing pooled genome-scale CRISPR libraries, which are biased toward the discovery of host proteins impacting early stages of viral replication. To identify host factors involved throughout the SARS-CoV-2 infectious cycle, we conducted an arrayed genome-scale siRNA screen. Resulting data were integrated with published functional screens and proteomics data to reveal (i) common pathways that were identified in all OMICs datasets-including regulation of Wnt signaling and gap junctions, (ii) pathways uniquely identified in this screen-including NADH oxidation, or (iii) pathways supported by this screen and proteomics data but not published functional screens-including arachionate production and MAPK signaling. The identified proviral host factors were mapped into the SARS-CoV-2 infectious cycle, including 32 proteins that were determined to impact viral replication and 27 impacting late stages of infection, respectively. Additionally, a subset of proteins was tested across other coronaviruses revealing a subset of proviral factors that were conserved across pandemic SARS-CoV-2, epidemic SARS-CoV-1 and MERS-CoV, and the seasonal coronavirus OC43-CoV. Further studies illuminated a role for the heparan sulfate proteoglycan perlecan in SARS-CoV-2 viral entry and found that inhibition of the non-canonical NF-kB pathway through targeting of BIRC2 restricts SARS-CoV-2 replication both in vitro and in vivo. These studies provide critical insight into the landscape of virus-host interactions driving SARS-CoV-2 replication as well as valuable targets for host-directed antivirals.
Collapse
Affiliation(s)
- Xin Yin
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yuan Pu
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Shuofeng Yuan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Lars Pache
- NCI Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Christopher Churas
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Stuart Weston
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Laura Riva
- Calibr-Skaggs at Scripps Research Institute, La Jolla, California, United States of America
| | - Lacy M Simons
- Division of Infectious Diseases, Departments of Medicine and Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - William J Cisneros
- Division of Infectious Diseases, Departments of Medicine and Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Thomas Clausen
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Grace Biddle
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Simon Doss-Gollin
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Meagan Deming
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Paul D De Jesus
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Ha Na Kim
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Daniel Fuentes
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - John M Whitelock
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Megan S Lord
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Ignacio Mena
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Tisch Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Judd F Hultquist
- Division of Infectious Diseases, Departments of Medicine and Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Matthew B Frieman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Trey Ideker
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, California, United States of America
| | - Dexter Pratt
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Laura Martin-Sancho
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Sumit K Chanda
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, United States of America
- Calibr-Skaggs at Scripps Research Institute, La Jolla, California, United States of America
| |
Collapse
|
17
|
Shitaye G, Ventserova N, D’Abrosca G, Dragone M, Maina EW, Fattorusso R, Iacovino R, Russo L, Isernia C, Malgieri G. The role of intrinsically disordered regions of SARS-CoV-2 nucleocapsid and non-structural protein 1 proteins. Front Chem 2025; 13:1597656. [PMID: 40568634 PMCID: PMC12187666 DOI: 10.3389/fchem.2025.1597656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 05/30/2025] [Indexed: 06/28/2025] Open
Abstract
Virus survival inside the host cell depends on the intricate mechanisms that recruit proteins involved in the arms race. Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) proteome exhibits important levels of structural order. However, some of the SARS-CoV-2 proteins, such as the Nucleocapsid (N) and Non-structural protein 1 (Nsp1), contain a considerably significant amount of intrinsically disordered regions (IDRs) that play indispensable roles in the intra-viral and virus-host interaction. Here, focusing on proteins that contain a relevant percentage of IDRs, we discuss experimental and computational studies sought to support IDRs as a key player in the interplay with ordered domains, the biological role as potential origin for variants of SARS-CoV-2, and their association with virus transmissibility. Furthermore, we also highlight the potential involvement of IDRs in the viral-host protein interaction and host cellular machinery. Thus, shading lights on the dark proteome of the virus and looking for therapeutic approaches beyond the classic structure-function paradigm may contribute to the efforts sparking the quest for therapeutics.
Collapse
Affiliation(s)
- Getasew Shitaye
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
- Department of Biomedical Sciences, College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
| | - Nataliia Ventserova
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | | | - Martina Dragone
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Eunice Wairimu Maina
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Roberto Fattorusso
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Rosa Iacovino
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Luigi Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Carla Isernia
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
- Interuniversity Research Centre on Bioactive Peptides, University of Naples “Federico II”, Naples, Italy
| | - Gaetano Malgieri
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
- Interuniversity Research Centre on Bioactive Peptides, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
18
|
Han Y, Xu F. PLpro Inhibitors as a Potential Treatment for COVID-19. Biomedicines 2025; 13:1417. [PMID: 40564136 DOI: 10.3390/biomedicines13061417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 05/28/2025] [Indexed: 06/28/2025] Open
Abstract
The advent of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the subsequent coronavirus disease 2019 (COVID-19) pandemic have posed a serious threat to human health and society [...].
Collapse
Affiliation(s)
- Yu Han
- Department of Infectious Diseases, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Feng Xu
- Department of Infectious Diseases, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
19
|
Luwen G, Liyan Z, Rahman A, Baloch Z. Deciphering the NSP12/7/8 complex: Key insights into coronavirus replication and potential therapeutic targets. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102505. [PMID: 40206654 PMCID: PMC11979514 DOI: 10.1016/j.omtn.2025.102505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Affiliation(s)
- Gao Luwen
- Jiangxi Medical College, Shangrao, Jiangxi, China
| | - Zhou Liyan
- Jiangxi Medical College, Shangrao, Jiangxi, China
| | - Abdul Rahman
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, Guangdong, China
- School of Food Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zulqarnain Baloch
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
20
|
Lovell TC, Dewling HAF, Li C, Lee HW, Gordon CJ, Kocincova D, Badmalia MD, Tchesnokov EP, Götte M, Cosa G. Single-Molecule Assay Reveals Binding Dynamics of SARS-CoV-2 Polymerase Components and Provides a New Tool to Distinguish Polymerase Inhibitors. ACS Infect Dis 2025. [PMID: 40465830 DOI: 10.1021/acsinfecdis.5c00062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
The genome replication of SARS-CoV-2, the causative agent of COVID-19, involves a multisubunit replication complex consisting of nonstructural proteins (nsps) 12, 7, and 8. While the structure of this complex is known, the dynamic behavior of the subunits interacting with RNA is missing. Here we report a single-molecule protein induced fluorescence enhancement (SM-PIFE) assay to monitor binding dynamics between the reconstituted or coexpressed replication complex and RNA. Increasing binding times were observed, in this order, with nsp7 (none), nsp8, and nsp12, in nsp8 nsp12 mixtures and in reconstituted mixtures bearing all three proteins. Unstable, unstable→stable, and stable binding modes were recorded in the latter case, indicating that complexation is dynamic and the correct conformation must be achieved before stable RNA binding can occur. Notably, the coexpressed protein yields mostly stable binding even at low concentrations, while the reconstituted proteins exhibit unstable binding indicating inefficient complexation with reduced protein. The SM-PIFE assay distinguishes inhibitors that impact protein binding from those that prevent replication, as demonstrated with suramin and remdesivir, respectively. The data reveals a correlation between binding lifetime/affinity and protein activity and underscores differences between coexpressed vs reconstituted mixtures, suggesting the existence of trapped conformations that may not evolve to productive binding.
Collapse
Affiliation(s)
- Terri C Lovell
- Department of Chemistry and Quebec Center for Advanced Materials (QCAM), McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Heidi A F Dewling
- Department of Chemistry and Quebec Center for Advanced Materials (QCAM), McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Cynthia Li
- Department of Chemistry and Quebec Center for Advanced Materials (QCAM), McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| | - Hery W Lee
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Calvin J Gordon
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Dana Kocincova
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Maulik D Badmalia
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Egor P Tchesnokov
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Matthias Götte
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Gonzalo Cosa
- Department of Chemistry and Quebec Center for Advanced Materials (QCAM), McGill University, 801 Sherbrooke Street West, Montreal, Quebec H3A 0B8, Canada
| |
Collapse
|
21
|
Nukaga S, Fujiwara-Tani R, Mori T, Kawahara I, Nishida R, Miyagawa Y, Goto K, Ohmori H, Fujii K, Sasaki T, Nakashima C, Luo Y, Mori S, Kishi S, Ogata R, Kuniyasu H. SARS-CoV-2-Derived RNA Fragment Induces Myocardial Dysfunction via siRNA-like Suppression of Mitochondrial ATP Synthase. Int J Mol Sci 2025; 26:5392. [PMID: 40508201 PMCID: PMC12156209 DOI: 10.3390/ijms26115392] [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: 04/29/2025] [Revised: 05/31/2025] [Accepted: 06/02/2025] [Indexed: 06/16/2025] Open
Abstract
Myocardial injury is a critical determinant of prognosis in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection; however, its underlying mechanisms remain incompletely understood. In this study, we examined the effects of SARS-CoV-2-derived RNA fragments on human cardiomyocytes. We identified a 19-nucleotide sequence within the viral genome that shares complete sequence homology with the human F1F0 ATP synthase subunit alpha gene (ATP5A). This sequence was found to associate with Argonaute 2 (AGO2) and downregulate ATP5A expression via a mechanism analogous to RNA interference. Consequently, oxidative phosphorylation was suppressed in cardiomyocytes, leading to impaired myocardial maturation and the emergence of heart failure-like phenotypes. Notably, exosome-mimetic liposomal delivery of this RNA fragment to cardiomyocytes reproduced the ATP5A-suppressive effect. These findings suggest that SARS-CoV-2-derived RNA fragments may contribute to myocardial injury through the siRNA-like modulation of mitochondrial gene expression. Further validation in animal models and patient-derived materials is warranted.
Collapse
Grants
- 25K14462 Ministry of Education, Culture, Sports, Science and Technology
- 25K14487 Ministry of Education, Culture, Sports, Science and Technology
- 24K20535 Ministry of Education, Culture, Sports, Science and Technology
- 24K14281 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Shota Nukaga
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
- Division of Rehabilitation, Hanna Central Hospital, Ikoma 630-0243, Japan
| | - Rina Fujiwara-Tani
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
| | - Takuya Mori
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
- Department of Medical Ethics and Genetics, Kyoto University, Kyoto 606-8501, Japan
| | - Isao Kawahara
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
- Division of Rehabilitation, Hanna Central Hospital, Ikoma 630-0243, Japan
| | - Ryoichi Nishida
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
| | - Yoshihiro Miyagawa
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
| | - Kei Goto
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
| | - Hitoshi Ohmori
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
| | - Kiyomu Fujii
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
| | - Takamitsu Sasaki
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
| | - Chie Nakashima
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
| | - Yi Luo
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
| | - Shiori Mori
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Osaka 573-1010, Japan
| | - Shingo Kishi
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
- Department of Pathological Diagnosis, Nozaki Tokushukai Hospital, Daito 574-0074, Japan
| | - Ruiko Ogata
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University School of Medicine, Kashihara 634-8521, Japan
| |
Collapse
|
22
|
Wu X, Tang L, Huang W, Gao M, Xu C, Li P, Kong X. Membrane Protein of SARS-CoV-2 Promotes the Production of CXCL10 and Apoptosis of Myocardial Cells. Cardiovasc Toxicol 2025; 25:830-840. [PMID: 40293660 DOI: 10.1007/s12012-025-10001-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025]
Abstract
SARS-CoV-2 infections directly or indirectly cause unconscionable vascular events, significantly increasing the morbidity and mortality of COVID-19. Biomarkers associated with cardiac injury often elevate in individuals with COVID-19. Cytokine storm is a mechanism underlying myocardial cell injury caused by SARS-CoV-2 infections. Cell apoptosis in AC16 cells overexpressing structural and helper proteins of SARS-CoV-2 was detected by Western blot, MTT and TUNEL assay. The M protein was determined to play the most pronounced role in inducing apoptosis. Transcriptome sequencing on AC16 cells overexpressing the M protein was performed to screen differentially expressed genes (DEGs), which were further subjected to the gene set enrichment analysis. The regulatory effect of CXCL10 on cell apoptosis of AC16 cells overexpressing M protein was finally explored. Overexpression of M protein significantly increased the Bax/Bcl-2 and cleaved caspase-3/caspase-3(CC3/C3) ratios and the percentage of TUNEL-positive cells in AC16 cells, while markedly reducing cell viability. CXCL10 was the most prominent DEG in AC16 cells overexpressing M protein. Knockdown of CXCL10 partially reversed the increases in the Bax/Bcl-2 and cleaved caspase-3/caspase-3 ratios, the percentage of TUNEL-positive cells, as well as the release of pro-inflammatory cytokines in AC16 cells overexpressing M protein. The M protein of SARS-CoV-2 triggers the production of CXCL10 and apoptosis of myocardial cells.
Collapse
Affiliation(s)
- Xiaoguang Wu
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Lu Tang
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Wen Huang
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Min Gao
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Changhao Xu
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China.
| | - Xiangqing Kong
- Department of Cardiology, The First Affiliated Hospital With Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
23
|
Calandria JM, Bazan HEP, Bhattacharjee S, Kautzmann MI, Maness NJ, Bazan NG. ELV-N34, RvD6-Isomer, or NPD1 Halt Replication of SARS-CoV-2 Omicron BA.5 Virus in Human Lung and Nasal Cells. FASEB J 2025; 39:e70563. [PMID: 40407038 PMCID: PMC12100679 DOI: 10.1096/fj.202403197r] [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: 12/11/2024] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025]
Abstract
Current vaccines rely on the sequence of Spike (S) protein to induce immunity against the severe acute respiratory coronavirus-2 (SARS-CoV-2) virus. Because of the high mutation rate of the viral S protein, new mutant strains are developed to generate new infectivity profiles. Bioactive lipid mediators (LMs) derived from docosahexaenoic acid (DHA) are synthesized on demand to sustain homeostasis. The purpose of this study was to determine the action of selected LMs in the viral replication of SARS-CoV-2 Omicron BA.5 variant in human lung and nasal epithelial cells. Cells from healthy donors were infected with Omicron BA.5 for one hour and treated with 500 nM Elovanoid (ELV)-N32, ELV-N34, Resolvin D6 isomer (RvD6i), Neuroprotection D1 (NPD1), or vehicle before and after infection. Impedance was recorded to determine cell death by infectivity. Cells were then immunostained for nucleocapsid (N) protein, microtubule-associated protein 1B-light chain 3 (LC3B), and autophagic proteins. N and S RNA were measured to assess the synthesis of viral components. The addition of ELV-N34 or RvD6i decreased the synthesis of N RNA by 76.7% and 96.9%, respectively, in lung primary culture, while NPD1 exerted the same effect in nasal epithelial cells (61.7% reduction). In lung cells, transcription of autophagy-related gene-3 (ATG3) and Sequestosome 1 (SQSTM1/p62), components of the autophagy initiation process, decreased compared to the non-treated infected cells. The results suggest that specific LMs prevent viral autophagy machinery hijacking, leading to a decrease in BA.5 replication. This novel effect of the bioactive LMs as antivirals, regardless of the protein sequence, would potentially complement vaccination and other prevention and treatment therapeutics.
Collapse
Affiliation(s)
- Jorgelina M. Calandria
- Neuroscience Center of ExcellenceLouisiana State University Health New OrleansNew OrleansLouisianaUSA
| | - Haydee E. P. Bazan
- Neuroscience Center of ExcellenceLouisiana State University Health New OrleansNew OrleansLouisianaUSA
| | - Surjyadipta Bhattacharjee
- Neuroscience Center of ExcellenceLouisiana State University Health New OrleansNew OrleansLouisianaUSA
| | - Marie‐Audrey I. Kautzmann
- Neuroscience Center of ExcellenceLouisiana State University Health New OrleansNew OrleansLouisianaUSA
| | | | - Nicolas G. Bazan
- Neuroscience Center of ExcellenceLouisiana State University Health New OrleansNew OrleansLouisianaUSA
| |
Collapse
|
24
|
Stasko N, Arwood L, Jandick N, Spragion D, Roberts RC, Setién M, Henson I, Annas A, Fulcher ML, Brotton M, Kummer L, Szaba F, Reagan M, Lanzer K, Cookenham T, Casey S, Kothapalli N, Hart T, Bradrick SS, Emerson D, Cockrell AS, Randell SH, Kocher JF. The pan-variant potential of light: 425 nm light inactivates SARS-CoV-2 variants of concern and non-cytotoxic doses reduce viral titers in human airway epithelial cells. mSphere 2025:e0023025. [PMID: 40434113 DOI: 10.1128/msphere.00230-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) prolonged the coronavirus disease 2019 (COVID-19) pandemic. The continued development of novel pan-variant therapeutics to treat currently circulating and future VOCs is critically important. Photomedicine may offer broadly applicable, pan-variant treatments. In this study, we show that visible light centered around 425 nm inactivates each of the five SARS-CoV-2 VOC lineages that have been identified by the World Health Organization (Alpha, Beta, Delta, Gamma, and Omicron) in cell-free suspensions in a dose-dependent manner, including bamlanivimab-resistant variants. Specifically, 60 J/cm2 of 425 nm light reduced SARS-CoV-2 titers by >4 log10 relative to unilluminated controls. We observed that 425 nm light inactivates SARS-CoV-2 through restricted entry to host cells. In addition, a non-cytotoxic dosing regimen of 32 J/cm2 of 425 nm light reduced infectious virus titers in well-differentiated air-liquid interface (ALI) human airway epithelial (HAE) cells infected with the Beta, Delta, and Omicron variants that incorporate mutations associated with immune evasion and/or increased transmissibility. Infectious SARS-CoV-2 titers were reduced when dosing began during the early stages of infection or in more established infections. Finally, we translated these findings to the RD-X19, a novel medical device that emits 425 nm light; our results showed that the RD-X19 restricted spike binding to ACE-2 and reduced SARS-CoV-2 titers in cell-free suspensions (by >2 log10) and in the ALI HAE model (by >1 log10). These findings indicate that photomedicine utilizing 425 nm visible light may serve as a novel, pan-variant treatment modality for COVID-19.IMPORTANCEThe continued spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to the emergence of variants that can evade public health measures, including vaccines and therapeutics. Thus, the continued development of broadly applicable measures to supplement current public health measures and standards of care remains critical. Photomedicine is one such approach. In this study, we show that non-ultraviolet visible light can inactivate each SARS-CoV-2 variant of concern (VOC) by preventing entry to host cells. Furthermore, visible light reduced the amount of virus produced in an infection model of the human airway at multiple stages of infection, demonstrating the antiviral capability of visible light. This study provides preclinical support for the development of visible light to serve as a SARS-CoV-2 countermeasure and warrants further investigation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - M Leslie Fulcher
- The Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Marisa Brotton
- The Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Frank Szaba
- Trudeau Institute, Saranac Lake, New York, USA
| | - Matt Reagan
- Trudeau Institute, Saranac Lake, New York, USA
| | | | | | - Sean Casey
- Trudeau Institute, Saranac Lake, New York, USA
| | | | - Tricia Hart
- Trudeau Institute, Saranac Lake, New York, USA
| | | | | | | | - Scott H Randell
- The Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | |
Collapse
|
25
|
Khan Z, Farooq S, Atia-Tul-Wahab, Iqbal H, Naz F, Iftner T, Khan KM, Yusuf M, Choudhary MI. Sulfonohydrazide as a potential inhibitor of SARS-CoV-2 infection. Sci Rep 2025; 15:18732. [PMID: 40437072 PMCID: PMC12119938 DOI: 10.1038/s41598-025-03685-2] [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: 12/05/2024] [Accepted: 05/21/2025] [Indexed: 06/01/2025] Open
Abstract
The COVID-19 pandemic caused immense mortality and morbidity reporting 704,753,890 cases worldwide. The repercussions of this pandemic are still being felt in the form of newly evolving variants and infections. The pandemic has pointed towards the need for the development of new and effective agents against SARS-CoV-2 infection. Sulfonohydrazides are a class of compounds with a wide range of therapeutic potential. The present study aims to identify the anti-SARS-CoV-2 potential of Sulfonohydrazide compounds. Twenty-five Sulfonohydrazides derivatives were evaluated for anti-viral potential via plaque reduction assay (PRA) and cytopathic effect (CPE) analysis in-vitro. Treatment point assay was employed for the strategic evaluation of antiviral compound at the particular stages of the SARS-CoV-2 life cycle. Gene expression analysis was also carried out, which was supported by immunofluorescence assays targeting the N and S proteins of SARS-CoV-2, alongside fold-change analysis, to identify a robust and multifaceted approach for the understanding of viral dynamics. Moreover, ligand-inhibitor interactions were assessed by in- silico studies. Compound 24 (4(E)-4-methyl-N'-(2,3,4-trihydroxybenzylidene)benzenesulfonohydrazide) was identified as the most potent molecule that inhibited SARS-CoV-2 infection (92.85 ± 3.57%) via PRA. The time point assay revealed that the effect of the compound might be at the entry point, which might be due to the down-regulation of the Spike (S) and Angiotensin-converting enzyme 2 (ACE-2) genes by the compound. The gene expression analysis of ORF1a/b by qRT-PCR indicated reduction in viral load after compound treatment, as indicated by a higher cycle threshold (Ct) value. Moreover, the compound 24 also downregulated the expression of S, RdRp, and ACE-2. Furthermore, the interaction of compound 24 with S, RdRp, and ACE-2 was predicted via molecular docking, which validated the interaction and possible anti-SARS-CoV-2 effect. Additionally, immunofluorescence staining analysis of spike and nucleocapsid proteins also showed downregulation in SARS-CoV-2 infected cells. Overall, the acquired data suggested that Sulfonohydrazide derivative 24 inhibits SARS-CoV-2 entry and replication.
Collapse
Affiliation(s)
- Zoha Khan
- National Institute of Virology, Dr. Panjwani Centre for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
- Dr. Panjwani Centre for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Saba Farooq
- National Institute of Virology, Dr. Panjwani Centre for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
- Dr. Panjwani Centre for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| | - Atia-Tul-Wahab
- Dr. Panjwani Centre for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Hana'a Iqbal
- National Institute of Virology, Dr. Panjwani Centre for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
- Dr. Panjwani Centre for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Farzana Naz
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Thomas Iftner
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital and Medical Faculty, Eberhard Karls University, 72076, Tuebingen, Germany
| | - Khalid M Khan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Muhammad Yusuf
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jl Ir. Soekarno KM 21, Jatinangor, 45363, West Java, Indonesia
| | - M Iqbal Choudhary
- National Institute of Virology, Dr. Panjwani Centre for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
- Dr. Panjwani Centre for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21412, Saudi Arabia.
| |
Collapse
|
26
|
Nguyen AN, Kouyate TS, Ryff K, Plotkin AL, Doss-Gollin S, Thomas S, McEnaney K, Ozonoff A, Diray-Arce J, Levy O, Odumade OA, Baden LR, van Haren SD, Smolen KK, IMPACC - Boston Team. Patients Hospitalized with COVID-19 Demonstrate Distinct Plasma Cytokine and Chemokine Concentrations in vivo and TLR-Mediated Cytokine and Chemokine Production in Whole Blood in vitro. J Innate Immun 2025; 17:288-301. [PMID: 40435967 PMCID: PMC12185065 DOI: 10.1159/000545432] [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: 11/27/2024] [Accepted: 03/18/2025] [Indexed: 06/28/2025] Open
Abstract
INTRODUCTION SARS-CoV-2's continued global health impact underscores the importance of ongoing pathogenesis research. Insights into the host's first line of defense against severe COVID-19 identify actionable biomarkers, informing disease management or therapeutics. Yet, the innate immune response, including cytokines, chemokines, adenosine deaminases (ADAs) and Toll-like receptors (TLRs), relevant to COVID-19 remain incompletely characterized. METHODS Peripheral blood was longitudinally collected between May 2020 and March 2021 from COVID-19 hospitalized adults (N = 79) and healthy controls (HCs) (N = 14; not tested, assumed COVID-negative, no viral exposure or symptoms). Heparinized blood was fractionated for plasma cryopreservation and in vitro whole blood TLR-stimulation employing TLR-3, -4, and -7/8 agonists. Post-stimulation culture supernatants were analyzed using multiplex and enzymatic assays. RESULTS Upon hospitalization, plasma concentrations of IFNγ, IL-6, CXCL10, and ADAs were significantly upregulated compared to convalescent time points and HCs. Participants with fatal COVID-19 exhibited higher IL-27, CXCL10, and ADAs concentrations upon admission. Plasma cytokines, chemokines, and ADAs were positively correlated and associated with distinct temporal patterns. TLR-stimulated cell cultures from patients produced reduced IFNα2, IFNγ, IL-12p40, and IL-12p70 compared to HCs or later time points. CONCLUSION Higher plasma concentrations of IL-27, CXCL10, and ADAs at admission were associated with severe COVID-19 and mortality. Reduced TLR-mediated IFNα2, IFNγ, and IL-12p70 production suggests COVID dampens Th1-polarizing innate immune responses, providing insight into immunological sequelae of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Athena N Nguyen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Thomas S Kouyate
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA,
| | - Kevin Ryff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Alec L Plotkin
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Simon Doss-Gollin
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Sanya Thomas
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Kerry McEnaney
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA
| | - Oludare A Odumade
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | | | - Simon D van Haren
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Kinga K Smolen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
27
|
Fischer HL, Kline C, Duprex WP, McCarthy KR, Watkins SC, Conway JF, Ambrose Z. Deletion of the Envelope gene attenuates SARS-CoV-2 infection by altered Spike localization and increased cell-to-cell transmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.20.655126. [PMID: 40475451 PMCID: PMC12140015 DOI: 10.1101/2025.05.20.655126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes COVID-19, a highly transmissible acute respiratory infection that can result in severe pneumonia and death. Many details of SARS-CoV-2 infection are not fully understood, including the cell biology and host-virus interactions involved in coronavirus assembly and release, in which the Envelope (E) structural protein is instrumental. Deletion of E in other coronaviruses has been shown previously to either attenuate or abrogate infection. To determine the role of E on SARS-CoV-2 virus production and infectivity, we produced reporter SARS-CoV-2 with or without the E gene deleted using a bacterial artificial chromosome. Replication of ΔE SARS-CoV-2 was attenuated in Vero E6 cells expressing human ACE2 and TMPRSS2 and in human epithelial cell lines. Electron and immunofluorescence microscopy and virology assays showed that ΔE SARS-CoV-2 increased cell surface expression of Spike (S) glycoprotein, leading to reduced S incorporation into ΔE SARS-CoV-2 particles and promotion of increased cell-to-cell transmission that evades neutralizing antibody inhibition. Trans-complementation of E partially rescued ΔE SARS-CoV-2 S incorporation and restored cell-free transmission. In addition to validating the role of E in retention of S in the ER-Golgi intermediate complex (ERGIC), our results showed that a lack of E led to reorganization of the ERGIC during SARS-CoV-2 infection. Improved understanding of E in SARS-CoV-2 replication and host pathogenesis may help development of novel therapeutics. Importance Non-S coronavirus structural proteins, including E, are conserved, making them potential pan-coronavirus therapeutic targets. Many details about these proteins and their roles in viral replication and host pathogenesis are unknown. In this study, we showed that SARS-CoV-2 replicates without E but is attenuated and impaired for virus particle formation, with less S incorporated into virions and more S expressed on the cell surface compared to wild-type virus. SARS-CoV-2 lacking E spread primarily via cell fusion and evaded neutralizing antibodies. In addition, the absence of E resulted in the reorganization of the ERGIC cell secretory compartment during SARS-CoV-2 infection. A better understanding of how E influences SARS-CoV-2 replication could guide directed design of novel therapeutics for treatment of COVID-19 patients, as well as the potential for pan-coronavirus protection against future coronavirus outbreaks.
Collapse
Affiliation(s)
- Hannah L. Fischer
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA
| | - Christopher Kline
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - W. Paul Duprex
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA
| | - Kevin R. McCarthy
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA
| | - Simon C. Watkins
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - James F. Conway
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Zandrea Ambrose
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
28
|
Hou Y, Shi H, Wang H, Tian L, Huan C, Liu Y, Wang H, Zhang W. HERC5-mediated ISGylation of SARS-CoV-2 nsp8 facilitates its degradation and inhibits viral replication. Int J Biol Macromol 2025; 315:144546. [PMID: 40409630 DOI: 10.1016/j.ijbiomac.2025.144546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/19/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 non-structural protein 8 (SARS-CoV-2 nsp8) is a multifunctional protein essential for viral replication and immune evasion. However, the host factors that regulate nsp8 stability and function remain unclear. In this study, we identify HECT and RCC-like domain-containing protein 5 (HERC5) as an essential E3 ligase that regulates nsp8 stability through ISGylation, a ubiquitin-like post-translational modification that facilitates proteasome-dependent degradation. HERC5 overexpression significantly enhances nsp8 degradation in an enzymatic activity-dependent manner, whereas SARS-CoV-2 papain-like protease (PLpro) counteracts this process by deconjugating interferon-stimulated gene 15 (ISG15) from nsp8-thereby preventing its degradation and facilitating viral replication. Mass spectrometry and mutational analyses revealed that the N2 domain of nsp8 is indispensable for ISGylation, with multiple lysine residues acting as primary modification sites. Additionally, we demonstrated that the ISGylation system, including HERC5, ubiquitin-like modifier activating enzyme 7 (UBA7), and ISG15, effectively suppresses SARS-CoV-2 replication across multiple variants, including Omicron BA.5 and XBB.1.5.15. These findings provide novel insights into the role of ISGylation in host antiviral defense and highlight the interplay between HERC5 and PLpro in modulating viral replication. This study establishes a foundation for developing therapeutic strategies targeting HERC5 or PLpro to inhibit SARS-CoV-2 replication.
Collapse
Affiliation(s)
- Yubao Hou
- Institute of Virology and AIDS Research, the First Hospital of Jilin University, Changchun, China; Centre of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, the First Hospital of Jilin University, Changchun, China
| | - Hongyun Shi
- Institute of Virology and AIDS Research, the First Hospital of Jilin University, Changchun, China; Centre of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, the First Hospital of Jilin University, Changchun, China
| | - Huihan Wang
- Institute of Virology and AIDS Research, the First Hospital of Jilin University, Changchun, China; Centre of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, the First Hospital of Jilin University, Changchun, China
| | - Li Tian
- Institute of Virology and AIDS Research, the First Hospital of Jilin University, Changchun, China; Centre of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, the First Hospital of Jilin University, Changchun, China
| | - Chen Huan
- Institute of Virology and AIDS Research, the First Hospital of Jilin University, Changchun, China; Centre of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, the First Hospital of Jilin University, Changchun, China
| | - Yan Liu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130000, Jilin, China.
| | - Hong Wang
- Institute of Virology and AIDS Research, the First Hospital of Jilin University, Changchun, China; Centre of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, the First Hospital of Jilin University, Changchun, China.
| | - Wenyan Zhang
- Institute of Virology and AIDS Research, the First Hospital of Jilin University, Changchun, China; Centre of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, the First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
29
|
Kawai M, Malla TR, Chan HTH, Tumber A, Brewitz L, Salah E, Terasaka N, Katoh T, Kawamura A, Schofield CJ, Duarte F, Suga H. RaPID discovery of cell-permeable helical peptide inhibitors con-taining cyclic β-amino acids against SARS-CoV-2 main protease. RSC Chem Biol 2025:d5cb00021a. [PMID: 40406165 PMCID: PMC12093385 DOI: 10.1039/d5cb00021a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 05/02/2025] [Indexed: 05/26/2025] Open
Abstract
Structurally constrained cyclic β-amino acids are attractive building blocks for peptide drugs because they induce unique and stable conformations. Introduction of (1S,2S)-2-aminocyclopentanecarboxylic acid [(1S,2S)-2-ACPC] into peptides stabilizes helical conformations, so improving proteolytic stability and cell membrane permeability. We report on the ribosomal synthesis of a helical peptide library incorporating (1S,2S)-2-ACPC at every third position and its application for the discovery of SARS-CoV-2 main protease (Mpro) inhibitors. We identified two peptide sequences containing multiple (1S,2S)-2-ACPC residues, which exhibit helical conformations and superior proteolytic stability compared with their α-Ala or β-Ala counterparts. Studies using the chloroalkane cell-penetration assay showed that their cell permeability values (CP50) are comparable with or even slightly better than that of the cell-penetrating nona-arginine (R9) peptide. The new approach is thus a highly efficient method that combines a helical peptide library containing structurally constrained cyclic β-amino acids with the classical RaPID discovery method, enabling de novo discovery of proteolytically stable and cell-penetrating bioactive peptides that target intracellular proteins.
Collapse
Affiliation(s)
- Marina Kawai
- Department of Chemistry, Graduate School of Science, The University of Tokyo Tokyo Japan
| | - Tika R Malla
- Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, Chemistry Research Laboratory, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - H T Henry Chan
- Physical and Theoretical Chemistry Laboratory, University of Oxford South Parks Road Oxford OX1 3QZ UK
| | - Anthony Tumber
- Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, Chemistry Research Laboratory, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Lennart Brewitz
- Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, Chemistry Research Laboratory, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Eidarus Salah
- Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, Chemistry Research Laboratory, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Naohiro Terasaka
- Department of Chemistry, Graduate School of Science, The University of Tokyo Tokyo Japan
| | - Takayuki Katoh
- Department of Chemistry, Graduate School of Science, The University of Tokyo Tokyo Japan
| | - Akane Kawamura
- Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, Chemistry Research Laboratory, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
- Chemistry - School of Natural and Environmental Sciences, Newcastle University Newcastle upon Tyne UK
| | - Christopher J Schofield
- Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, Chemistry Research Laboratory, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Fernanda Duarte
- Physical and Theoretical Chemistry Laboratory, University of Oxford South Parks Road Oxford OX1 3QZ UK
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo Tokyo Japan
| |
Collapse
|
30
|
Braconi L, Sosic A, Crocetti L. Recent breakthroughs in synthetic small molecules targeting SARS-CoV-2 M pro from 2022 to 2024. Bioorg Med Chem 2025; 128:118247. [PMID: 40413978 DOI: 10.1016/j.bmc.2025.118247] [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: 03/21/2025] [Revised: 05/07/2025] [Accepted: 05/17/2025] [Indexed: 05/27/2025]
Abstract
Among the identified targets for developing anti-coronavirus therapies, SARS-CoV-2 Mpro stands out as one of the most promising due to its crucial role in viral replication and its low mutability across various coronaviruses, making it a potential broad-spectrum target. Currently, although the approved drugs targeting Mpro are peptidomimetic inhibitors with an adequate efficacy, they exhibit relatively poor pharmacokinetic properties commonly associated with peptide-based compounds. On the contrary, using non-peptidic small-molecules Mpro inhibitors can offer many advantages, including reduced off-target toxicity, improved metabolic stability and drug-like properties more appropriate for oral administration. This topic has sparked interest in the scientific community, leading to the publication of numerous studies in recent years. In this review, we summarize the most recent progress over the past two years in the identification and development of synthetic small-molecule inhibitors of SARS-CoV-2 Mpro.
Collapse
Affiliation(s)
- Laura Braconi
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Alice Sosic
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy.
| | - Letizia Crocetti
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy.
| |
Collapse
|
31
|
Goldmann O, Medina E. Revisiting Pathogen Exploitation of Clathrin-Independent Endocytosis: Mechanisms and Implications. Cells 2025; 14:731. [PMID: 40422234 DOI: 10.3390/cells14100731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/04/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025] Open
Abstract
Endocytosis is a specialized transport mechanism in which the cell membrane folds inward to enclose large molecules, fluids, or particles, forming vesicles that are transported within the cell. It plays a crucial role in nutrient uptake, immune responses, and cellular communication. However, many pathogens exploit the endocytic pathway to invade and survive within host cells, allowing them to evade the immune system and establish infection. Endocytosis can be classified as clathrin-mediated (CME) or clathrin-independent (CIE), based on the mechanism of vesicle formation. Unlike CME, which involves the formation of clathrin-coated vesicles that bud from the plasma membrane, CIE does not rely on clathrin-coated vesicles. Instead, other mechanisms facilitate membrane invagination and vesicle formation. CIE encompasses a variety of pathways, including caveolin-mediated, Arf6-dependent, and flotillin-dependent pathways. In this review, we discuss key features of CIE pathways, including cargo selection, vesicle formation, routes taken by internalized cargo, and the regulatory mechanisms governing CIE. Many viruses and bacteria hijack host cell CIE mechanisms to facilitate intracellular trafficking and persistence. We also revisit the exploitation of CIE by bacterial and viral pathogens, highlighting recent discoveries in entry mechanisms, intracellular fate, and host-pathogen interactions. Understanding how pathogens manipulate CIE in host cells can inform the development of novel antimicrobial and immunomodulatory interventions, offering new avenues for disease prevention and treatment.
Collapse
Affiliation(s)
- Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| |
Collapse
|
32
|
Sun J, Sun D, Yang Q, Wang D, Peng J, Guo H, Ding X, Chen Z, Yuan B, Ivanenkov YA, Yuan J, Zagribelnyy BA, He Y, Su J, Wang L, Tang J, Li Z, Li R, Li T, Hu X, Liang X, Zhu A, Wei P, Fan Y, Liu S, Zheng J, Guan X, Aliper A, Yang M, Bezrukov DS, Xie Z, Terentiev VA, Peng G, Polykovskiy DA, Malyshev AS, Malkov MN, Zhu Q, Aspuru-Guzik A, Ding X, Cai X, Zhang M, Zhao J, Zhong N, Ren F, Chen X, Zhavoronkov A, Zhao J. A novel, covalent broad-spectrum inhibitor targeting human coronavirus M pro. Nat Commun 2025; 16:4546. [PMID: 40374668 PMCID: PMC12081877 DOI: 10.1038/s41467-025-59870-4] [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: 03/04/2025] [Accepted: 05/06/2025] [Indexed: 05/17/2025] Open
Abstract
Human coronaviruses (CoV) cause respiratory infections that range from mild to severe. CoVs are a large family of viruses with considerable genetic heterogeneity and a multitude of viral types, making preventing and treating these viruses difficult. Comprehensive treatments that inhibit CoV infections fulfill a pressing medical need and may be immensely valuable in managing emerging and endemic CoV infections. As the main protease (Mpro) is highly conserved across many CoVs, this protease has been identified as a route for broad CoV inhibition. We utilize the advanced generative chemistry platform Chemistry42 for de novo molecular design and obtained novel small-molecule, non-peptide-like inhibitors targeting the SARS-CoV-2 Mpro. ISM3312 is identified as an irreversible, covalent Mpro inhibitor from extensive virtual screening and structure-based optimization efforts. ISM3312 exhibits low off-target risk and outstanding antiviral activity against multiple human coronaviruses, including SARS-CoV-2, MERS-CoV, 229E, OC43, NL63, and HKU1 independent of P-glycoprotein (P-gp) inhibition. Furthermore, ISM3312 shows significant inhibitory effects against Nirmatrelvir-resistant Mpro mutants, suggesting ISM3312 may contribute to reduced viral escape in these settings. Incorporating ISM3312 and Nirmatrelvir into antiviral strategy could improve preparedness and reinforce defenses against future coronavirus threats.
Collapse
Affiliation(s)
- Jing Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Deheng Sun
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China
| | - Qi Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, Guangdong Province, 510005, China
| | - Dong Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Jingjing Peng
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China
| | - Hu Guo
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Xiaoyu Ding
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China
| | - Zhao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Bin Yuan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Yan A Ivanenkov
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Jinwei Yuan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Bogdan A Zagribelnyy
- Insilico Medicine AI Limited, Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE
| | - Yiyun He
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Jingyi Su
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Ling Wang
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China
| | - Jielin Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, Guangdong Province, 510005, China
| | - Zhun Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Rong Li
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, Shanghai Tech University, Shanghai, 201210, China
| | - Taotao Li
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China
| | - Xiaoyu Hu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Xing Liang
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Peilan Wei
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Yaya Fan
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China
| | - Sang Liu
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China
| | - Jie Zheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Xin Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Alex Aliper
- Insilico Medicine AI Limited, Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE
| | - Minglei Yang
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, Shanghai Tech University, Shanghai, 201210, China
| | - Dmitry S Bezrukov
- Insilico Medicine AI Limited, Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE
| | - Zhanhong Xie
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Victor A Terentiev
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Guilin Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China
| | - Daniil A Polykovskiy
- Insilico Medicine Canada Inc., 3710-1250 Ren´e-L´evesque west, Montreal, QC, H3B 4W8, Canada
| | - Alexander S Malyshev
- Insilico Medicine Hong Kong Ltd., Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Maxim N Malkov
- Insilico Medicine AI Limited, Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE
| | - Qingsong Zhu
- Insilico Medicine AI Limited, Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE
| | - Alán Aspuru-Guzik
- Department of Chemistry, Department of Computer Science, University of Toronto, Vector Institute for Artificial Intelligence, Canadian Institute for Advanced Research, Toronto, ON, M5S 3H6, Canada
| | - Xiao Ding
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China
| | - Xin Cai
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China
| | - Man Zhang
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China
| | - Jingxian Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China.
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China.
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, Guangdong Province, 510005, China.
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China.
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, Guangdong Province, 510005, China.
| | - Feng Ren
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China.
| | - Xinwen Chen
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, Guangdong Province, 510005, China.
| | - Alex Zhavoronkov
- Insilico Medicine Shanghai Ltd, Suite 902, Tower C, Changtai Plaza, 2889 Jinke Road, Pudong New District, Shanghai, 201203, China.
- Insilico Medicine AI Limited, Level 6, Unit 08, Block A, IRENA HQ Building, Masdar City, Abu Dhabi, UAE.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Centre for Respiratory Disease, National Centre for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China.
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong Province, 510182, China.
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, Guangdong Province, 510005, China.
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, Shanghai Tech University, Shanghai, 201210, China.
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, 518005, China.
| |
Collapse
|
33
|
de Morais Gomes V, Santos DM, Macedo-da-Silva J, Lazari LC, Machado RRG, Dos Santos AF, Araujo DB, Coutinho JVP, Arini GS, Angeli CB, de Souza EE, Marques RF, Boscardin SB, Wrenger C, Marinho CRF, Oliveira DBL, Durigon EL, Labriola L, Rosa-Fernandes L, Palmisano G. P.1 and P.2 SARS-CoV-2 Brazilian variants activate the unfolded protein response with a time and pathway specificity. J Proteomics 2025; 315:105397. [PMID: 39909104 DOI: 10.1016/j.jprot.2025.105397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/07/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
COVID-19 is a human respiratory syndrome caused by the infection of the SARS-CoV-2 virus that has a high rate of infection and mortality. Viruses modulate the host machinery by altering cellular mechanisms that favor their replication. One of the mechanisms that viruses exploit is the protein folding and processing of post-translational modifications that occur in the endoplasmic reticulum (ER). When ER function is impaired, there is an accumulation of misfolded proteins leading to endoplasmic reticulum stress (ER stress). To maintain homeostasis, cells trigger an adaptive signaling mechanism called the Unfolded Protein Response (UPR) which helps cells deal with stress, but under severe conditions, can activate the apoptotic cell death mechanism. This study elucidated an activation of a diversity of molecular mechanisms by Brazilian variants of SARS-CoV-2 by a time-resolved and large-scale characterization of SARS-CoV-2-infected cells proteomics and immunoblotting. Furthermore, it was shown that pharmacological UPR modulation could reduce viral release by counteracting the different viral activations of its cellular response. Analysis of human clinical specimens and disease outcomes focusing on ER stress reinforces the importance of UPR modulation as a host regulatory mechanism during viral infection and could point to novel therapeutic targets. SIGNIFICANCE: Since the emergence of SARS-CoV-2 and the consequent COVID-19 pandemic, the rapid emergence of variants of this new coronavirus has been a cause for concern since many of them have significantly higher rates of transmissibility and virulence, being called Variants of Concern (VOC). In this work, we studied the VOCs Gamma (P.1) and Zeta (P.2), also known as Brazilian variants. Constant evidence has reported that there are particularities related to each variant of SARS-CoV-2, with different rates of transmissibility, replication and modulation of host biological processes being observed, in addition to the mutations present in the variants. For this reason, this work focused on infections caused by the Brazilian variants of SARS-CoV-2 in different cell lines, in which we were able to observe that the infections caused by the variants induced endoplasmic reticulum stress in the infected cells and activated the UPR pathways, presenting specific modulations of each variant in this pathway. Furthermore, transcriptome analysis of patients revealed a correlation between ER-related genes and COVID-19 progression. Finally, we observed that the use of UPR modulators in host cells decreased viral release of all variants without affecting cell viability. The data presented in this work complement the observations of other studies that aim to understand the pathogenicity of SARS-CoV-2 VOCs and possible new therapeutic strategies, mainly targeting biological processes related to the endoplasmic reticulum.
Collapse
Affiliation(s)
| | - Deivid Martins Santos
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Janaina Macedo-da-Silva
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Lucas C Lazari
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | | | | | - Danielle Bastos Araujo
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil
| | | | - Gabriel Santos Arini
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Brazil
| | - Claudia B Angeli
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Edmarcia E de Souza
- Unit for Drug Discovery, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Rodolfo F Marques
- Laboratory of Antigen Targeting for Dendritic Cells, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Laboratory of Antigen Targeting for Dendritic Cells, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, ICB, University of São Paulo, Brazil
| | | | - Danielle B L Oliveira
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil
| | - Edison L Durigon
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil; Scientific Platform Pasteur USP, Sao Paulo, Brazil
| | - Leticia Labriola
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Brazil
| | - Livia Rosa-Fernandes
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil; Laboratory of Experimental Immunoparasitology, Department of Parasitology, ICB, University of São Paulo, Brazil; Centre for Motor Neuron Disease Research, Faculty of Medicine, Health & Human Sciences, Macquarie Medical School, Sydney, Australia
| | - Giuseppe Palmisano
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil; School of Natural Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|
34
|
Loonen S, van Steenis L, Bauer M, Šoštarić N. Phosphorylation Changes SARS-CoV-2 Nucleocapsid Protein's Structural Dynamics and Its Interaction With RNA. Proteins 2025. [PMID: 40375582 DOI: 10.1002/prot.26842] [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: 11/05/2024] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/18/2025]
Abstract
The SARS-CoV-2 nucleocapsid protein, or N-protein, is a structural protein that plays an important role in the SARS-CoV-2 life cycle. The N-protein takes part in the regulation of viral RNA replication and drives highly specific packaging of full-length genomic RNA prior to virion formation. One regulatory mechanism that is proposed to drive the switch between these two operating modes is the phosphorylation state of the N-protein. Here, we assess the dynamic behavior of non-phosphorylated and phosphorylated versions of the N-protein homodimer through atomistic molecular dynamics simulations. We show that the introduction of phosphorylation yields a more dynamic protein structure and decreases the binding affinity between the N-protein and RNA. Furthermore, we find that secondary structure is essential for the preferential binding of particular RNA elements from the 5' UTR of the viral genome to the N-terminal domain of the N-protein. Altogether, we provide detailed molecular insights into N-protein dynamics, N-protein:RNA interactions, and phosphorylation. Our results corroborate the hypothesis that phosphorylation of the N-protein serves as a regulatory mechanism that determines N-protein function.
Collapse
Affiliation(s)
- Stefan Loonen
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, HZ, the Netherlands
| | - Lina van Steenis
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, HZ, the Netherlands
| | - Marianne Bauer
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, HZ, the Netherlands
| | - Nikolina Šoštarić
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, HZ, the Netherlands
| |
Collapse
|
35
|
Chen Y, Klute S, Sparrer KMJ, Serra-Moreno R. RAB5 is a host dependency factor for the generation of SARS-CoV-2 replication organelles. mBio 2025; 16:e0331424. [PMID: 40167317 PMCID: PMC12077180 DOI: 10.1128/mbio.03314-24] [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: 10/23/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains a threat due to the emergence of variants with increased transmissibility and enhanced escape from immune responses. Like other coronaviruses before, SARS-CoV-2 likely emerged after its transmission from bats. The successful propagation of SARS-CoV-2 in humans might have been facilitated by usurping evolutionarily conserved cellular factors to execute crucial steps in its life cycle, such as the generation of replication organelles-membrane structures where coronaviruses assemble their replication-transcription complex. In this study, we found that RAB5, which is highly conserved across mammals, is a critical host dependency factor for the replication of the SARS-CoV-2 genome. Our results also suggest that SARS-CoV-2 uses RAB5+ membranes to build replication organelles with the aid of COPB1, a component of the COP-I complex, and that the virus protein NSP6 participates in this process. Hence, targeting NSP6 represents a promising approach to interfere with SARS-CoV-2 RNA synthesis and halt its propagation.IMPORTANCEIn this study, we sought to identify the host dependency factors that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses for the generation of replication organelles: cellular membranous structures that SARS-CoV-2 builds in order to support the replication and transcription of its genome. We uncovered that RAB5 is an important dependency factor for SARS-CoV-2 replication and the generation of replication organelles, and that the viral protein NSP6 participates in this process. Hence, NSP6 represents a promising target to halt SARS-CoV-2 replication.
Collapse
Affiliation(s)
- Yuexuan Chen
- Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Susanne Klute
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Konstantin Maria Johannes Sparrer
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Ruth Serra-Moreno
- Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
36
|
Freiberg C, Dotan A, Arnheim D, Aviel YB. Investigating the association between SARS-CoV-2 infection, COVID-19 vaccination, and autoimmune diseases in a pediatric population: a comprehensive analysis. Pediatr Rheumatol Online J 2025; 23:52. [PMID: 40369546 PMCID: PMC12080261 DOI: 10.1186/s12969-025-01093-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND During the COVID-19 pandemic there were reports of an increased association between COVID 19 and various autoimmune diseases (AID) in adults. This study aims to investigate the incidence of AIDs in children before and during the pandemic and explores potential links to SARS-CoV-2 vaccination. METHODS We analyzed 493,705 anonymized medical records from Maccabi Healthcare Services, Israel's second-largest healthcare provider, to study AID incidence during 2014-2022. The study period was divided into three phases: two pre-pandemic phases of equal duration (A and B) and a pandemic phase (C). RESULTS Of 4,596 (0.9%) patients diagnosed with an AID in the cohort, incidence rates were 0.9% for Group A (2014-2016), 1.0% for Group B (2017-2019), and 0.9% for Group C (2020-2022) (p = 0.13). Logistic regression showed no significant differences in overall autoimmune disease incidence between the pre-COVID and COVID periods. Notably, specific conditions like celiac disease showed reduced incidence in Group A (OR 0.8309, p = 0.0071) while arthritis was significantly more common in Groups A and B. Additionally, COVID-19 diagnosis was not significantly associated with increased autoimmune disease risk (HR 1.092, p = 0.491); however, receiving at least one COVID vaccine was linked to higher risk (HR 1.2323, p = 0.0033). CONCLUSION Our findings suggest that the overall incidence of new-onset autoimmune diseases in children remained relatively stable during the COVID-19 pandemic. The study indicates a potential association between COVID-19 vaccination and an increased risk of developing autoimmune diseases, necessitating further research to elucidate long-term effects in the pediatric population.
Collapse
Affiliation(s)
| | - Arad Dotan
- Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
- Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Dana Arnheim
- Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
- Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Yonatan Butbul Aviel
- Department of Pediatrics and Pediatric Rheumatology Service, Rambam Health Care Campus, Ruth Children's Hospital, Haifa, Israel
| |
Collapse
|
37
|
Aierken D, Zhang V, Sealfon R, Marecki JC, Raney KD, Gladfelter AS, Joseph JA, Roden CA. Biomolecular condensates control and are defined by RNA-RNA interactions that arise in viral replication. RESEARCH SQUARE 2025:rs.3.rs-6378534. [PMID: 40470224 PMCID: PMC12136198 DOI: 10.21203/rs.3.rs-6378534/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2025]
Abstract
Cells must limit RNA-RNA interactions to avoid irreversible RNA entanglement. Cells may prevent deleterious RNA-RNA interactions by genome organization to avoid complementarity however, RNA viruses generate long, perfectly complementary antisense RNA during replication. How do viral RNAs avoid irreversible entanglement? One possibility is RNA sequestration into biomolecular condensates. To test this, we reconstituted critical SARS-CoV-2 RNA-RNA interactions in Nucleocapsid condensates. We observed that RNAs with low propensity RNA-RNA interactions resulted in more round, liquid-like condensates while those with high sequence complementarity resulted in more heterogeneous networked morphology independent of RNA structure stability. Residue-resolution molecular simulations and direct sequencing-based detection of RNA-RNA interactions support that these properties arise from degree of trans RNA contacts. We propose that extensive RNA-RNA interactions in cell and viral replication are controlled via a combination of genome organization, timing, RNA sequence content, RNA production ratios, and emergent biomolecular condensate material properties.
Collapse
Affiliation(s)
- Dilimulati Aierken
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, USA
- Omenn–Darling Bioengineering Institute, Princeton University, Princeton, New Jersey, USA
| | - Vita Zhang
- Department of Cell Biology, Duke University, Durham, North Carolina, USA
- Department of Biochemistry, Duke University, Durham, North Carolina, USA
| | - Rachel Sealfon
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | - John C. Marecki
- Department of Biochemistry, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kevin D. Raney
- Department of Biochemistry, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Amy S. Gladfelter
- Department of Cell Biology, Duke University, Durham, North Carolina, USA
| | - Jerelle A. Joseph
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey, USA
- Omenn–Darling Bioengineering Institute, Princeton University, Princeton, New Jersey, USA
| | - Christine A. Roden
- Department of Cell Biology, Duke University, Durham, North Carolina, USA
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC, Canada
- Lead contact
| |
Collapse
|
38
|
Tao X, Wang Y, Jin J, Yan H, Yang H, Wan X, Li P, Xiao Y, Yu Q, Liu L, Liu Y, Han T, Zhang W. NSP6 regulates calcium overload-induced autophagic cell death and is regulated by KLHL22-mediated ubiquitination. J Adv Res 2025:S2090-1232(25)00350-9. [PMID: 40373961 DOI: 10.1016/j.jare.2025.05.031] [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: 01/11/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025] Open
Abstract
INTRODUCTION Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a substantial global threat. SARS-CoV-2 nonstructural proteins (NSPs) are essential for impeding the host replication mechanism while also assisting in the production and organization of new viral components. However, NSPs are not incorporated into viral particles, and their subsequent fate within host cells remains poorly understood. Additionally, their role in viral pathogenesis requires further investigation. OBJECTIVES This study aimed to discover the ultimate fate of NSP6 in host cells and to elucidate its role in viral pathogenesis. METHODS We investigated the effects of NSP6 on cell death and explored the underlying mechanism; moreover, we examined the degradation mechanism of NSP6 in human cells, along with analysing its correlation with coronavirus disease 2019 (COVID-19) severity in patient peripheral blood mononuclear cells (PBMCs). RESULTS NSP6 was demonstrated to induce cell death. Specifically, NSP6 interacted with EI24 autophagy-associated transmembrane protein (EI24) to increase intracellular Ca2+ levels, thereby enhancing the interactions between unc-51-like autophagy activating kinase 1 (ULK1) and RB1 inducible coiled-coil 1 (RB1CC1/FIP200), as well as beclin 1 (BECN1) and phosphatidylinositol 3-kinase catalytic subunit type 3 (PIK3C3). This cascade ultimately triggers autophagy, thus resulting in cell death. Additionally, we discovered that the homeostasis of the NSP6 protein was regulated by K48-linked ubiquitination. We identified kelch-like protein 22 (KLHL22) as the E3 ligase that was responsible for ubiquitinating and degrading NSP6, restoring intracellular calcium homeostasis and reversing NSP6-induced autophagic cell death. Moreover, NSP6 expression levels were observed to be positively associated with the severity of SARS-CoV-2-induced disease. CONCLUSION This study reveals that KLHL22-mediated ubiquitination controls NSP6 stability and that NSP6 induces autophagic cell death via calcium overload, highlighting its cytotoxic role and suggesting therapeutic strategies that target calcium signaling or promote NSP6 degradation as potential interventions against COVID-19.
Collapse
Affiliation(s)
- Xingyu Tao
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Yanan Wang
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Jiangbo Jin
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China
| | - Huilin Yan
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Hui Yang
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Xiaorui Wan
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Ping Li
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Yanghua Xiao
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Qi Yu
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Lingjiao Liu
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China
| | - Yang Liu
- China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China; Department of Clinical Microbiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China.
| | - Tianyu Han
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China.
| | - Wei Zhang
- Jiangxi Institute of Respiratory Disease, The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang City 330006 Jiangxi, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang City 330006 Jiangxi, China; China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang City 330200 Jiangxi, China.
| |
Collapse
|
39
|
Tian L, Zhao Z, Gao W, Liu Z, Li X, Zhang W, Li Z. SARS-CoV-2 nsp16 is regulated by host E3 ubiquitin ligases, UBR5 and MARCHF7. eLife 2025; 13:RP102277. [PMID: 40358464 PMCID: PMC12074641 DOI: 10.7554/elife.102277] [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] [Indexed: 05/15/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), remains a global public health threat with considerable economic consequences. The nonstructural protein 16 (nsp16), in complex with nsp10, facilitates the final viral mRNA capping step through its 2'-O-methylase activity, helping the virus to evade host immunity and prevent mRNA degradation. However, nsp16 regulation by host factors remains poorly understood. While various E3 ubiquitin ligases interact with SARS-CoV-2 proteins, their roles in targeting nsp16 for degradation remain unclear. In this study, we demonstrate that nsp16 undergoes ubiquitination and proteasomal degradation mediated by the host E3 ligases UBR5 and MARCHF7. UBR5 induces K48-linked ubiquitination, whereas MARCHF7 promotes K27-linked ubiquitination, independently suppressing SARS-CoV-2 replication in cell cultures and in mice. Notably, UBR5 and MARCHF7 also degrade nsp16 variants from different viral strains, exhibiting broad-spectrum antiviral activity. Our findings reveal novel antiviral mechanisms of the ubiquitin-proteasome system (UPS) and highlight their potential therapeutic targets against COVID-19.
Collapse
Affiliation(s)
- Li Tian
- Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin UniversityChangchunChina
| | - Zongzheng Zhao
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChangchunChina
| | - Wenying Gao
- Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin UniversityChangchunChina
| | - Zirui Liu
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChangchunChina
| | - Xiao Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural SciencesChangchunChina
| | - Wenyan Zhang
- Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin UniversityChangchunChina
| | - Zhaolong Li
- Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
40
|
Hartmann S, Radochonski L, Ye C, Martinez-Sobrido L, Chen J. SARS-CoV-2 ORF3a drives dynamic dense body formation for optimal viral infectivity. Nat Commun 2025; 16:4393. [PMID: 40355429 PMCID: PMC12069715 DOI: 10.1038/s41467-025-59475-x] [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: 05/08/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
SARS-CoV-2 hijacks multiple organelles for virion assembly, of which the mechanisms have not been fully understood. Here, we identified a SARS-CoV-2-driven membrane structure named the 3a dense body (3DB). 3DBs are unusual electron-dense and dynamic structures driven by the accessory protein ORF3a via remodeling a specific subset of the trans-Golgi network (TGN) and early endosomal membrane. 3DB formation is conserved in related bat and pangolin coronaviruses but was lost during the evolution to SARS-CoV. During SARS-CoV-2 infection, 3DB recruits the viral structural proteins spike (S) and membrane (M) and undergoes dynamic fusion/fission to maintain the optimal unprocessed-to-processed ratio of S on assembled virions. Disruption of 3DB formation resulted in virions assembled with an abnormal S processing rate, leading to a dramatic reduction in viral entry efficiency. Our study uncovers the crucial role of 3DB in maintaining maximal SARS-CoV-2 infectivity and highlights its potential as a target for COVID-19 prophylactics and therapeutics.
Collapse
Affiliation(s)
- Stella Hartmann
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Lisa Radochonski
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Jueqi Chen
- Department of Microbiology, University of Chicago, Chicago, IL, USA.
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA.
| |
Collapse
|
41
|
Pang X, Xu W, Liang J, Liu Y, Li H, Chen L. Research progress and perspectives of dual-target inhibitors. Eur J Med Chem 2025; 289:117453. [PMID: 40024166 DOI: 10.1016/j.ejmech.2025.117453] [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: 12/07/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
The occurrence and development of diseases are complex, and single-target drugs that affect only a single target or pathway often fail to achieve the expected therapeutic effect. The simultaneous effect on two key targets could not only increase patient tolerance but also accelerate disease remission. Dual-target inhibitors have already been studied the most intensively in the development of dual-target drugs. This article briefly introduces the function of drug therapy targets, and mainly summarizes the design strategies and research progress of dual-target inhibitors in neurodegenerative diseases, infectious diseases, metabolic diseases and cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaojing Pang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Wen Xu
- Institute of Structural Pharmacology & TCM Chemical Biology, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Jing Liang
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yang Liu
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hua Li
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China; Institute of Structural Pharmacology & TCM Chemical Biology, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China.
| | - Lixia Chen
- Wuya College of Innovation, Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
42
|
Sentis C, Parraud D, Billaud G, Valette M, Bouscambert‐Duchamp M, Lina B, Morfin F, Gaymard A. Performance of Subgenomic RT-PCR for Predicting SARS-CoV-2 Infectivity Compared to Genomic RT-PCR and Culture Isolation. J Med Virol 2025; 97:e70363. [PMID: 40272020 PMCID: PMC12020333 DOI: 10.1002/jmv.70363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/21/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
SARS-CoV-2 clinical samples can be detected as positive for a long period of time using real-time RT-PCR, even when patients are no longer infectious. Viral culture is the gold standard for assessing a patient's infectivity, but it is a time-consuming technique and lacks sensitivity. SARS-CoV-2 subgenomic RNA (sgRNA) detection has been used as a proxy for assessing the infectivity but only a limited number of studies have described its use in vitro and in clinical samples. This study aimed to evaluate the correlation between results from viral culture, genomic RT-PCR (gRT-PCR), and subgenomic RT-PCR (sgRT-PCR) during in vitro infection and in clinical samples. In vitro viral replication kinetics showed that both genomic RNA (gRNA) and subgenomic RNA (sgRNA) levels remained stable up to 21 days in the absence of replication-competent virus. Using clinical samples, sgRNA was detected in 87.5% of culture-positive samples, demonstrating better performances than gRT-PCR (Positive predictive value (PPV) 93.3% and Negative predictive value (NPV) of 87.5%) and an almost perfect agreement with culture results (Cohen κ = 0.81 [95% CI: 0.66-0.95]). These findings suggest that testing for sgRNA and/or using a gRNA Ct cut-off of 21.2 could be used as a proxy to determine the presence of SARS-CoV-2 replication-competent virus.
Collapse
Affiliation(s)
- Célia Sentis
- Hospices Civils de Lyon, Institut des Agents Infectieux, service de Virologie, Laboratoire Associé au Centre National de Référence des Virus des Infections RespiratoiresLyonFrance
| | - Delphine Parraud
- Hospices Civils de Lyon, Institut des Agents Infectieux, service de Virologie, Laboratoire Associé au Centre National de Référence des Virus des Infections RespiratoiresLyonFrance
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1LyonFrance
| | - Geneviève Billaud
- Hospices Civils de Lyon, Institut des Agents Infectieux, service de Virologie, Laboratoire Associé au Centre National de Référence des Virus des Infections RespiratoiresLyonFrance
| | - Martine Valette
- Hospices Civils de Lyon, Institut des Agents Infectieux, service de Virologie, Laboratoire Associé au Centre National de Référence des Virus des Infections RespiratoiresLyonFrance
| | - Maude Bouscambert‐Duchamp
- Hospices Civils de Lyon, Institut des Agents Infectieux, service de Virologie, Laboratoire Associé au Centre National de Référence des Virus des Infections RespiratoiresLyonFrance
| | - Bruno Lina
- Hospices Civils de Lyon, Institut des Agents Infectieux, service de Virologie, Laboratoire Associé au Centre National de Référence des Virus des Infections RespiratoiresLyonFrance
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1LyonFrance
| | - Florence Morfin
- Hospices Civils de Lyon, Institut des Agents Infectieux, service de Virologie, Laboratoire Associé au Centre National de Référence des Virus des Infections RespiratoiresLyonFrance
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1LyonFrance
| | - Alexandre Gaymard
- Hospices Civils de Lyon, Institut des Agents Infectieux, service de Virologie, Laboratoire Associé au Centre National de Référence des Virus des Infections RespiratoiresLyonFrance
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1LyonFrance
| |
Collapse
|
43
|
Venkataraman S, Savithri HS, Murthy MRN. Recent advances in the structure and assembly of non-enveloped spherical viruses. Virology 2025; 606:110454. [PMID: 40081202 DOI: 10.1016/j.virol.2025.110454] [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: 11/11/2024] [Revised: 02/03/2025] [Accepted: 02/12/2025] [Indexed: 03/15/2025]
Abstract
Non-enveloped spherical viruses (NSVs) are characterized by their highly symmetrical capsids that serve to protect and encapsulate the genomes. The stability and functionality of the capsids determine their ability for survival and proliferation in harsh environments. Over four decades of structural studies using X-ray crystallography and NMR have provided static, high-resolution snapshots of several viruses. Recently, advances in cryo-electron microscopy, together with AI-based structure predictions and traditional methods, have aided in elucidating not only the structural details of complex NSVs but also the mechanistic processes underlying their assembly. The knowledge thus generated has been instrumental in critical understanding of the conformational changes and interactions associated with the coat proteins, the genome, and the auxiliary factors that regulate the capsid dynamics. This review seeks to summarize current literature regarding the structure and assembly of the NSVs and discusses how the data has facilitated a deeper understanding of their biology and phylogeny.
Collapse
Affiliation(s)
| | | | - M R N Murthy
- Indian Institute of Science, Bengaluru, 560012, India.
| |
Collapse
|
44
|
Proal AD, Aleman S, Bomsel M, Brodin P, Buggert M, Cherry S, Chertow DS, Davies HE, Dupont CL, Deeks SG, Ely EW, Fasano A, Freire M, Geng LN, Griffin DE, Henrich TJ, Hewitt SM, Iwasaki A, Krumholz HM, Locci M, Marconi VC, Mehandru S, Muller-Trutwin M, Painter MM, Pretorius E, Price DA, Putrino D, Qian Y, Roan NR, Salmon D, Tan GS, VanElzakker MB, Wherry EJ, Van Weyenbergh J, Yonker LM, Peluso MJ. Targeting the SARS-CoV-2 reservoir in long COVID. THE LANCET. INFECTIOUS DISEASES 2025; 25:e294-e306. [PMID: 39947217 DOI: 10.1016/s1473-3099(24)00769-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/28/2024] [Accepted: 11/12/2024] [Indexed: 03/15/2025]
Abstract
There are no approved treatments for post-COVID-19 condition (also known as long COVID), a debilitating disease state following SARS-CoV-2 infection that is estimated to affect tens of millions of people. A growing body of evidence shows that SARS-CoV-2 can persist for months or years following COVID-19 in a subset of individuals, with this reservoir potentially driving long-COVID symptoms or sequelae. There is, therefore, an urgent need for clinical trials targeting persistent SARS-CoV-2, and several trials of antivirals or monoclonal antibodies for long COVID are underway. However, because mechanisms of SARS-CoV-2 persistence are not yet fully understood, such studies require important considerations related to the mechanism of action of candidate therapeutics, participant selection, duration of treatment, standardisation of reservoir-associated biomarkers and measurables, optimal outcome assessments, and potential combination approaches. In addition, patient subgroups might respond to some interventions or combinations of interventions, making post-hoc analyses crucial. Here, we outline these and other key considerations, with the goal of informing the design, implementation, and interpretation of trials in this rapidly growing field. Our recommendations are informed by knowledge gained from trials targeting the HIV reservoir, hepatitis C, and other RNA viruses, as well as precision oncology, which share many of the same hurdles facing long-COVID trials.
Collapse
Affiliation(s)
- Amy D Proal
- PolyBio Research Foundation, Medford, MA, USA.
| | - Soo Aleman
- Department of Infectious Diseases and Unit of Post-COVID Huddinge, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Morgane Bomsel
- HIV entry and Laboratory of Mucosal Immunity, Institut Cochin, Paris, France; Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Petter Brodin
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Department of Immunology and Inflammation, Imperial College London, London, UK; Medical Research Council Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Marcus Buggert
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel S Chertow
- Emerging Pathogens Section, Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA; Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Helen E Davies
- Department of Respiratory Medicine, University Hospital Llandough, Cardiff, UK; University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - Christopher L Dupont
- Division of Genomic Medicine, Environment & Sustainability, J Craig Venter Institute, University of California San Diego, La Jolla, CA, USA
| | - Steven G Deeks
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA
| | - E Wes Ely
- The Critical Illness, Brain Dysfunction, Survivorship Center at Vanderbilt University Medical Center, Nashville, TN, USA; Veteran's Affairs Tennessee Valley Geriatric Research Education Clinical Center, Nashville, TN, USA
| | - Alessio Fasano
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA; Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Marcelo Freire
- Department of Infectious Diseases, J Craig Venter Institute, University of California San Diego, La Jolla, CA, USA
| | - Linda N Geng
- J Craig Venter Institute, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Diane E Griffin
- W Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California, San Francisco, CA, USA
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Center for Infection and Immunity, Yale University School of Medicine, New Haven, CT, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Harlan M Krumholz
- Center for Infection and Immunity, Yale University School of Medicine, New Haven, CT, USA; Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, CT, USA; Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Health Policy and Management, Yale School of Public Health, New Haven, CT, USA
| | - Michela Locci
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vincent C Marconi
- Emory University School of Medicine and Rollins School of Public Health, Atlanta, GA, USA; Atlanta Veterans Affairs Medical Center, Decatur, GA, USA
| | - Saurabh Mehandru
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Henry D Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michaela Muller-Trutwin
- Institut Pasteur, Université Paris-Cité, HIV, Inflammation and Persistence Unit, Paris, France
| | - Mark M Painter
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa; Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, UK
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK; Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - David Putrino
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yu Qian
- Department of Informatics, J Craig Venter Institute, University of California San Diego, La Jolla, CA, USA
| | - Nadia R Roan
- Gladstone Institutes, University of California, San Francisco, CA, USA; Department of Urology, University of California, San Francisco, CA, USA
| | - Dominique Salmon
- Department of Infectious Diseases, Institut Fournier, Paris, France; Direction of International Relations Assistance Publique Hôpitaux de Paris, Paris, France
| | - Gene S Tan
- Department of Infectious Diseases, J Craig Venter Institute, University of California San Diego, La Jolla, CA, USA
| | - Michael B VanElzakker
- PolyBio Research Foundation, Medford, MA, USA; Division of Neurotherapeutics, Massachusetts General Hospital, Boston, MA, USA
| | - E John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Johan Van Weyenbergh
- Laboratory of Clinical and Epidemiological Virology, Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Lael M Yonker
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA; Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Michael J Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
45
|
Yu N, Huang YY, Feng L, Liu Q. Coronavirus disease 2019-associated liver injury in pregnancy: A case report. J Int Med Res 2025; 53:3000605251344150. [PMID: 40433845 PMCID: PMC12120285 DOI: 10.1177/03000605251344150] [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: 02/15/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Liver injury is an increasingly recognized extrapulmonary manifestation of coronavirus disease 2019 (COVID-19), particularly during pregnancy due to overlapping physiological and pathological changes. A 26-year-old primigravida at 24+4 weeks of gestation presented with fever, chills, weakness, and sore throat, who was later confirmed to be severe acute respiratory syndrome coronavirus 2-positive. Laboratory tests revealed lymphopenia, elevated liver enzymes, low platelet count, increased D-dimer level, high total bilirubin level, and elevated lactate dehydrogenase levels. After thorough evaluation, these findings were attributed to COVID-19-associated liver injury. The patient was provided with supportive care and symptomatic treatment, resulting in gradual normalization of liver function. She was discharged at 26 weeks of gestation in stable condition. This case highlights the importance of considering COVID-19-associated liver injury in pregnant patients presenting with hepatic dysfunction, where prompt recognition and conservative management can help achieve favorable maternal outcomes.
Collapse
Affiliation(s)
- Nan Yu
- Gynaecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Yi-Yao Huang
- Gynaecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Ling Feng
- Gynaecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Qian Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, China
| |
Collapse
|
46
|
Zhang M, Zhu Y, Li N, Aishanjiang K, Zhu S, Tang A, Li G, Liu G. Development of a monoclonal antibody-based colloidal gold immunochromatographic strip for rapid detection of feline coronavirus. Int J Biol Macromol 2025; 309:142683. [PMID: 40169048 DOI: 10.1016/j.ijbiomac.2025.142683] [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: 12/24/2024] [Revised: 03/02/2025] [Accepted: 03/28/2025] [Indexed: 04/03/2025]
Abstract
Feline infectious peritonitis (FIP), caused by feline coronavirus (FCoV), is a fatal disease with no effective vaccine. Early detection is crucial for FIP management, and a rapid, accurate diagnostic method is urgently needed. Hence, the purpose of this study was to establish a rapid, sensitive, specific immunochromatographic strip (ICS) for clinical detection of FIP. We selected the highly conserved N protein of FIPV and expressed recombinant N protein as an immunogen to prepare monoclonal antibodies (mAbs). Five mAbs specific to FIPV were produced. The antigenic epitopes recognized by the 2B10 and 10E7 mAbs used for ICS preparation were identified, and the structure and conservation of the epitopes were analyzed. Subsequently, we paired the 2B10 and 10E7 mAbs, assembled the ICS, and implemented several optimization measures. The specificity of the ICS was confirmed by positive reactions with FIPV-positive samples and negative reactions with FHV, FPV, and FCV. Sensitivity testing detected FIPV suspensions (TCID₅₀ = 106.5/mL) diluted to 1: 512. The ICS showed 98.3 % agreement with RT-PCR results in detecting 60 suspected samples and remained stable for 6 months at room temperature. In conclusion, this study developed a simple, sensitive, and specific ICS for the detection of FIPV.
Collapse
Affiliation(s)
- Miao Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Yingqi Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Na Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Kelimujiang Aishanjiang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Shiqiang Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Aoxing Tang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China
| | - Guoxin Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China.
| | - Guangqing Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CAAS), Shanghai 200241, China.
| |
Collapse
|
47
|
Zhang R, Li D, Gao P, Ruan W, Qiao S, Xu S, Dai L, Luo T, Zhao X, Gao GF. A SARS-CoV and SARS-CoV-2 RBD Heterodimer Vaccine Candidate. J Med Virol 2025; 97:e70367. [PMID: 40317517 DOI: 10.1002/jmv.70367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 05/07/2025]
Abstract
The continuous evolution of SARS-CoV-2 through accumulating mutations, combined with the persistent risk of zoonotic sarbecovirus transmission events, highlights the critical demand for broadly protective vaccines. Building on our previous findings that a heterodimeric receptor-binding domain (RBD) design substantially improves cross-reactive immunogenicity in vaccine candidates, we propose this strategy as a foundation for developing pan-sarbecovirus vaccines with cross-neutralizing capacity against diverse and emerging variants. In this study, we developed a sarbecovirus immunogen, utilizing a heterodimeric strategy incorporating the RBDs from both SARS-CoV and SARS-CoV-2. Pseudovirus neutralization assays revealed that mice immunized with the SARS-CoV-2 prototype (PT)-SARS-CoV heterodimer (PT-SARS) developed 39.9- to 305.6-fold higher neutralizing antibody (NAb) titers against SARS-CoV-2 sub-variants compared to the SARS-CoV RBD homodimer (SARS-SARS). Furthermore, PT-SARS elicited 17.6- and 31.2-fold enhanced neutralization against WIV1 and SARS-CoV, respectively, relative to the SARS-CoV-2 PT homodimer (PT-PT). To address evolving Omicron sub-variants, we further updated BA.1-SARS and BA.2-SARS immunogens. Notably, BA.2-SARS exhibited a 6.2-fold increase in neutralizing potency against BA.2.86 compared to PT-SARS. Crucially, the heterodimeric immunogen induced balanced and broadly reactive NAbs against multiple sarbecoviruses, including RaTG13, Pangolin GD, SARS-CoV, and SARS-CoV-2 variants/sub-variants, demonstrating its potential as a sarbecovirus immunogen candidate.
Collapse
Affiliation(s)
- Rong Zhang
- College of Animal Sciences and Veterinary Medicine, Guangxi University (GXU), Nanning, China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Dedong Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Pengyue Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
- School of Life Science, University of Science and Technology of China (USTC), Hefei, China
- Department of Infectious Diseases, Shenzhen Children's Hospital, Shenzhen, China
| | - Wenjing Ruan
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
- School of Life Science, University of Science and Technology of China (USTC), Hefei, China
| | - Shitong Qiao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
- Beijing Life Science Academy, Beijing, China
| | - Senyu Xu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Lianpan Dai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Tingrong Luo
- College of Animal Sciences and Veterinary Medicine, Guangxi University (GXU), Nanning, China
| | - Xin Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
| | - George F Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Chinese Academy of Sciences (CAS), Beijing, China
| |
Collapse
|
48
|
Liu Y, Tang H, Xu P, Zhou X, Li S. SARS-CoV-2 N protein interacts with SLC7A11 to cause ferroptosis in acute lung injury. Allergol Immunopathol (Madr) 2025; 53:23-30. [PMID: 40342111 DOI: 10.15586/aei.v53i3.1340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 04/02/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND The nucleocapsid protein (N protein) in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is elevated in bodily fluids at the onset of infection and has recently been found to have a direct role in lung damage. However, the exact mode of action of the N protein in acute lung injury is still unknown. METHOD Recombinant N protein was used to treat mice and A549 cells in vivo and in vitro. Enzyme-linked immunosorbent assay and hematoxylin and eosin staining were used to detect the levels of inflammatory factors and lung damage in lung tissue. The total iron and Fe2+ contents and the expression of ferroptosis markers in mouse lung tissues and cells were detected. Co-immunoprecipitation detects the binding of N protein and solute carrier family 7 member 11 (SLC7A11). Replenishment experiments were conducted by activating SLC7A11 to study the effect of SLC7A11 on N protein-induced lung injury. RESULT Recombinant N protein caused acute lung injury and lung inflammation, increased total iron and Fe2+ contents in vivo and in vitro, promoted the expression of ACSL4, inhibited the expression of GPX4 and FTH1, and triggered ferroptosis. Recombinant N protein can interact with SLC7A11, and activating SLC7A11 can reverse N protein-induced ferroptosis and acute lung injury. CONCLUSION SARS-CoV-2 N protein can directly interact with SLC7A11 to cause ferroptosis, which produces a lot of inflammatory factors and results in lung injury in mice.
Collapse
Affiliation(s)
- Yi Liu
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hui Tang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Pan Xu
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xiaoqi Zhou
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shiying Li
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China;
| |
Collapse
|
49
|
van der Klugt T, van Gent M. The dynamic interactions between virus infections and nonsense-mediated decay. Hum Mol Genet 2025:ddae151. [PMID: 40292718 DOI: 10.1093/hmg/ddae151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 04/30/2025] Open
Abstract
Humans are continuously exposed to a wide array of viruses that cause a significant amount of morbidity and mortality worldwide. Over recent years, the evolutionarily conserved host RNA degradation pathway nonsense-mediated decay (NMD) has emerged as a broad antiviral defense mechanism that controls infection of a variety of RNA and DNA viruses. Besides regulating the abundance of host transcripts, NMD directly destabilizes virus genomic RNA, replication intermediates, and viral transcripts to interfere with replication. In turn, viruses have evolved strategies to modulate cellular NMD activity or repurpose NMD factors to facilitate their replication. In this review, we describe our current understanding of the role of NMD in controlling virus infections as well as the strategies employed by viruses to interfere with NMD.
Collapse
Affiliation(s)
- Teun van der Klugt
- HerpesLabNL, Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Michiel van Gent
- HerpesLabNL, Department of Viroscience, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| |
Collapse
|
50
|
Yılmaz S, Eken A, Sezer Z, Bağcı BŞ, Erdem S, Sarıkaya MD, Kaplan B, Inal A, Bayram A, Kalın Unuvar G, Zararsız G, Yerlitas Sİ, Cakir N, Pavel STI, Uygut MA, Yetiskin H, Kara A, Ozdarendeli A. Vaccination with inactivated SARS-CoV-2 vaccine TURKOVAC induces durable humoral and cellular immune responses up to 8 months. Front Med (Lausanne) 2025; 12:1524393. [PMID: 40357274 PMCID: PMC12066321 DOI: 10.3389/fmed.2025.1524393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Background The rapid spread of the SARS-CoV-2 virus has led to a global health crisis, necessitating swift responses in medical science, mainly through vaccination strategies. While short-term vaccine effectiveness is evident, immune protection's long-term effects and duration remain incompletely understood. Systematic monitoring of these responses is essential for optimizing vaccination strategies. Aims This study aimed to explore the durability of antigen-specific T and B cell responses and antibody levels up to 8 months post-immunization with the inactivated TURKOVAC vaccine in volunteers. Additionally, the impact of two versus three doses of vaccination on these parameters was analyzed. Methods Volunteers (n = 80) received two or three doses of TURKOVAC. Spike-specific B cells, CD4+ T cells, CD8+ T cells, and antibody levels were measured at multiple time points post-immunization. Results Spike-specific B cells remained elevated up to 8 months post-immunization. SARS-CoV-2-specific CD4+ and CD8+ T cells peaked at 4 months but declined thereafter. TURKOVAC resulted in durable antigen-specific humoral and cellular immune memory with distinct kinetics. Still, most assessments observed no significant differences between two and three doses, except for antigen specific-IL-2 and CD4+ LAMP1 responses. Conclusion TURKOVAC vaccination induces durable immune responses, with spike-specific B cells persisting up to 8 months and T cell responses peaking at 4 months before declining. These findings suggest that TURKOVAC contributes to long-term immune protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Seçil Yılmaz
- Genome and Stem Cell Center, Erciyes University, Kayseri, Türkiye
| | - Ahmet Eken
- Genome and Stem Cell Center, Erciyes University, Kayseri, Türkiye
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Zafer Sezer
- Department of Medical Pharmacology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
- Good Clinical Practise Centre (IKUM), Erciyes University, Kayseri, Türkiye
| | - Burcu Şen Bağcı
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, Kayseri, Türkiye
| | - Serife Erdem
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | | | - Busra Kaplan
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, Kayseri, Türkiye
- Department of Medical Microbiology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Ahmet Inal
- Department of Medical Pharmacology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
- Good Clinical Practise Centre (IKUM), Erciyes University, Kayseri, Türkiye
| | - Adnan Bayram
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Gamze Kalın Unuvar
- Infectious Diseases Clinic, Department of Infectious Diseases, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Gokmen Zararsız
- Department of Biostatistics, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Serra İlayda Yerlitas
- Department of Biostatistics, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Nuri Cakir
- Department of Medical Microbiology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | | | - Muhammet Ali Uygut
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, Kayseri, Türkiye
| | - Hazel Yetiskin
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, Kayseri, Türkiye
| | - Ates Kara
- Pediatric Infectious Department, Faculty of Medicine, Hacettepe University Hospitals, Ankara, Türkiye
| | - Aykut Ozdarendeli
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, Kayseri, Türkiye
- Department of Medical Microbiology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| |
Collapse
|