101
|
Chen J, Wang J, Yang W, Zhao L, Xu X. Identifying Inhibitor-SARS-CoV2-3CL pro Binding Mechanism Through Molecular Docking, GaMD Simulations, Correlation Network Analysis and MM-GBSA Calculations. Molecules 2025; 30:805. [PMID: 40005117 PMCID: PMC11857935 DOI: 10.3390/molecules30040805] [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: 12/23/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
The main protease of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), known as 3CLpro, is crucial in the virus's life cycle and plays a pivotal role in COVID-19. Understanding how small molecules inhibit 3CLpro's activity is vital for developing anti-COVID-19 therapeutics. To this end, we employed Gaussian accelerated molecular dynamics (GaMD) simulations to enhance the sampling of 3CLpro conformations and conducted correlation network analysis (CNA) to explore the interactions between different structural domains. Our findings indicate that a CNA-identified node in domain II of 3CLpro acts as a conduit, transferring conformational changes from the catalytic regions in domains I and II, triggered by the binding of inhibitors (7YY, 7XB, and Y6G), to domain III, thereby modulating 3CLpro's activity. Normal mode analysis (NMA) and principal component analysis (PCA) revealed that inhibitor binding affects the structural flexibility and collective movements of the catalytic sites and domain III, influencing 3CLpro's function. The binding free energies, predicted by both MM-GBSA and QM/MM-GBSA methods, showed a high correlation with experimental data, validating the reliability of our analyses. Furthermore, residues L27, H41, C44, S46, M49, N142, G143, S144, C145, H163, H164, M165, and E166, identified through residue-based free energy decomposition, present promising targets for the design of anti-COVID-19 drugs and could facilitate the development of clinically effective 3CLpro inhibitors.
Collapse
Affiliation(s)
- Jianzhong Chen
- School of Science, Shandong Jiaotong University, Jinan 250357, China; (J.W.); (W.Y.); (L.Z.); (X.X.)
| | | | | | | | | |
Collapse
|
102
|
Arthurs AL, Dietrich B, Knöfler M, Lushington CJ, Thomas PQ, Adikusuma F, Williamson JM, Babikha S, Damhuis T, Jankovic-Karasoulos T, Smith MD, Pringle KG, Roberts CT. Genetically edited human placental organoids cast new light on the role of ACE2. Cell Death Dis 2025; 16:78. [PMID: 39920116 PMCID: PMC11806113 DOI: 10.1038/s41419-025-07400-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] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/05/2025] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
ACE2 expression is altered in pregnancy disorders and ACE2 gene variants are associated with several major pregnancy complications including small-for-gestational-age, fetal growth restriction and preeclampsia. This study utilised gene-editing to generate both ACE2 knockout and ACE2 rs2074192 placental organoids, facilitating mechanistic studies into the role of ACE2 in placental development, and the effect of fetal carriage of ACE2 rs2074192 CC, CT and TT genotypes. Parameters of cell and organoid growth were measured, together with qPCR, Western Blotting, and ELISA assessments, in all groups from both organoid models. Here, we report that ACE2 knockout results in delayed placental cell growth and increased cell death. ACE2 knockout organoids had lower ACE protein expression, reduced organoid diameters and asymmetrical growth. Placental organoids with the ACE2 rs2074192 TT genotype had significantly higher expression of ACE2 mRNA and ACE2 protein with elevated ACE2:ACE expression ratio and no change in ACE protein. Despite increased expression of ACE2 protein, ACE2 enzyme activity was significantly decreased in ACE2 rs2074192 TT placental organoids. TT organoids also had reduced diameters and asymmetrical growth. Our research provides a new molecular understanding of the role of ACE2 in placental development, with potential implications for pregnancy in the carriage of the ACE2 rs2074192 gene variant.
Collapse
Affiliation(s)
- Anya L Arthurs
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia.
| | - Bianca Dietrich
- Placental Development Group, Medical University of Vienna, Vienna, Austria
| | - Martin Knöfler
- Placental Development Group, Medical University of Vienna, Vienna, Austria
| | - Caleb J Lushington
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
- Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Paul Q Thomas
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
- Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- South Australian Genome Editing (SAGE), South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Fatwa Adikusuma
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
- Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Jessica M Williamson
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Susan Babikha
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Tyla Damhuis
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Tanja Jankovic-Karasoulos
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Melanie D Smith
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW, Australia
| | - Claire T Roberts
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
103
|
Parra RG, Komives EA, Wolynes PG, Ferreiro DU. Frustration In Physiology And Molecular Medicine. ARXIV 2025:arXiv:2502.03851v1. [PMID: 39975445 PMCID: PMC11838788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Molecules provide the ultimate language in terms of which physiology and pathology must be understood. Myriads of proteins participate in elaborate networks of interactions and perform chemical activities coordinating the life of cells. To perform these often amazing tasks, proteins must move and we must think of them as dynamic ensembles of three dimensional structures formed first by folding the polypeptide chains so as to minimize the conflicts between the interactions of their constituent amino acids. It is apparent however that, even when completely folded, not all conflicting interactions have been resolved so the structure remains 'locally frustrated'. Over the last decades it has become clearer that this local frustration is not just a random accident but plays an essential part of the inner workings of protein molecules. We will review here the physical origins of the frustration concept and review evidence that local frustration is important for protein physiology, protein-protein recognition, catalysis and allostery. Also, we highlight examples showing how alterations in the local frustration patterns can be linked to distinct pathologies. Finally we explore the extensions of the impact of frustration in higher order levels of organization of systems including gene regulatory networks and the neural networks of the brain.
Collapse
Affiliation(s)
- R. Gonzalo Parra
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Spain
| | | | - Peter G. Wolynes
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005
| | - Diego U. Ferreiro
- Protein Physiology Lab, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Consejo Nacional de Investigaciones Científicas y Técnicas - Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| |
Collapse
|
104
|
Trempelis KP, Kosmeri C, Kalavas P, Ladomenou F, Siomou E, Makis A. SARS-CoV-2 Variants and Their Impact on Pediatric COVID-19: Clinical Manifestations and Hematological Profiles. Diseases 2025; 13:48. [PMID: 39997055 PMCID: PMC11854181 DOI: 10.3390/diseases13020048] [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: 01/06/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND The aim of this study was to analyze data on pediatric cases of COVID-19 admitted to a tertiary referral hospital in northwest Greece. METHODS A retrospective analysis was conducted on the most common clinical manifestations and laboratory findings, stratified by age group and SARS-CoV-2 strain. RESULTS A total of 254 children were hospitalized, with a mean age of 4.5 years. Underlying conditions were present in 10.2% of cases; two children required pediatric intensive care unit (PICU) admission, and one child died. The most common hematological manifestations, in general, were neutropenia (30%) and lymphopenia (23%), whereas the findings varied when the children were stratified by age group. Eight children developed multisystem inflammatory syndrome (MIS-C), with the most common findings being anemia (75%), lymphopenia (50%), and thrombocytopenia (25%). Analysis of the SARS-CoV-2 strains revealed the proportions of the dominant strain over time. Fever was the predominant symptom across all strains, particularly in the Omicron group, which also had a high incidence of gastrointestinal symptoms. The longest hospital admission occurred in children with the Omicron strain, followed by the Wuhan, Alpha, and Delta strains. CONCLUSIONS Fever was the most consistent symptom across all age groups and virus strains. The most common hematological manifestations were neutropenia (30%) and lymphopenia (23%). The Omicron strain was associated with the longest hospital stay.
Collapse
Affiliation(s)
- Konstantinos Paris Trempelis
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece; (K.P.T.); (F.L.); (E.S.)
| | - Chrysoula Kosmeri
- Department of Pediatrics, University Hospital of Ioannina, 45500 Ioannina, Greece
| | - Panagiotis Kalavas
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece; (K.P.T.); (F.L.); (E.S.)
| | - Fani Ladomenou
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece; (K.P.T.); (F.L.); (E.S.)
- Department of Pediatrics, University Hospital of Ioannina, 45500 Ioannina, Greece
| | - Ekaterini Siomou
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece; (K.P.T.); (F.L.); (E.S.)
- Department of Pediatrics, University Hospital of Ioannina, 45500 Ioannina, Greece
| | - Alexandros Makis
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece; (K.P.T.); (F.L.); (E.S.)
- Department of Pediatrics, University Hospital of Ioannina, 45500 Ioannina, Greece
| |
Collapse
|
105
|
Barozi V, Tastan Bishop Ö. Impact of African-Specific ACE2 Polymorphisms on Omicron BA.4/5 RBD Binding and Allosteric Communication Within the ACE2-RBD Protein Complex. Int J Mol Sci 2025; 26:1367. [PMID: 39941135 PMCID: PMC11818624 DOI: 10.3390/ijms26031367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/27/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
Severe acute respiratory symptom coronavirus 2 (SARS-CoV-2) infection occurs via the attachment of the spike (S) protein's receptor binding domain (RBD) to human ACE2 (hACE2). Natural polymorphisms in hACE2, particularly at the interface, may alter RBD-hACE2 interactions, potentially affecting viral infectivity across populations. This study identified the effects of six naturally occurring hACE2 polymorphisms with high allele frequency in the African population (S19P, K26R, M82I, K341R, N546D and D597Q) on the interaction with the S protein RBD of the BA.4/5 Omicron sub-lineage through post-molecular dynamics (MD), inter-protein interaction and dynamic residue network (DRN) analyses. Inter-protein interaction analysis suggested that the K26R variation, with the highest interactions, aligns with reports of enhanced RBD binding and increased SARS-CoV-2 susceptibility. Conversely, S19P, showing the fewest interactions and largest inter-protein distances, agrees with studies indicating it hinders RBD binding. The hACE2 M82I substitution destabilized RBD-hACE2 interactions, reducing contact frequency from 92 (WT) to 27. The K341R hACE2 variant, located distally, had allosteric effects that increased RBD-hACE2 contacts compared to WThACE2. This polymorphism has been linked to enhanced affinity for Alpha, Beta and Delta lineages. DRN analyses revealed that hACE2 polymorphisms may alter the interaction networks, especially in key residues involved in enzyme activity and RBD binding. Notably, S19P may weaken hACE2-RBD interactions, while M82I showed reduced centrality of zinc and chloride-coordinating residues, hinting at impaired communication pathways. Overall, our findings show that hACE2 polymorphisms affect S BA.4/5 RBD stability and modulate spike RBD-hACE2 interactions, potentially influencing SARS-CoV-2 infectivity-key insights for vaccine and therapeutic development.
Collapse
Affiliation(s)
- Victor Barozi
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry, Microbiology and Bioinformatics, Rhodes University, Makhanda 6139, South Africa;
| | - Özlem Tastan Bishop
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry, Microbiology and Bioinformatics, Rhodes University, Makhanda 6139, South Africa;
- National Institute for Theoretical and Computational Sciences (NITheCS), Matieland 7602, South Africa
| |
Collapse
|
106
|
Jiang C, Liu Z, Wang Y, Yao G, He J, Li S, Rong R, Liang Z, Liu J. Severity and risk to inhalation of pathogen-laden aerosol in large public spaces: Insights from fangcang shelter hospitals under multi-location release. JOURNAL OF HAZARDOUS MATERIALS 2025; 483:136695. [PMID: 39616847 DOI: 10.1016/j.jhazmat.2024.136695] [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: 09/10/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 01/28/2025]
Abstract
The key to strengthening the inherent safety of large public spaces and implementing precise preventive measures lies in clarifying the transmission risks of respiratory infectious diseases based on multiple factors. This work innovatively improves a pathogen inhalation infection risk prediction model and attempts to apply it to a Fangcang Shelter Hospital to investigate the effect of pathogen release location on risk distribution and the role of airflow distribution in risk control mechanisms. The model used in the study improved in resolution and accuracy, shedding light on the airflow distribution mechanisms involved in pathogen transport and risk control, thus providing a quantitatively realistic landscape of the spread of respiratory infectious diseases in large indoor environments. Predictions reveal a significant unevenness in the spatial distribution of infection probabilities within the multi-patient shelter unit, which is further exacerbated by different release locations, and that extreme infection risks can reach 4 to 14 times the average. Additionally, the study noted that the infection probability in the medical staff area due to the long-distance transmission of contaminants can reach as high as 1.72 % and that patients from ward 6# could potentially infect a healthcare worker every four days.
Collapse
Affiliation(s)
- Chuan Jiang
- Department of Power Engineering, School of Energy Power and Mechanical Engineering, North China Electric Power University, Baoding, Hebei 071003, China
| | - Zhijian Liu
- Department of Power Engineering, School of Energy Power and Mechanical Engineering, North China Electric Power University, Baoding, Hebei 071003, China.
| | - Yongxin Wang
- Department of Power Engineering, School of Energy Power and Mechanical Engineering, North China Electric Power University, Baoding, Hebei 071003, China
| | - Guangpeng Yao
- Department of Power Engineering, School of Energy Power and Mechanical Engineering, North China Electric Power University, Baoding, Hebei 071003, China
| | - Junzhou He
- Department of Power Engineering, School of Energy Power and Mechanical Engineering, North China Electric Power University, Baoding, Hebei 071003, China
| | - Shiyue Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Rui Rong
- Department of Power Engineering, School of Energy Power and Mechanical Engineering, North China Electric Power University, Baoding, Hebei 071003, China
| | - Zhenyu Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Jingwei Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
107
|
Bibi N, Shah M, Khan S, Chohan MS, Kamal MA. Insilico targeting of virus entry facilitator NRP1 to block SARS-CoV2 entry. PLoS One 2025; 20:e0310855. [PMID: 39908250 DOI: 10.1371/journal.pone.0310855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/07/2024] [Indexed: 02/07/2025] Open
Abstract
The entry and infectivity of a virus are determined by its interaction with the host. SARS-CoV-2, the virus responsible for COVID-19, utilizes the spike (S) protein to attach to and enter host cells. Recent studies have identified neuropilin-1 (NRP1) as a crucial facilitator for the entry of SARS-CoV-2. The binding of the spike protein to the b1 domain of NRP1 has been shown to enhance viral infection twofold. Consequently, targeting NRP1 to disrupt this interaction represents a promising strategy to mitigate viral infection. In this study, a small molecule library of approximately 10,000 compounds was screened to identify those that could inhibit the interaction between NRP1 and the spike protein by targeting the b1 domain of NRP1. The crystallographic structure of the b1 domain of human NRP1 (PDB entry: 7JJC) was used for this purpose. Following virtual screening, docking studies, and evaluation of binding affinity and ADMET properties, 10 compounds were shortlisted. The top two candidates, AZD3839 and LY2090314, were selected for molecular dynamics simulation studies over 100 ns to assess binding stability. MM/GBSA calculations indicated that both AZD3839 and LY2090314 exhibited strong and stable binding to the b1 domain of NRP1. Computational modeling of the interaction between the b1 domain of NRP1 and the receptor-binding domain of the spike protein suggested that AZD3839 and LY2090314 could effectively hinder the NRP1-spike interaction. Therefore, these compounds may serve as potential drug candidates to reduce SARS-CoV-2 infectivity.
Collapse
Affiliation(s)
- Nousheen Bibi
- Departments of Bioinformatics, Shaheed Benazir Bhutto Women University Peshawar, Peshawar, Pakistan
| | - Maleeha Shah
- Departments of Bioinformatics, Shaheed Benazir Bhutto Women University Peshawar, Peshawar, Pakistan
| | - Shahzad Khan
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Al Hofuf, Al-Ahsa Saudi Arabia
| | - Muhammad Shahzad Chohan
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Al Hofuf, Al-Ahsa Saudi Arabia
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Faculty of Allied Health Sciences, Department of Pharmacy, Daffodil International University, Dhaka, Bangladesh
- Enzymoics, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| |
Collapse
|
108
|
Zhao Y, Tang Y, Wang QY, Li J. Ocular neuroinflammatory response secondary to SARS-CoV-2 infection-a review. Front Immunol 2025; 16:1515768. [PMID: 39967658 PMCID: PMC11832381 DOI: 10.3389/fimmu.2025.1515768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
With the consistent occurrence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the prevalence of various ocular complications has increased over time. SARS-CoV-2 infection has been shown to have neurotropism and therefore to lead to not only peripheral inflammatory responses but also neuroinflammation. Because the receptor for SARS-CoV-2, angiotensin-converting enzyme 2 (ACE2), can be found in many intraocular tissues, coronavirus disease 2019 (COVID-19) may also contribute to persistent intraocular neuroinflammation, microcirculation dysfunction and ocular symptoms. Increased awareness of neuroinflammation and future research on interventional strategies for SARS-CoV-2 infection are important for improving long-term outcomes, reducing disease burden, and improving quality of life. Therefore, the aim of this review is to focus on SARS-CoV-2 infection and intraocular neuroinflammation and to discuss current evidence and future perspectives, especially possible connections between conditions and potential treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Jia Li
- Department of Glaucoma, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
109
|
Elbadawy M, Saito N, Kato Y, Hayashi K, Abugomaa A, Kobayashi M, Yoshida T, Shibutani M, Kaneda M, Yamawaki H, Sasaki K, Usui T, Omatsu T. Establishment of a bat lung organoid culture model for studying bat-derived infectious diseases. Sci Rep 2025; 15:4035. [PMID: 39900611 PMCID: PMC11791068 DOI: 10.1038/s41598-025-88621-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/29/2025] [Indexed: 02/05/2025] Open
Abstract
Bat is considered a natural reservoir of various important pathogens, including severe acute respiratory syndrome coronavirus (SARS-CoV), SARS-CoV-2, Ebola virus, and Nipah virus. To study these viruses' pathogenicity and proliferation efficacy and viral tolerance mechanisms in bats, bat-derived cell lines, and primary cultured cells are used. However, these do not adequately reflect the exact biology of bats, and establishing new bat-related research models is necessary. Organoid culture can recapitulate organ structure, functions, and diseases. The respiratory tract is one of the primary routes of viral infection, and the establishment of bat lung organoids (BLO) is necessary to study the viral susceptibility in bats. Therefore, we aimed to establish a culture method of BLO from Rousettus leschenaultia that died of natural causes. The generated BLO successfully recapitulated the characteristics of pulmonary epithelial structure and morphology. BLO expressed the entry receptors for coronavirus, Angiotensin-converting enzyme 2 (ACE2), and Transmembrane Protease Serine 2 (TMPRSS2), and alveolar type 2 cells were successfully sorted from BLO, which has an important role for the development of viral infection in the respiratory system. Furthermore, we showed that BLO had no susceptibility to Pteropine orthoreovirus (PRV) compared to bat intestinal organoids. Collectively, our established bat organoid culture models including this BLO might become promising in vitro biomaterials to study the biology of bat-derived infectious diseases.
Collapse
Affiliation(s)
- Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, 183-8509, Tokyo, Japan
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, 13736, Elqaliobiya, Egypt
| | - Nagisa Saito
- Research and Education Center for Prevention of Global Infectious Diseases of Animals, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, 183-8509, Tokyo, Japan
| | - Yuki Kato
- Research and Education Center for Prevention of Global Infectious Diseases of Animals, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, 183-8509, Tokyo, Japan
| | - Kimika Hayashi
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, 183-8509, Tokyo, Japan
| | - Amira Abugomaa
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, 183-8509, Tokyo, Japan
- Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Mio Kobayashi
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, 183-8509, Tokyo, Japan
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, 183-8509, Tokyo, Japan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, 183-8509, Tokyo, Japan
| | - Masahiro Kaneda
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, 183-8509, Tokyo, Japan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, 35-1, Higashi 23 ban-cho, Towada, 034-8628, Aomori, Japan
| | - Kazuaki Sasaki
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, 183-8509, Tokyo, Japan
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, 183-8509, Tokyo, Japan.
| | - Tsutomu Omatsu
- Research and Education Center for Prevention of Global Infectious Diseases of Animals, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu, 183-8509, Tokyo, Japan.
| |
Collapse
|
110
|
Levinger R, Tussia-Cohen D, Friedman S, Lender Y, Nissan Y, Fraimovitch E, Gavriel Y, Tearle JLE, Kolodziejczyk AA, Moon KM, Gomes T, Kunowska N, Weinberg M, Donati G, Foster LJ, James KR, Yovel Y, Hagai T. Single-cell and Spatial Transcriptomics Illuminate Bat Immunity and Barrier Tissue Evolution. Mol Biol Evol 2025; 42:msaf017. [PMID: 39836373 PMCID: PMC11817796 DOI: 10.1093/molbev/msaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/26/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025] Open
Abstract
Bats have adapted to pathogens through diverse mechanisms, including increased resistance-rapid pathogen elimination, and tolerance-limiting tissue damage following infection. In the Egyptian fruit bat (an important model in comparative immunology), several mechanisms conferring disease tolerance were discovered, but mechanisms underpinning resistance remain poorly understood. Previous studies on other species suggested that the elevated basal expression of innate immune genes may lead to increased resistance to infection. Here, we test whether such transcriptional patterns occur in Egyptian fruit bat tissues through single-cell and spatial transcriptomics of gut, lung, and blood cells, comparing gene expression between bat, mouse, and human. Despite numerous recent loss and expansion events of interferons in the bat genome, interferon expression and induction are remarkably similar to that of mouse. In contrast, central complement system genes are highly and uniquely expressed in key regions in bat lung and gut epithelium, unlike in human and mouse. Interestingly, the unique expression of these genes in the bat gut is strongest in the crypt, where developmental expression programs are highly conserved. The complement system genes also evolve rapidly in their coding sequences across the bat lineage. Finally, the bat complement system displays strong hemolytic activity. Together, these results indicate a distinctive transcriptional divergence of the complement system, which may be linked to bat resistance, and highlight the intricate evolutionary landscape of bat immunity.
Collapse
Affiliation(s)
- Roy Levinger
- Shmunis School of Biomedicine and Cancer Research, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dafna Tussia-Cohen
- Shmunis School of Biomedicine and Cancer Research, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Sivan Friedman
- Shmunis School of Biomedicine and Cancer Research, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yan Lender
- Shmunis School of Biomedicine and Cancer Research, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yomiran Nissan
- School of Zoology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Evgeny Fraimovitch
- Shmunis School of Biomedicine and Cancer Research, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yuval Gavriel
- Shmunis School of Biomedicine and Cancer Research, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Jacqueline L E Tearle
- Translational Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | | | - Kyung-Mee Moon
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, BC, Canada
| | - Tomás Gomes
- Fundação GIMM - Gulbenkian Institute for Molecular Medicine, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Natalia Kunowska
- Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Maya Weinberg
- School of Zoology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Giacomo Donati
- Department of Life Sciences and Systems Biology, University of Turin, Torino, Italy
- Molecular Biotechnology Center, University of Turin, Torino, Italy
| | - Leonard J Foster
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, BC, Canada
| | - Kylie R James
- Translational Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Yossi Yovel
- School of Zoology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tzachi Hagai
- Shmunis School of Biomedicine and Cancer Research, George S Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
111
|
Jones LR. Intra-host variability of SARS-CoV-2: Patterns, causes and impact on COVID-19. Virology 2025; 603:110366. [PMID: 39724740 DOI: 10.1016/j.virol.2024.110366] [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: 10/30/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Intra-host viral variability is related to pathogenicity, persistence, drug resistance, and the emergence of new clades. This work reviews the large amount of data on SARS-CoV-2 intra-host variability accumulated to date, addressing known and potential implications in COVID-19 and the emergence of VOCs and lineage-defining mutations. Topics covered include the distribution of intra-host polymorphisms across the genome, the corresponding mutational signatures, their patterns of emergence and extinction throughout infection, and the processes governing their abundance, frequency, and type (synonymous, nonsynonymous, indels, nonsense). Besides, evidence is reviewed that the virus can replicate and mutate in isolation at different anatomical compartments, which may imply that what we have learned from respiratory samples could be part of a broader picture.
Collapse
Affiliation(s)
- Leandro R Jones
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Avenida Rivadavia 1917, C1083ACA Ciudad Autónoma de Buenos Aires, Argentina; Laboratorio de Virología y Genética Molecular (LVGM), Facultad de Ciencias Naturales y Ciencias de la Salud, Universidad Nacional de la Patagonia San Juan Bosco, Belgrano 160, Trelew, CP, 9100, Argentina.
| |
Collapse
|
112
|
Trivedi R, Ashiq PL, Garg N, Jha P, Gadly T, Chakraborty B. Comparative study of alpha, beta, and omicron spike protein by computing the IR/Raman frequencies and UV-vis adsorption - A computational analysis through DFT. Microb Pathog 2025; 199:107195. [PMID: 39645095 DOI: 10.1016/j.micpath.2024.107195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 11/20/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
The outbreak of COVID-19 coronavirus disease around the end of 2019 was a pandemic. The virus has been mutated and so many strains like Alpha, Beta, and Omicron are present in different parts of the world. Hence, timely detection technique is important to overcome the diagnostic challenges. Considering the need for this pandemic situation, we used a spectroscopy test methodology to distinguish different strains of Covid-19 by computing the infrared & Raman frequencies and the optical absorption data. Optimization of spike protein of Alpha, and Omicron mutations showed the high value of dipole moment (4.44, and 4.36) Debye, and polarizability [Alpha (233.62), Omicron (228.65)] indicating more bioactivity of Alpha and Omicron instead of Beta. Molecular electrostatic potential map exhibits the presence of electrophilic and nucleophilic region suggesting charge transfer of spikes to accept/donate electrons and hence the system increased reactiveness. UV-Vis absorption analysis also shows electronic transitions (σ to π∗ and π to π∗) due to that protein probe mechanism of Alpha and Omicron becomes increasingly become unsaturated thus confirming its easy binding ability to the target human protein as compared to binding affinity of Beta spike protein.
Collapse
Affiliation(s)
- Ravi Trivedi
- High Pressure and Synchrotron Radiation Physics Division, Bhabha Atomic Research Centre, Trombay, Mumbai, India; Department of Physics, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India; Centre for Computational Physics, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India.
| | - P L Ashiq
- School of Pure and Applied Physics, Mahatma Gandhi University, Priyadarsini Hills P O, Kottayam, 686560, India
| | - Nandini Garg
- High Pressure and Synchrotron Radiation Physics Division, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | - Prafulla Jha
- Department of Physics, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - Trilochan Gadly
- Bio-Organic Divisions, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | - Brahmananda Chakraborty
- High Pressure and Synchrotron Radiation Physics Division, Bhabha Atomic Research Centre, Trombay, Mumbai, India; Homi Bhabha National Institute, India.
| |
Collapse
|
113
|
Hong SJ, Resnick SJ, Iketani S, Cha JW, Albert BA, Fazekas CT, Chang CW, Liu H, Dagan S, Abagyan MR, Fajtová P, Culbertson B, Brace B, Reddem ER, Forouhar F, Glickman JF, Balkovec JM, Stockwell BR, Shapiro L, O'Donoghue AJ, Sabo Y, Freundlich JS, Ho DD, Chavez A. A multiplex method for rapidly identifying viral protease inhibitors. Mol Syst Biol 2025; 21:158-172. [PMID: 39762652 PMCID: PMC11790949 DOI: 10.1038/s44320-024-00082-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 02/05/2025] Open
Abstract
With current treatments addressing only a fraction of pathogens and new viral threats constantly evolving, there is a critical need to expand our existing therapeutic arsenal. To speed the rate of discovery and better prepare against future threats, we establish a high-throughput platform capable of screening compounds against 40 diverse viral proteases simultaneously. This multiplex approach is enabled by using cellular biosensors of viral protease activity combined with DNA-barcoding technology, as well as several design innovations that increase assay sensitivity and correct for plate-to-plate variation. Among >100,000 compound-target interactions explored within our initial screen, a series of broad-acting inhibitors against coronavirus proteases were uncovered and validated through orthogonal assays. A medicinal chemistry campaign was performed to improve one of the inhibitor's potency while maintaining its broad activity. This work highlights the power of multiplex screening to efficiently explore chemical space at a fraction of the time and costs of previous approaches.
Collapse
Affiliation(s)
- Seo Jung Hong
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Samuel J Resnick
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
- Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sho Iketani
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Ji Won Cha
- Department of Pediatrics, University of California San Diego, La Jolla, CA, 92123, USA
| | - Benjamin Alexander Albert
- Department of Pediatrics, University of California San Diego, La Jolla, CA, 92123, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Christopher T Fazekas
- Department of Pediatrics, University of California San Diego, La Jolla, CA, 92123, USA
| | - Ching-Wen Chang
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, 07110, USA
| | - Hengrui Liu
- Department of Biological Sciences, Department of Chemistry, and Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
| | - Shlomi Dagan
- Fisher Drug Discovery Resource Center, The Rockefeller University, New York, NY, 10065, USA
| | - Michael R Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Pavla Fajtová
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Bruce Culbertson
- Medical Scientist Training Program, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Brooklyn Brace
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Eswar R Reddem
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, 10032, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, 10027, USA
| | - Farhad Forouhar
- Department of Pathology and Cell Biology and Columbia University Digestive and Liver Disease Research Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, 10032, USA
| | - J Fraser Glickman
- Fisher Drug Discovery Resource Center, The Rockefeller University, New York, NY, 10065, USA
| | - James M Balkovec
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, 07110, USA
| | - Brent R Stockwell
- Department of Biological Sciences, Department of Chemistry, and Department of Pathology and Cell Biology, Columbia University, New York, NY, 10032, USA
- Department of Pathology and Cell Biology and Columbia University Digestive and Liver Disease Research Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, 10032, USA
| | - Lawrence Shapiro
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, 10032, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, 10027, USA
| | - Anthony J O'Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yosef Sabo
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Joel S Freundlich
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University - New Jersey Medical School, Newark, NJ, 07103, USA
- Department of Medicine, Center for Emerging and Re-emerging Pathogens, Rutgers University - New Jersey Medical School, Newark, NJ, 07103, USA
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Alejandro Chavez
- Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA.
- Department of Pediatrics, University of California San Diego, La Jolla, CA, 92123, USA.
| |
Collapse
|
114
|
Batlle D, Hassler L, Wysocki J. ACE2, From the Kidney to SARS-CoV-2: Donald Seldin Award Lecture 2023. Hypertension 2025; 82:166-180. [PMID: 39624896 DOI: 10.1161/hypertensionaha.124.22064] [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: 01/18/2025]
Abstract
ACE2 (angiotensin-converting enzyme 2) is a monocarboxypeptidase that cleaves Ang II (angiotensin II) among other substrates. ACE2 is present in the cell membrane of many organs, most abundantly in epithelial cells of kidney proximal tubules and the small intestine, and also exists in soluble forms in plasma and body fluids. Membrane-bound ACE2 exerts a renoprotective action by metabolizing Ang II and therefore attenuating the undesirable actions of excess Ang II. Therefore, soluble ACE2, by downregulating this peptide, may exert a therapeutic action. Our laboratory has designed ACE2 truncates that pass the glomerular filtration barrier to target the kidney renin-angiotensin system directly and, therefore, compensate for loss of kidney membrane-bound ACE2. Membrane-bound ACE2 is also the essential receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Soluble ACE2 proteins have been studied as a way to intercept SARS-CoV-2 from binding to membrane-bound ACE2 and prevent cell entry of SARS-CoV-2 altogether. We bioengineered a soluble ACE2 protein, termed ACE2 618-DDC-ABD, with increased binding affinity for SARS-CoV-2 and prolonged duration of action, which, when administered intranasally, provides near-complete protection from lethality in k18hACE2 mice infected with different SARS-CoV-2 variants. The main advantage of soluble ACE2 proteins for the neutralization of SARS-CoV-2 is their immediate onset of action and universality for current and future emerging SARS-CoV-2 variants. It is notable that ACE2 is critically involved in 2 dissimilar functions: as a receptor for cell entry of many coronaviruses and as an enzyme in the metabolism of Ang II, and yet in both cases, it is a therapeutic target.
Collapse
Affiliation(s)
- Daniel Batlle
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Luise Hassler
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Jan Wysocki
- Division of Nephrology/Hypertension, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
115
|
Gallo E. Revolutionizing Synthetic Antibody Design: Harnessing Artificial Intelligence and Deep Sequencing Big Data for Unprecedented Advances. Mol Biotechnol 2025; 67:410-424. [PMID: 38308755 DOI: 10.1007/s12033-024-01064-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/02/2024] [Indexed: 02/05/2024]
Abstract
Synthetic antibodies (Abs) represent a category of engineered proteins meticulously crafted to replicate the functions of their natural counterparts. Such Abs are generated in vitro, enabling advanced molecular alterations associated with antigen recognition, paratope site engineering, and biochemical refinements. In a parallel realm, deep sequencing has brought about a paradigm shift in molecular biology. It facilitates the prompt and cost-effective high-throughput sequencing of DNA and RNA molecules, enabling the comprehensive big data analysis of Ab transcriptomes, including specific regions of interest. Significantly, the integration of artificial intelligence (AI), based on machine- and deep- learning approaches, has fundamentally transformed our capacity to discern patterns hidden within deep sequencing big data, including distinctive Ab features and protein folding free energy landscapes. Ultimately, current AI advances can generate approximations of the most stable Ab structural configurations, enabling the prediction of de novo synthetic Abs. As a result, this manuscript comprehensively examines the latest and relevant literature concerning the intersection of deep sequencing big data and AI methodologies for the design and development of synthetic Abs. Together, these advancements have accelerated the exploration of antibody repertoires, contributing to the refinement of synthetic Ab engineering and optimizations, and facilitating advancements in the lead identification process.
Collapse
Affiliation(s)
- Eugenio Gallo
- Avance Biologicals, Department of Medicinal Chemistry, 950 Dupont Street, Toronto, ON, M6H 1Z2, Canada.
- RevivAb, Department of Protein Engineering, Av. Ipiranga, 6681, Partenon, Porto Alegre, RS, 90619-900, Brazil.
| |
Collapse
|
116
|
Webb I, Erdmann M, Milligan R, Savage M, Matthews DA, Davidson AD. Examining the feasibility of replacing ORF3a with fluorescent genes to construct SARS-CoV-2 reporter viruses. J Gen Virol 2025; 106:002072. [PMID: 39937571 PMCID: PMC11822206 DOI: 10.1099/jgv.0.002072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
The SARS-CoV-2 genome encodes at least nine accessory proteins, including innate immune antagonist and putative viroporin ORF3a. ORF3a plays a role in many stages of the viral replication cycle, including immune modulation. We constructed two recombinant (r)SARS-CoV-2 viruses in which the ORF3a gene was replaced with mScarlet (mS) or mNeonGreen (mNG), denoted as rSARS-CoV-2-Δ3a-mS and rSARS-CoV-2-Δ3a-mNG, respectively. rSARS-CoV-2-Δ3a-mNG generated a fluorescent signal after infection in both A549-ACE-2-TMPRSS2 (AAT) and Vero-E6-TMPRSS2 (VTN) cells, unlike rSARS-CoV-2-Δ3a-mS. rSARS-CoV-2-Δ3a-mS mS protein could be detected immunologically in VTN but not AAT cells, indicating the expression of a non-fluorescent mS protein. The analysis of the viral transcriptomes in infected AAT cells by nanopore direct RNA sequencing (dRNAseq) revealed that the level of mS transcript was below the limit of detection in AAT cells. rSARS-CoV-2-Δ3a-mNG virus was found to be genetically stable in AAT and VTN cells, but rSARS-CoV-2-Δ3a-mS acquired partial deletions of the mS gene during sequential passaging in VTN cells, creating the virus rSARS-CoV-2-Δ3a-ΔmS. The mS deletion in VTN cells removes the chromophore coding sequence, and this may explain the presence of a non-fluorescent mS protein detected in VTN cells. The rSARS-CoV-2-Δ3a-mNG, rSARS-CoV-2-Δ3a-mS and rSARS-CoV-2-Δ3a-ΔmS viruses all replicated to a lower titre and produced smaller plaques than the parental rSARS-CoV-2-S-D614G. Interestingly, the rSARS-CoV-2-Δ3a-ΔmS virus produced higher virus titres and larger plaque sizes than rSARS-CoV-2-Δ3a-mS. This suggested that both the insertion of mS coding sequence and the deletion of ORF3a coding sequence contributed to attenuation. In comparison with rSARS-CoV-2, the rSARS-CoV-2-Δ3a-mS and rSARS-CoV-2-Δ3a-mNG viruses showed increased sensitivity to pre-treatment of cells with IFN-α but did not exhibit a dose-dependent increase in replication in the presence of the Janus kinase-signal transducer and activator of transcription signalling pathway inhibitor, ruxolitinib. In conclusion, the replacement of the ORF3a coding sequence with those of fluorescent reporter proteins attenuated the replication of SARS-CoV-2 and its ability to effectively evade the innate immune response in vitro.
Collapse
Affiliation(s)
- Isobel Webb
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - Maximillian Erdmann
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - Rachel Milligan
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - Megan Savage
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - David A. Matthews
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol BS8 1TD, UK
| | - Andrew D. Davidson
- School of Cellular and Molecular Medicine, Faculty of Health and Life Sciences, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
117
|
Verdugo C, Verdugo C, Fica A, Hernández F, Ramírez-Reveco A, Plaza A, Castro N, Hernández-Riquelme M, Acosta-Jamett G. An assessment of the Chilean COVID-19 surveillance program through the comparison between reported and true SARS-CoV-2 infection prevalence: A case study of three urban centers in southern Chile. Public Health 2025; 239:207-214. [PMID: 39884022 DOI: 10.1016/j.puhe.2024.12.033] [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: 07/04/2024] [Revised: 11/26/2024] [Accepted: 12/17/2024] [Indexed: 02/01/2025]
Abstract
OBJECTIVES Estimate the detection limits of the COVID-19 surveillance system (SS) in Chile, by estimating the SARS-CoV-2 true prevalence (TP) and the reported official positivity prevalence (OPP) gap. STUDY DESIGN Randomized cross-sectional. METHODS Two sampling campaigns (SC) were conducted (October-November 2020 and December 2020-January 2021) in the cities of Temuco, Valdivia, and Osorno. Blood was collected from adults from randomly selected households. Sera were analyzed using a commercial later flow test (LFT). A meta-analysis was performed to estimate LFT-performance in asymptomatic-cases. Data were analyzed using a Bayesian latent class model (BLCM) to estimate TP. Finally, BLCM outputs were compared with the OPP, by calculating the TP/OPP rate. RESULTS 1124 and 1017 households were visited during the 1st and 2nd SC, respectively. The BLCM rendered TP estimates of 6.5 %, 3.2 %, and 6.6 % for the cities of Temuco, Valdivia, and Osorno, respectively (1stSC), increasing to 9.4 %, 5.0 %, and 7.5 %, 60 days later (2ndSC). Depending on the city and SC, TP/OPP rates varied between 2.3 and 5.7. CONCLUSION The national SS was unable to detect 70-79 % of all infected cases, suggesting that mild and asymptomatic cases were scarcely detected.
Collapse
Affiliation(s)
- Cristóbal Verdugo
- Center for Surveillance and Evolution of Infectious Diseases, Universidad Austral de Chile, Valdivia, Chile; Instituto de Medicina Preventiva Veterinaria, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile.
| | - Claudio Verdugo
- Center for Surveillance and Evolution of Infectious Diseases, Universidad Austral de Chile, Valdivia, Chile; Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Alberto Fica
- Universidad Austral de Chile, Escuela de Medicina, Facultad de Medicina, Valdivia, Chile
| | - Felipe Hernández
- Center for Surveillance and Evolution of Infectious Diseases, Universidad Austral de Chile, Valdivia, Chile; Instituto de Medicina Preventiva Veterinaria, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Alfredo Ramírez-Reveco
- Universidad Austral de Chile, Instituto de Ciencia Animal, Facultad de Ciencias Veterinarias, Valdivia, Chile
| | - Anita Plaza
- Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Natalia Castro
- Center for Surveillance and Evolution of Infectious Diseases, Universidad Austral de Chile, Valdivia, Chile; Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Maximiliano Hernández-Riquelme
- Instituto de Medicina Preventiva Veterinaria, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Gerardo Acosta-Jamett
- Center for Surveillance and Evolution of Infectious Diseases, Universidad Austral de Chile, Valdivia, Chile; Instituto de Medicina Preventiva Veterinaria, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
118
|
Nurmi V, Mayne R, Knight C, Almonacid-Mendoza HL, Secret S, Estcourt L, Hepojoki J, Šuštić T, Lamikanra AA, Tsang HP, Menon DK, Shankar-Hari M, van der Schoot CE, Vidarsson G, Roberts DJ, Simmonds P, Hedman K, Harvala H. Individual patient and donor seroprofiles in convalescent plasma treatment of COVID-19 in REMAP-CAP clinical trial. J Infect 2025; 90:106412. [PMID: 39798805 DOI: 10.1016/j.jinf.2025.106412] [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: 11/03/2024] [Revised: 01/01/2025] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
OBJECTIVES Convalescent plasma (CP) treatment of COVID-19 has shown significant therapeutic effect only when administered early. We investigated the importance of patient and CP seroprofiles on treatment outcome in REMAP-CAP CP trial. METHODS We evaluated neutralising antibodies (nAb), anti-spike (S) IgM, IgG, IgG avidity, IgG fucosylation and respiratory viral loads in a sub-set of patients (n=80) and controls (n=51) before and after transfusion, comparing them to those in the CP units (n=157) they received. RESULTS Most patients were SARS-CoV-2 seropositive pre-transfusion (72% nAb; 89% S-IgG seropositivity). The majority (80%) had higher pre-transfusion S-IgG levels (median 1.7×106 arbitrary units (AU); 56%) or S-IgG production rates (median 1.1×106 AU/day; 64%) than they received from CP (median 2.2×105 AU). Only 22% of the patients demonstrated significant (median 24-fold) increase in their S-IgG levels acquired from transfusion. Better outcomes, measured by organ support-free days, were associated with increase in S-IgM levels (p=0.007), decreased S-IgG fucosylation (p<0.001), lower patient age (p<0.001) but not with receiving CP (p=0.337). CONCLUSIONS Based on our data, increased S-antibody levels linked to transfused CP were only observed in pre-seroconversion or immunodeficient patients lacking their own SARS-CoV-2 antibodies, representing the groups where CP treatment has previously shown most benefit.
Collapse
Affiliation(s)
- Visa Nurmi
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK; Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Richard Mayne
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Chanice Knight
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | | | - Shannah Secret
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK; Clinical Services, NHS Blood and Transplant, Oxford, UK
| | - Lise Estcourt
- Clinical Services, NHS Blood and Transplant, Oxford, UK; Radcliffe Department of Medicine and BRC Haematology Theme, University of Oxford, Oxford, UK
| | - Jussi Hepojoki
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Institute of Veterinary Pathology, Vetsuisse faculty, University of Zürich, Zürich, Switzerland
| | - Tonći Šuštić
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands; Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Centre for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - Abigail A Lamikanra
- Clinical Services, NHS Blood and Transplant, Oxford, UK; Radcliffe Department of Medicine and BRC Haematology Theme, University of Oxford, Oxford, UK
| | - Hoi Pat Tsang
- Clinical Services, NHS Blood and Transplant, Oxford, UK
| | - David K Menon
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Manu Shankar-Hari
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands; Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, the Netherlands; Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Centre for Biomolecular Research, Utrecht University, Utrecht, the Netherlands
| | - David J Roberts
- Clinical Services, NHS Blood and Transplant, Oxford, UK; Radcliffe Department of Medicine and BRC Haematology Theme, University of Oxford, Oxford, UK
| | - Peter Simmonds
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Klaus Hedman
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Helsinki University Hospital Diagnostics Centre, Helsinki, Finland
| | - Heli Harvala
- Radcliffe Department of Medicine and BRC Haematology Theme, University of Oxford, Oxford, UK; Microbiology Services, NHS Blood and Transplant, Colindale, UK; Infection and Immunity, University College of London, London, UK
| |
Collapse
|
119
|
Saravanan K, Elavarasi S, Revathi G, Karuppannan P, Ashokkumar M, Muthusamy C, Ram Kumar A. Targeting SARS-CoV2 spike glycoprotein: molecular insights into phytocompounds binding interactions - in-silico molecular docking. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025; 36:315-332. [PMID: 39225011 DOI: 10.1080/09205063.2024.2399395] [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: 05/23/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
This study utilized small molecular characterization and docking study to evaluate the binding affinity of seven antiviral phytocompounds with the SARS CoV-2 variants (SARS-CoV-2 Spike Glycoprotein, SARS-CoV-2 Spike Protein Variant in 1-RBD, Alpha Variant SARS-CoV2- Spike Protein). The results revealed that five of seven compounds, possesses excellent drug lead property reveled through in-silico ADMET analysis. In addition, six of seven except D-Glucosamine, exhibited excellent binding affinity. Six ligands possess significant binding affinity towards SARS-CoV-2 variants 6VXX, 7LWV and 7R13, which is certainly greater than Remdesivir. Fagaronine found to be the best drug candidate against SARS-CoV-2 variants, It was found that -7.4, -5.6 and -6.3 is the docking score respectively. Aranotin, Beta aescin, Gliotoxin, and Fagaronine formed hydrogen bonds with specific amino acids and exhibited significant binding interactions. These findings suggest that these phytocompounds could be promising candidates for developing antiviral therapies against SARS-CoV-2. Moreover, the study underscores the importance of molecular docking in understanding protein-ligand interactions and its role in drug discovery. The documented pharmacological properties of these compounds in the literature further support their potential therapeutic relevance in various diseases.
Collapse
Affiliation(s)
- K Saravanan
- PG and Research Dept. of Zoology, Nehru Memorial College (Autonomous), Puthanampatti, Thiruchirappalli, Tamilnadu, India
| | - S Elavarasi
- PG and Research Dept. of Zoology, Holy Cross College (Autonomous), Thiruchirappalli, Tamilnadu, India
| | - G Revathi
- PG and Research Dept. of Zoology, Nehru Memorial College (Autonomous), Puthanampatti, Thiruchirappalli, Tamilnadu, India
| | - P Karuppannan
- PG and Research Dept. of Zoology, Vivekananda College of Arts and Science for women (Autonomous), Tiruchengode, Tamilnadu, India
| | - M Ashokkumar
- Department of Physics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Thandalam, Chennai, India
| | - C Muthusamy
- Department of Food Technology, School of Liberal Arts and Applied Sciences, Hindustan Institute of Technology and Science, Padur, OMR, Chennai, Tamilnadu, India
| | - A Ram Kumar
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Thandalam, Chennai, Tamil Nadu, India
| |
Collapse
|
120
|
Saha A, Ganguly A, Kumar A, Srivastava N, Pathak R. Harnessing Epigenetics: Innovative Approaches in Diagnosing and Combating Viral Acute Respiratory Infections. Pathogens 2025; 14:129. [PMID: 40005506 PMCID: PMC11858160 DOI: 10.3390/pathogens14020129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Acute respiratory infections (ARIs) caused by viruses such as SARS-CoV-2, influenza viruses, and respiratory syncytial virus (RSV), pose significant global health challenges, particularly for the elderly and immunocompromised individuals. Substantial evidence indicates that acute viral infections can manipulate the host's epigenome through mechanisms like DNA methylation and histone modifications as part of the immune response. These epigenetic alterations can persist beyond the acute phase, influencing long-term immunity and susceptibility to subsequent infections. Post-infection modulation of the host epigenome may help distinguish infected from uninfected individuals and predict disease severity. Understanding these interactions is crucial for developing effective treatments and preventive strategies for viral ARIs. This review highlights the critical role of epigenetic modifications following viral ARIs in regulating the host's innate immune defense mechanisms. We discuss the implications of these modifications for diagnosing, preventing, and treating viral infections, contributing to the advancement of precision medicine. Recent studies have identified specific epigenetic changes, such as hypermethylation of interferon-stimulated genes in severe COVID-19 cases, which could serve as biomarkers for early detection and disease progression. Additionally, epigenetic therapies, including inhibitors of DNA methyltransferases and histone deacetylases, show promise in modulating the immune response and improving patient outcomes. Overall, this review provides valuable insights into the epigenetic landscape of viral ARIs, extending beyond traditional genetic perspectives. These insights are essential for advancing diagnostic techniques and developing innovative treatments to address the growing threat of emerging viruses causing ARIs globally.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (A.S.); (N.S.)
| | - Anirban Ganguly
- Department of Biochemistry, All India Institute of Medical Sciences, Deoghar 814152, India;
| | - Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, India;
| | - Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (A.S.); (N.S.)
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
121
|
Almatroudi A. Analysis of bioactive compounds of Olea europaea as potential inhibitors of SARS-CoV-2 main protease: a pharmacokinetics, molecular docking and molecular dynamics simulation studies. J Biomol Struct Dyn 2025; 43:1147-1158. [PMID: 38063160 DOI: 10.1080/07391102.2023.2291172] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/10/2023] [Indexed: 01/16/2025]
Abstract
COVID-19 is a highly infectious disease caused by a new type of extremely contagious coronavirus called SARS-CoV-2. The virus's main protease enzyme, SARS-CoV-2 Mpro, is essential for its replication and transcription processes. Targeting this enzyme presents a promising avenue for antiviral drug development. Researchers have explored the intricate three-dimensional configurations of the enzyme, analyzing its interactions with various inhibitors. These findings provide a foundation for designing specific and powerful inhibitors targeting SARS-CoV-2 Mpro. Certain plants possess medicinal attributes due to the presence of bioactive compounds that inhibit pathogens. The olive tree (Olea europaea) has served as a source of food and medicine, containing bioactive compounds in its leaves that hinder the proliferation of various pathogens including viruses. This study explores the potential of bioactive compounds from olive leaf extract (OLE) to inhibit SARS-CoV-2 Mpro. In-silico study was conducted to predict the pharmacokinetic and toxicity profiles of these compounds. Molecular docking was utilized to assess their binding affinity to SARS-CoV-2 Mpro and their potential interference with its function. The top three compounds, apigenin (Api), luteolin-7-O-glucoside (Lut) and rutin (Rut), were chosen based on their favorable drug-like properties and strong binding affinities to Mpro. Detailed molecular dynamics simulations demonstrated the stability of SARS-CoV-2 Mpro in conjunction with these compounds, showing minimal structural alterations over the simulation period. Particularly, Lut and Rut formed bonds with critical amino acid residues His41 and Cys145 of Mpro, suggesting their potential inhibitory effect. These findings suggest that these compounds hold promise as natural drug candidates for combating COVID-19.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
122
|
Yang L, Zeng XT, Luo RH, Tang Y, Ren SX, Long XY, Fu XH, Zhang WJ, Ren HY, Zheng YT, Cheng W. CRTC3 restricts SARS-CoV-2 replication and is antagonized by CREB. Virol Sin 2025; 40:92-108. [PMID: 39736320 PMCID: PMC11963146 DOI: 10.1016/j.virs.2024.12.006] [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: 08/05/2024] [Accepted: 12/26/2024] [Indexed: 01/01/2025] Open
Abstract
Virus-encoding RNA-dependent RNA polymerase (RdRp) is essential for genome replication and gene transcription of human coronaviruses (HCoVs), including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We previously identified the interaction between the catalytic subunit NSP12 of SARS-CoV-2 RdRp and the host protein CREB-regulated transcription coactivator 3 (CRTC3), a member of the CRTC family that regulates cyclic AMP response element-binding protein (CREB)-mediated transcriptional activation. Currently, the implication of CRTC3 in the pathogenesis of HCoVs is poorly understood. Herein, we demonstrated that CRTC3 attenuates RdRp activity and SARS-CoV-2 genome replication, therefore reducing the production of progeny viruses. The interaction of CRTC3 with NSP12 contributes to its inhibitory effect on RdRp activity. Furthermore, we expanded the suppressive effects of two other CRTC family members (CRTC1 and CRTC2) on the RdRp activities of lethal HCoVs, including SARS-CoV-2 and Middle East respiratory syndrome coronavirus (MERS-CoV), along with the CREB antagonization. Overall, our research suggests that CRTCs restrict the replication of HCoVs and are antagonized by CREB, which not only provides new insights into the replication regulation of HCoVs, but also offers important information for the development of anti-HCoV interventions.
Collapse
Affiliation(s)
- Li Yang
- Department of Pulmonary and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-related Molecular Network, and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xiao-Tao Zeng
- Department of Pulmonary and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-related Molecular Network, and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China; Research and Innovation Center, Pengzhou People's Hospital, Pengzhou 610000, China
| | - Rong-Hua Luo
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Ying Tang
- Department of Pulmonary and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-related Molecular Network, and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Si-Xue Ren
- Department of Pulmonary and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-related Molecular Network, and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xin-Yan Long
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China
| | - Xiang-Hui Fu
- Department of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wan-Jiang Zhang
- Department of Pathophysiology, Shihezi University School of Medicine, The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi 832003, China
| | - Hai-Yan Ren
- Department of Pulmonary and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-related Molecular Network, and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China.
| | - Yong-Tang Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, China.
| | - Wei Cheng
- Department of Pulmonary and Critical Care Medicine, Respiratory Infection and Intervention Laboratory of Frontiers Science Center for Disease-related Molecular Network, and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China.
| |
Collapse
|
123
|
Wang ZY, Ren YM, Hu SW, Zhang NX, Dong MX, Li Y, Yang Y, Guo ZJ, Xu SS, Chen J, Goh AH, Chen XY. 19F qNMR based pharmacokinetics, metabolism and mass balance studies of SARS-CoV-2-3CL protease inhibitor simnotrelvir (SIM0417) in humans. Acta Pharmacol Sin 2025; 46:489-499. [PMID: 39349765 PMCID: PMC11747505 DOI: 10.1038/s41401-024-01393-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 09/05/2024] [Indexed: 01/30/2025]
Abstract
Simnotrelvir (SIM0417), an inhibitor of the 3CL protease of SARS-CoV-2, has been identified as a CYP3A sensitive substrate. This study investigated the pharmacokinetics, metabolism, and mass balance of simnotrelvir following a single oral dose of 750 mg in six healthy Chinese male subjects, co-administered with four doses of 100 mg ritonavir. Analysis using 19F qNMR combined with LC-MS/MS showed that the parent drug M0 constituted over 90% of the drug-related components in plasma. Of the administered dose, 55.4% (54.3% of M0) was recovered in urine, while 36.7% (4.57% of M0) was excreted in feces. UPLC/Q-TOF MS was used to identify metabolites in human plasma, urine and feces. Notably, oxidative metabolites catalyzed by CYP3A were scarcely detected in these matrixes. The amide hydrolyzed metabolite M9 and the cyano hydrolyzed metabolite M10 were recognized as the predominant metabolites, with the main excretion being through feces (19.0% and 12.7% of the administered dose, respectively). In vitro experiments indicated that M10 is primarily formed in the duodenum and jejunum, with further metabolism to M9 by microbiota in the large intestine. Overall, the co-administration of simnotrelvir with ritonavir led to predominant metabolism by intestinal enzymes or microbiota, resulting in hydrolyzed metabolites. These findings highlight the critical role of intestinal metabolism in the pharmacokinetics of simnotrelvir and emphasize the need to consider interactions with antibiotics and individual differences of intestinal microbiota.
Collapse
Affiliation(s)
- Ze-Yu Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yong-Mei Ren
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shu-Wei Hu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Nai-Xia Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Meng-Xiao Dong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yun Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yang Yang
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210042, China
- Simcere Zaiming Pharmaceutical Co. Ltd., Nanjing, 210042, China
| | - Zi-Jia Guo
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210042, China
- Jiangsu Simcere Pharmaceutical Co., Ltd, Nanjing, 210042, China
| | - Shan-Sen Xu
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210042, China
- Jiangsu Simcere Pharmaceutical Co., Ltd, Nanjing, 210042, China
| | - Jia Chen
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210042, China
- Simcere Zaiming Pharmaceutical Co. Ltd., Nanjing, 210042, China
| | - Aik Han Goh
- State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, 210042, China
- Jiangsu Simcere Pharmaceutical Co., Ltd, Nanjing, 210042, China
| | - Xiao-Yan Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
124
|
Yan X, Liu Y, Hu T, Huang Z, Li C, Guo L, Liu Y, Li N, Zhang H, Sun Y, Yi L, Wu J, Feng J, Zhang F, Jiang T, Tu C, He B. A compendium of 8,176 bat RNA viral metagenomes reveals ecological drivers and circulation dynamics. Nat Microbiol 2025; 10:554-568. [PMID: 39833544 DOI: 10.1038/s41564-024-01884-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 11/13/2024] [Indexed: 01/22/2025]
Abstract
Bats are natural hosts for many emerging viruses for which spillover to humans is a major risk, but the diversity and ecology of bat viruses is poorly understood. Here we generated 8,176 RNA viral metagenomes by metatranscriptomic sequencing of organ and swab samples from 4,143 bats representing 40 species across 52 locations in China. The resulting database, the BtCN-Virome, expands bat RNA virus diversity by over 3.4-fold. Some viruses in the BtCN-Virome are traced to mammals, birds, arthropods, mollusks and plants. Diet, infection dynamics and environmental parameters such as humidity and forest coverage shape virus distribution. Compared with those in the wild, bats dwelling in human settlements harboured more diverse viruses that also circulated in humans and domestic animals, including Nipah and Lloviu viruses not previously reported in China. The BtCN-Virome provides important insights into the genetic diversity, ecological drivers and circulation dynamics of bat viruses, highlighting the need for surveillance of bats near human settlements.
Collapse
Affiliation(s)
- Xiaomin Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China
| | - Yang Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China
| | - Tingsong Hu
- Southern Center for Diseases Control and Prevention, Guangzhou, Guangdong Province, China
| | - Zhenglanyi Huang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, Jilin Province, China
| | - Chenxi Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China
| | - Lei Guo
- Division of Wildlife and Plant Conservation, State Forestry and Grassland Administration, Changchun, Jilin Province, China
| | - Yuhang Liu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China
| | - Nan Li
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China
| | - Hailin Zhang
- Yunnan Institute of Endemic Diseases Control and Prevention, Dali, Yunnan Province, China
| | - Yue Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China
| | - Le Yi
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China
| | - Jianmin Wu
- Guangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jiang Feng
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, Jilin Province, China
| | - Fuqiang Zhang
- Southern Center for Diseases Control and Prevention, Guangzhou, Guangdong Province, China.
| | - Tinglei Jiang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, Jilin Province, China.
| | - Changchun Tu
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China.
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, Jiangsu Province, China.
| | - Biao He
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, Jilin Province, China.
| |
Collapse
|
125
|
Kebriaei A, Besharati R, Namdar Ahmad Abad H, Havakhah S, Khosrojerdi M, Azimian A. The relationship between microRNAs and COVID-19 complications. Noncoding RNA Res 2025; 10:16-24. [PMID: 39296641 PMCID: PMC11406673 DOI: 10.1016/j.ncrna.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/21/2024] Open
Abstract
Over the past three years, since the onset of COVID-19, several scientific studies have concentrated on understanding susceptibility to the virus, the progression of the illness, and possible long-term complexity. COVID-19 is broadly recognized with effects on multiple systems in the body, and various factors related to society, medicine, and genetics/epigenetics may contribute to the intensity and results of the disease. Additionally, a SARS-CoV-2 infection can activate pathological activities and expedite the emergence of existing health issues into clinical problems. Forming easily accessible, distinctive, and permeable biomarkers is essential for categorizing patients, preventing the disease, predicting its course, and tailoring treatments for COVID-19 individually. One promising candidate for such biomarkers is microRNAs, which could serve various purposes in understanding diverse forms of COVID-19, including susceptibility, intensity, disease progression, outcomes, and potential therapeutic options. This review provides an overview of the most significant findings related to the involvement of microRNAs in COVID-19 pathogenesis. Furthermore, it explores the function of microRNAs in a broad span of effects that may arise from accompanying or underlying health status. It underscores the value of comprehending how diverse conditions, such as neurological disorders, diabetes, cardiovascular diseases, and obesity, interact with COVID-19.
Collapse
Affiliation(s)
- Abdollah Kebriaei
- Department of Pathobiology and Laboratory Sciences, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Reza Besharati
- Department of Pathobiology and Laboratory Sciences, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hasan Namdar Ahmad Abad
- Department of Pathobiology and Laboratory Sciences, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Shahrzad Havakhah
- Department of Physiology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mahsa Khosrojerdi
- Department of Immunology and Allergy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Azimian
- Department of Pathobiology and Laboratory Sciences, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| |
Collapse
|
126
|
Wang D, Li L, Ren Z, Yu Y, Zhang Z, Zhou J, Zhao H, Zhao Z, Shi P, Mi X, Jin X, Deng Z, Li J, Chen J. Host Specificity and Geographic Dispersion Shape Virome Diversity in Rhinolophus Bats. Mol Ecol 2025; 34:e17645. [PMID: 39825599 DOI: 10.1111/mec.17645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/15/2024] [Accepted: 12/20/2024] [Indexed: 01/20/2025]
Abstract
Rhinolophus bats have been identified as natural reservoirs for viruses with global health implications, including severe acute respiratory syndrome-related coronaviruses (SARSr-CoV) and swine acute diarrhoea syndrome-related coronavirus (SADSr-CoV). In this study, we characterised the individual viromes of 603 bats to systematically investigate the diversity, abundance and geographic distribution of viral communities within R. affinis, R. sinicus and 11 other bat species. The massive metatranscriptomic data revealed substantial viral genome resources of 133 vertebrate-infecting viral clusters, which contain occasional cross-species transmission across mammalian orders and especially across bat families. Notably, those viruses included nine clusters closely related to human and/or livestock pathogens, such as SARS-CoVs and SADS-CoVs. The investigation also highlighted distinct features of viral diversity between and within bat colonies, which appear to be influenced by the distinct host population genetics of R. affinis and R. sinicus species. The comparison of SARSr-CoVs further showed varied impact of host specificity along genome-wide diversification and modular viral evolution among Rhinolophus species. Overall, the findings point to a complex interaction between host genetic diversity, and the way viruses spread and structure within natural populations, calling for continued surveillance efforts to understand factors driving viral transmission and emergence in human populations. These results present the underestimated spillover risk of bat viruses, highlighting the importance of enhancing preparedness and surveillance for emerging zoonotic viruses.
Collapse
Affiliation(s)
- Daxi Wang
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI Research, Shenzhen, China
| | - Linmiao Li
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Zirui Ren
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI Research, Shenzhen, China
| | - Yepin Yu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Zhipeng Zhang
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI Research, Shenzhen, China
| | - Jiabin Zhou
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Hailong Zhao
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI Research, Shenzhen, China
| | - Zhiwen Zhao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Peibo Shi
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xinrui Mi
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xin Jin
- BGI Research, Shenzhen, China
| | - Ziqing Deng
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI Research, Shenzhen, China
| | - Junhua Li
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI Research, Shenzhen, China
- BGI Research, Shenzhen, China
| | - Jinping Chen
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
127
|
Maison DP, Tasissa H, Deitchman A, Peluso MJ, Deng Y, Miller FD, Henrich TJ, Gerschenson M. COVID-19 clinical presentation, management, and epidemiology: a concise compendium. Front Public Health 2025; 13:1498445. [PMID: 39957982 PMCID: PMC11826932 DOI: 10.3389/fpubh.2025.1498445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/21/2025] [Indexed: 02/18/2025] Open
Abstract
Coronavirus Disease 2019, caused by severe acute respiratory coronavirus 2, has been an ever-evolving disease and pandemic, profoundly impacting clinical care, drug treatments, and understanding. In response to this global health crisis, there has been an unprecedented increase in research exploring new and repurposed drugs and advancing available clinical interventions and treatments. Given the widespread interest in this topic, this review aims to provide a current summary-for interested professionals not specializing in COVID-19-of the clinical characteristics, recommended treatments, vaccines, prevention strategies, and epidemiology of COVID-19. The review also offers a historical perspective on the pandemic to enhance understanding.
Collapse
Affiliation(s)
- David P. Maison
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Hawi Tasissa
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Amelia Deitchman
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Michael J. Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - F. DeWolfe Miller
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Timothy J. Henrich
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| |
Collapse
|
128
|
Yilmaz E, Yilmaz D, Yildiz CG, Cacan E. Upregulation of the MAP2K4 gene triggers endothelial-mesenchymal transition in COVID-19. Mol Biol Rep 2025; 52:180. [PMID: 39888478 DOI: 10.1007/s11033-025-10289-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND SARS-CoV-2 infection is marked by an excessive inflammatory response, leading to elevated production of pro-inflammatory cytokines through activation of intracellular pathways like mitogen-activated protein kinase (MAPK). Viruses can use the MAPK signaling pathway to their advantage, but the relationship of this pathway to the severe SARS-CoV-2 period has not been fully elucidated. MAP2K4 is involved in the MAPK signaling pathway and affects cellular processes such as cell-cell junction, cell proliferation, differentiation and apoptosis. METHODS AND RESULTS In this study, we sought to determine the associated biomarkers that are involved in the MAP2K4 pathway and elucidate its possible roles in terms of some clinical features associated with COVID-19. We evaluated the expressions of MAP2K4, SNAI1, SLUG, ZEB1 and E-Cadherin. For this purpose, we prospectively recruited 66 individuals, 39 of whom were women and had a mean age of 65 years. The results revealed that MAP2K4 upregulation increased SNAI1 gene expression level whereas E- Cadherin level was decreased in SARS-CoV-2 positive participants. In addition, negative correlations were determined with PLT, Lymphocyte and CKMB and E- Cadherin levels in positive participants. We also observed a negative correlation between the MAP2K4 and AST, and a positive correlation between SLUG and BUN, ZEB1 and CK. CONCLUSIONS We conclude that SARS-CoV-2 infection triggers fibrosis by increasing MAP2K4 regulation. Additionally, this is the first study to demonstrate the possible contribution of MAP2K4 in influencing COVID-19 clinical features, which may be relevant for identifying COVID-19 positive participants with severe complications.
Collapse
Affiliation(s)
- Esra Yilmaz
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Tokat Gaziosmanpasa University, Tokat, 60200, Türkiye
| | - Dilek Yilmaz
- Department of Infectious Diseases and Clinical Microbiology, Yozgat City Hospital, Tokat, 66100, Türkiye
| | - Can Gokay Yildiz
- Department of Emergency Medicine, Tokat City Hospital, Tokat, 60200, Türkiye
| | - Ercan Cacan
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Tokat Gaziosmanpasa University, Tokat, 60200, Türkiye.
| |
Collapse
|
129
|
Kakizaki M, Hashimoto R, Nagata N, Yamamoto T, Okura T, Katoh H, Kitai Y, Akahori Y, Shirato K, Ryo A, Takayama K, Takeda M. The respective roles of TMPRSS2 and cathepsins for SARS-CoV-2 infection in human respiratory organoids. J Virol 2025; 99:e0185324. [PMID: 39601592 PMCID: PMC11784140 DOI: 10.1128/jvi.01853-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
A critical aspect of the mechanism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is the protease-mediated activation of the viral spike (S) protein. The type II transmembrane serine protease TMPRSS2 is crucial for SARS-CoV-2 infection in lung epithelial Calu-3 cells and murine airways. However, the importance of TMPRSS2 needs to be re-examined because the ability to utilize TMPRSS2 is significantly reduced in the Omicron variants that spread globally. For this purpose, replication profiles of SARS-CoV-2 were analyzed in human respiratory organoids. All tested viruses, including Omicron variants, replicated efficiently in these organoids. Notably, all SARS-CoV-2 strains retained replication ability in TMPRSS2-gene knockout (KO) respiratory organoids, suggesting that TMPRSS2 is not essential for SARS-CoV-2 infection in human respiratory tissues. However, TMPRSS2-gene knockout significantly reduces the inhibitory effect of nafamostat, indicating the advantage of TMPRSS2-utilizing ability for the SARS-CoV-2 infection in these organoids. Interestingly, Omicron variants regained the TMPRSS2-utilizing ability in recent subvariants. The basal infectivity would be supported mainly by cathepsins because the cathepsin inhibitor, EST, showed a significant inhibitory effect on infection with any SARS-CoV-2 strains, mainly when used with nafamostat. A supplementary contribution of other serine proteases was also suggested because the infection of the Delta variant was still inhibited partially by nafamostat in TMPRSS2 KO organoids. Thus, various proteases, including TMPRSS2, other serine proteases, and cathepsins, co-operatively contribute to SARS-CoV-2 infection significantly in the respiratory organoids. Thus, SARS-CoV-2 infection in the human respiratory tissues would be more complex than observed in cell lines or mice. IMPORTANCE We explored how the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus infects human respiratory organoids, which are a cultured cell model made to mimic the physiological conditions of the human airways. We focused on understanding the role of different proteases of host cells in activating the virus spike proteins. Specifically, we looked at TMPRSS2, a transmembrane serine protease, and cathepsin L, a lysosomal enzyme, which helps the virus enter cells by cutting the viral spike protein. We discovered that while TMPRSS2 is crucial for the virus in certain cells and animal models, other proteases, including cathepsins and various serine proteases, also play significant roles in the SARS-CoV-2 infection of human respiratory organoids. We suggest that SARS-CoV-2 uses a more complex mechanism involving multiple proteases to infect human airways, differing from what we see in conventional cell lines or animal models. This complexity might help explain how different variants can spread and infect people effectively.
Collapse
Affiliation(s)
- Masatoshi Kakizaki
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| | - Rina Hashimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Takashi Okura
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hiroshi Katoh
- Department of Microbiology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuki Kitai
- Department of Microbiology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukiko Akahori
- Department of Microbiology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuya Shirato
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| | - Akihide Ryo
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Makoto Takeda
- Department of Microbiology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Pandemic Preparedness, Infection and Advanced Research Center, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
130
|
Batool S, Chokkakula S, Jeong JH, Baek YH, Song MS. SARS-CoV-2 drug resistance and therapeutic approaches. Heliyon 2025; 11:e41980. [PMID: 39897928 PMCID: PMC11786845 DOI: 10.1016/j.heliyon.2025.e41980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
In light of the transition of COVID-19 from a pandemic to an endemic phase, there is still a dire need to address challenges associated with drug resistance, particularly among immunocompromised and high-risk populations. This review explores the current state of research on SARS-CoV-2 drug resistance and underscores the ongoing need for effective therapeutic strategies. It critically evaluates existing knowledge on resistance mechanisms and therapeutic options, aiming to consolidate information and highlight areas for future research. By examining the complex interactions between the virus and its host, the review advocates for a multifaceted approach, including combination therapies, targeted drug development, and continuous surveillance of viral mutations. It also emphasizes the impact of evolving viral variants on antiviral efficacy and suggests adaptive treatment protocols. This review aims to enhance our understanding of SARS-CoV-2 drug resistance and contribute to more effective management of COVID-19 through a discussion of promising strategies such as drug repurposing and combination therapies.
Collapse
Affiliation(s)
- Sania Batool
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| | - Santosh Chokkakula
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| | - Ju Hwan Jeong
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| | - Yun Hee Baek
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| | - Min-Suk Song
- Department of Microbiology, Chungbuk National University, College of Medicine and Medical Research Institute, Cheongju 28644, Chungbuk, Republic of Korea
| |
Collapse
|
131
|
Zhang W, Xia L, Yuan Z, Liu M, Jiao Y, Wang Z. Simultaneous determination of nirmatrelvir, ritonavir, and beta-D-N4-hydroxycytidine in human plasma and epithelial lining fluid using LC-MS/MS and its clinical application to compare rates of achieving effective concentrations. Heliyon 2025; 11:e41737. [PMID: 39882486 PMCID: PMC11774765 DOI: 10.1016/j.heliyon.2025.e41737] [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: 07/22/2024] [Revised: 12/28/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
Currently, the trials found that the clinical efficacy of molnupiravir is lower than ritonavir-boosted nirmatrelvir. An explanation for these different efficacies in clinical treatments is still limited. The analysis method was developed and validated to simultaneously quantify nirmatrelvir, ritonavir, and beta-D-N4-hydroxycytidine (NHC) in human plasma and bronchoalveolar lavage fluid (BALF) by electrospray ionization mass spectrometry. Our method was validated over a linear range of 30-10000 ng/mL for both matrices, with precision and accuracy within 15 % across four concentrations. Recovery rates for both analytes from plasma and BALF were between 90.7-102.2 % and 90.5-107.7 %, respectively. The analytical method was then applied to monitor therapeutic drug concentrations in 59 plasma samples from 23 patients receiving ritonavir-boosted nirmatrelvir or molnupiravir. By setting target plasma concentrations of 292 ng/mL for nirmatrelvir and 1205 ng/mL for NHC, based on in vitro antiviral 90 % virus inhibitory concentrations (EC90), the drug's molecular weight and its binding to human plasma proteins, we observed that ritonavir-boosted nirmatrelvir had substantially greater rates of achieving target plasma concentrations. Additionally, we monitored epithelial lining fluid in 4 BALF samples from 4 patients and observed that NHC exhibited higher permeability in lung tissue (approximately 20 % higher than nirmatrelvir). However, subtherapeutic antiviral concentrations of NHC were also present in epithelial lining fluid. These findings highlight the importance of considering these factors in determining the efficacy of these drugs in treating coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Pharmacy, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Lin Xia
- Department of Pharmacy, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zhilong Yuan
- Department of Pharmacy, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- School of Pharmacy, Bengbu Medical College, Bengbu, 233004, China
| | - Mengdan Liu
- Department of Pharmacy, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- College of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, China
| | - Yang Jiao
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zhuo Wang
- Department of Pharmacy, Shanghai Changhai Hospital, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
132
|
Denz PJ, Papa JL, McFadden MI, Rao PR, Roettger J, Forero A, Yount JS. Accelerated Adaptation of SARS-CoV-2 Variants in Mice Lacking IFITM3 Preserves Distinct Tropism and Pathogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635150. [PMID: 39975176 PMCID: PMC11838348 DOI: 10.1101/2025.01.27.635150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Here we investigated whether interferon induced transmembrane protein 3 (IFITM3), a key antiviral protein deficient in certain human populations, affects interspecies adaptation of SARS-CoV-2. We found that SARS-CoV-2 Beta and Omicron variants passaged through IFITM3-deficient versus wild type mice exhibit enhanced replication and pathogenesis in this new host species. Enhancements associated with amino acid substitutions in the viral genome, suggesting that IFITM3 limits accumulation of adaptive mutations. Mouse-adapted viruses enabled comparative studies of variants in mice. Beta caused lung dysfunction and altered cilia-associated gene programs, consistent with broad viral antigen distribution in lungs. Omicron, which shows low pathogenicity and upper respiratory tract preference in humans, replicated to high nasal titers while showing restrained spatial distribution in lungs and diminished lung inflammatory responses compared to Beta. Our findings demonstrate that IFITM3 deficiency accelerates coronavirus adaptation and reveal that intrinsic SARS-CoV-2 variant traits shape tropism, immunity, and pathogenesis across hosts. HIGHLIGHTS IFITM3 is a critical barrier to SARS-CoV-2 adaptation in new host speciesMouse-adapted SARS-CoV-2 strains enable comparative pathologyOmicron favors nose and large airways, leading to mild lung pathologyBeta exhibits broad lung replication, driving severe inflammation and dysfunction.
Collapse
|
133
|
Shoemaker SR, Luo M, Dam KMA, Pak JE, Hoffmann MAG, Marqusee S. The Interplay of Furin Cleavage and D614G in Modulating SARS-CoV-2 Spike Protein Dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635166. [PMID: 39975226 PMCID: PMC11838386 DOI: 10.1101/2025.01.27.635166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
We report a detailed analysis of the full-length SARS-CoV-2 spike dynamics within a native-like membrane environment and variants inaccessible to studies on soluble constructs by conducting hydrogen-deuterium exchange mass spectrometry (HDX-MS) on enveloped virus-like particles (eVLPs) displaying various spike constructs. We find that the previously identified open-interface trimer conformation is sampled in all eVLP-displayed spike variants studied including sequences from engineered vaccine constructs and native viral sequences. The D614G mutation, which arose early in the pandemic, favors the canonical 'closed-interface' prefusion conformation, potentially mitigating premature S1 shedding in the presence of a cleaved furin site and providing an evolutionary advantage to the virus. Remarkably, furin cleavage at the S1/S2 boundary allosterically increases the flexibility of the S2' site, which may facilitate increased TMPRSS2 processing, enhancing viral infectivity. The use of eVLPs in HDX-MS studies provides a powerful platform for studying viral and membrane proteins in near-native environments.
Collapse
|
134
|
Han Y, Guo J, Li X, Zhong Z. Differences in clinical characteristics between coronavirus disease 2019 (COVID-19) and influenza: a systematic review and meta-analysis. NPJ Prim Care Respir Med 2025; 35:8. [PMID: 39875405 PMCID: PMC11775258 DOI: 10.1038/s41533-025-00414-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19) epidemic has brought major challenges to the global health system, and influenza is also a problem that cannot be ignored. We aimed to explore and compare the clinical characteristics of COVID-19 and influenza to deepen the understanding of these two diseases and provide some guidance for clinicians to make differential diagnoses. We searched PubMed, Embase and Web of Science for articles and performed a meta-analysis using Stata 14.0 with a random-effects model. This meta-analysis was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. One hundred articles involving 226,913 COVID-19 patients and 201,617 influenza patients were included, and all the articles included patients with these two diseases as experimental and control groups. Compared to influenza, COVID-19 was more common among men (OR = 1.46, 95% CI: 1.23-1.74) and people with a higher body mass index (MD = 1.43, 95% CI: 1.09-1.77). The proportion of current smokers among COVID-19 patients was lower than that among influenza patients (OR = 0.25, 95% CI: 0.18-0.33). Patients with COVID-19 had longer stays in the hospital (MD = 3.20, 95% CI: 2.58-3.82) and ICU (MD = 3.10, 95% CI: 1.44-4.76), required mechanical ventilation more frequently (OR = 2.30, 95% CI: 1.77-3.00), and had higher mortality (OR = 2.22, 95% CI: 1.93-2.55). We also found significant differences in some blood parameters between the two groups of patients. Upper respiratory symptoms were more obvious in influenza patients, and the proportion of comorbidities was higher than that among COVID-19 patients. There are some differences in the major characteristics, symptoms, laboratory findings and comorbidities between COVID-19 patients and influenza patients. COVID-19 patients often require more medical resources and have worse clinical outcomes.
Collapse
Affiliation(s)
- Yingying Han
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jia Guo
- Department of Respiratory, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xingzhao Li
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zhuan Zhong
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
135
|
Espejo Mambié M, San Jose-Saras D, Bischofberger Valdés C, Díaz-Agero Pérez C, Galán Montemayor JC, Martínez-García L, Abreu Di-Berardino M, Moreno-Nunez P, Vicente-Guijarro J, Aranaz-Andrés J. Environmental biocontamination by SARS-CoV-2 Virus in the hospital setting. CURRENT RESEARCH IN MICROBIAL SCIENCES 2025; 8:100355. [PMID: 39995442 PMCID: PMC11847735 DOI: 10.1016/j.crmicr.2025.100355] [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] [Indexed: 02/26/2025] Open
Abstract
Background Demonstrating the capability to isolate biological material from the environment was fundamental to supporting any transmission route. Various and inconsistent methodologies have been used to address this issue; however, the debate in scientific societies about the possibility of airborne transmission as a source of SARS-CoV-2 spread remained open. Objective To analyze SARS-CoV-2 contamination in the air and on surfaces in a hospital setting during the COVID-19 pandemic. Methods This study involved air and surface sampling in the emergency, hospitalization, and intensive care unit areas of the Ramón y Cajal University Hospital. A consistent methodology was used for all samples, and clinical and environmental parameters and characterization of each location were recorded. Results A total of 234 samples were collected, comprising 160 surface samples and 74 air samples, of which 6.84 % tested positive (13/160 surface samples and 3/74 air samples). High-contact surfaces had the highest proportion of positive samples (12/13). All positive air samples were identified within 2 m of patients who had recently developed symptoms (<5 days). High dependency and elevated temperatures seemed to indicate a higher risk of environmental biocontamination. Additionally, there was a higher risk of contamination in the intensive care units than in the hospitalization or emergency units.
Collapse
Affiliation(s)
- M Espejo Mambié
- Universidad de Alcalá, Facultad de Medicina y Ciencias de la Salud, Departamento de Biología de Sistemas, Alcalá de Henares, Spain
- Asuncion Klinika, Tolosa, Guipuzcoa, Spain
- Servicio de Medicina Preventiva y Salud Pública, Hospital Universitario Ramón y Cajal, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - D San Jose-Saras
- Servicio de Medicina Preventiva y Salud Pública, Hospital Universitario Ramón y Cajal, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- Facultad de Ciencias de la Salud, Universidad Internacional de La Rioja, Logroño, La Rioja, Spain
| | - C Bischofberger Valdés
- Servicio de Medicina Preventiva y Salud Pública, Hospital Universitario Ramón y Cajal, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - C Díaz-Agero Pérez
- Servicio de Medicina Preventiva y Salud Pública, Hospital Universitario Ramón y Cajal, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - JC Galán Montemayor
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Madrid. Spain
| | - L Martínez-García
- Servicio de Medicina Preventiva y Salud Pública, Hospital Universitario Ramón y Cajal, Spain
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Madrid. Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | | | - P Moreno-Nunez
- Universidad de Alcalá, Facultad de Medicina y Ciencias de la Salud, Departamento de Biología de Sistemas, Alcalá de Henares, Spain
- Servicio de Medicina Preventiva y Salud Pública, Hospital Universitario Ramón y Cajal, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- Facultad de Ciencias de la Salud, Universidad Internacional de La Rioja, Logroño, La Rioja, Spain
| | - J Vicente-Guijarro
- Servicio de Medicina Preventiva y Salud Pública, Hospital Universitario Ramón y Cajal, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- Facultad de Ciencias de la Salud, Universidad Internacional de La Rioja, Logroño, La Rioja, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - J.M Aranaz-Andrés
- Servicio de Medicina Preventiva y Salud Pública, Hospital Universitario Ramón y Cajal, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- Facultad de Ciencias de la Salud, Universidad Internacional de La Rioja, Logroño, La Rioja, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| |
Collapse
|
136
|
Mendoza-Ramírez NJ, García-Cordero J, Hernández-Galicia G, Moreno-Licona NJ, Hernandez J, Cabello-Gutierrez C, Zúñiga-Ramos JA, Morales-Rios E, Pérez-Tapia SM, Ortiz-Navarrete V, Espinosa-Cantellano M, Fernández-Benavides DA, Cedillo-Barrón L. Vaccination with Plasmids Encoding the Fusion Proteins D-S1, D-S1N and O-SN from SARS-CoV-2 Induces an Effective Humoral and Cellular Immune Response in Mice. Vaccines (Basel) 2025; 13:134. [PMID: 40006682 PMCID: PMC11860763 DOI: 10.3390/vaccines13020134] [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: 12/10/2024] [Revised: 01/08/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Next-generation vaccines against coronavirus disease 2019 (COVID-19) focus on inducing a long-lasting immune response against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and its emerging variants. To achieve this, antigens other than spike proteins have been proposed, and different platforms have been evaluated. Nucleic acid-based vaccines are fundamental for this process. Preclinical data have shown that the SARS-CoV-2 nucleocapsid protein induces a protective cellular immune response, and when combined with the spike protein, the resulting humoral and cellular immune responses are effective against some SARS-CoV-2 variants. METHODS We designed a DNA vaccine against the spike and nucleocapsid proteins of SARS-CoV-2 to generate fusion proteins based on the Delta and Omicron B.5 strains. The most immunogenic regions of the spike and nucleocapsid proteins of the Delta and Omicron B strains were selected using bioinformatics. The nucleotide sequences were cloned into pcDNA3.1, and named pcDNA3.1/D-S1, pcDNA3.1/D-S1N, and pcDNA3.1/O-SN. The immunogenicity of the generated fusion proteins was evaluated by analyzing the humoral and cellular responses elicited after the immunization of BALB/c mice. RESULTS DNA immunization induced antibody production, neutralization activity, and IFN-γ production. The inclusion of the nucleocapsid regions in the plasmid greatly enhanced the immune response. Moreover, cross-reactions with the variants of interest were confirmed. CONCLUSIONS Plasmids-encoding fusion proteins combining the most immunogenic regions of the spike and nucleocapsid proteins present a promising strategy for designing new and effective vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Noe Juvenal Mendoza-Ramírez
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| | - Julio García-Cordero
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| | - Gabriela Hernández-Galicia
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| | - Nicole Justine Moreno-Licona
- Departamento de Bioquímica Cinvestav, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-L.); (E.M.-R.)
| | - Jesus Hernandez
- Laboratorio de Inmunología, Centro de Investigación en Alimentación y Desarrollo A. C (CIAD) Carretera a la Victoria km 0.6, Hermosillo Sonora 83304, Mexico
| | - Carlos Cabello-Gutierrez
- Departamento de Investigación en Virología y Micología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Calzada de Tlalpan 4502, Belisario Domínguez, Tlalpan 14080, Mexico; (C.C.-G.); (J.A.Z.-R.)
| | - Joaquín Alejandro Zúñiga-Ramos
- Departamento de Investigación en Virología y Micología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Calzada de Tlalpan 4502, Belisario Domínguez, Tlalpan 14080, Mexico; (C.C.-G.); (J.A.Z.-R.)
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Monterrey 64710, Mexico
| | - Edgar Morales-Rios
- Departamento de Bioquímica Cinvestav, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-L.); (E.M.-R.)
| | - Sonia Mayra Pérez-Tapia
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City 11340, Mexico;
| | - Vianney Ortiz-Navarrete
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| | - Martha Espinosa-Cantellano
- Departamento de Infectómica y Patogénesis Molecular, CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico;
| | - David Andrés Fernández-Benavides
- Centro de Ingeniería y Desarrollo Industrial (CIDESI), Av. Playa Pie de la Cuesta No. 702, Desarrollo San Pablo, Querétaro 76125, Mexico;
| | - Leticia Cedillo-Barrón
- Departamento de Biomedicina Molecular CINVESTAV, Av. IPN # 2508 Col, San Pedro Zacatenco 07360, Mexico; (N.J.M.-R.); (J.G.-C.); (G.H.-G.)
| |
Collapse
|
137
|
Wong AP, Lau SP, Woo PY. Bats as a mixing vessel for generation of novel coronaviruses: Co-circulation and co-infection of coronaviruses and other viruses. Virology 2025; 604:110426. [PMID: 39922026 DOI: 10.1016/j.virol.2025.110426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/10/2025]
Abstract
Bats are the hosts of a wide variety of coronaviruses (CoVs) of the genera Alphacoronavirus and Betacoronavirus. The presence of more than one CoV species or strain in a single bat species greatly enhances the chance of genetic exchange among the CoVs, mainly through homologous recombination, and hence enhance the generation of novel CoV species or strains that may adapt to human or other animals and result in future epidemics. In this article, we review the evidence for co-circulation and/or co-infection of two or more CoVs in the same bat species, including co-infection with different strains of a CoV, co-circulation/co-infection of different alphaCoVs or betaCoVs, and co-circulation/co-infection of alphaCoVs and betaCoVs together. With next-generation sequencing, there has been a recent explosion of such discoveries. It is anticipated that countless more similar findings will be made in the near future.
Collapse
Affiliation(s)
- AntonioC P Wong
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China
| | - SusannaK P Lau
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China.
| | - PatrickC Y Woo
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, China; Doctoral Program in Translational Medicine and Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan; The iEGG and Animal Biotechnology Research Center, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
138
|
Figueiroa T, Galvão Bueno M, Bento Moura PE, de Oliveira MB, Passos Cordeiro JL, Santos-Cavalcante N, Camacho Antevere Mazzarotto GA, Wallau GL, Corrêa da Silva Junior L, Resende PC, Siqueira MMM, Ogrzewalska M. Alpha and Betacoronavirus Detection in Neotropical Bats from Northeast Brazil Suggests Wide Geographical Distribution and Persistence in Natural Populations. Animals (Basel) 2025; 15:332. [PMID: 39943102 PMCID: PMC11816360 DOI: 10.3390/ani15030332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/19/2024] [Accepted: 07/16/2024] [Indexed: 02/16/2025] Open
Abstract
The emergence of zoonotic viral diseases, notably exemplified by the recent coronavirus disease pandemic in 2019 (COVID-19), underscores the critical need to understand the dynamics of viruses circulating in wildlife populations. This study aimed to investigate the diversity of coronaviruses in bat populations from northeastern Brazil, particularly in the state of Ceará, where little research on bat pathogens has been conducted previously. Bat sampling was performed between March 2021 and March 2022 across three municipalities, resulting in the collection of oral and rectal swabs from 298 captured individuals. Molecular analyses revealed alphacoronaviruses in multiple bat species. Additionally, a novel Betacoronavirus was identified in Artibeus planirostris, which did not fall within an established subgenus. Phylogenetic placement of these new coronavirus sequences suggests that closely related coronavirus lineages can infect a wide range of bat species sampled in distantly related Brazilian states and biomes. No SARS-CoV-2 and influenza A viruses were found in the sampled bats. These findings expand our understanding of coronavirus diversity in Brazilian bats. The detection of coronaviruses in various bat species underscores the importance of bats as reservoirs for these viruses. The absence of SARS-CoV-2 in the sampled bats indicates a lack of spillback events from human or environmental sources. However, the potential for future transmission events underscores the importance of ongoing surveillance and transmission mitigation protocols in wildlife management practices.
Collapse
Affiliation(s)
- Thays Figueiroa
- Fundação Oswaldo Cruz, IOC, Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Rio de Janeiro 21040-900, RJ, Brazil; (T.F.); (L.C.d.S.J.); (P.C.R.); (M.M.M.S.)
| | - Marina Galvão Bueno
- Fundação Oswaldo Cruz, IOC, Laboratório de Virologia Comparada e Ambiental, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Patricia Emilia Bento Moura
- Fundação Oswaldo Cruz, IOC, Laboratório de Virologia Comparada e Ambiental, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Marcione Brito de Oliveira
- Museu Nacional, Departamento de Vertebrados, Setor de Mastozoologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 20940-040, RJ, Brazil;
| | - José Luís Passos Cordeiro
- Fundação Oswaldo Cruz, Unidade do Ceará, Área de Saúde e Ambiente, Eusébio, Ceará 61773-270, CE, Brazil;
- Plataforma Internacional para Ciência, Tecnologia e Inovação em Saúde (PICTIS), Via do Conhecimento, Edifício Central, 3830-352 Ílhavo, Portugal
| | - Nádia Santos-Cavalcante
- Museu de História Natural do Ceará Prof. Dias da Rocha, Universidade Estadual do Ceará, Pacoti, Ceará 62770-000, CE, Brazil;
- Fundação Oswaldo Cruz, Instituto Lêonidas and Maria Deane (ILDM), Unidade da Amazônia, Manaus 69057-070, AM, Brazil
| | - Giovanny A. Camacho Antevere Mazzarotto
- Fundação Oswaldo Cruz, Unidade do Ceará, Laboratório Analítico de Competências Moleculares e Epidemiológicas, Plataforma de Camelídeos e Produção de Nanocorpos, Eusébio, Ceará 61773-270, CE, Brazil;
| | - Gabriel Luz Wallau
- Fundação Oswaldo Cruz, Departamento de Entomologia e Núcleo de Bioinformática, Instituto Aggeu Magalhães (IAM), Cidade Universitária, Recife 50740-465, PE, Brazil;
- Department of Arbovirology and Entomology, Bernhard Nocht Institute for Tropical Medicine, WHO Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, National Reference Center for Tropical Infectious Diseases, 20359 Hamburg, Germany
| | - Leonardo Corrêa da Silva Junior
- Fundação Oswaldo Cruz, IOC, Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Rio de Janeiro 21040-900, RJ, Brazil; (T.F.); (L.C.d.S.J.); (P.C.R.); (M.M.M.S.)
| | - Paola Cristina Resende
- Fundação Oswaldo Cruz, IOC, Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Rio de Janeiro 21040-900, RJ, Brazil; (T.F.); (L.C.d.S.J.); (P.C.R.); (M.M.M.S.)
| | - Marilda M. Mendonça Siqueira
- Fundação Oswaldo Cruz, IOC, Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Rio de Janeiro 21040-900, RJ, Brazil; (T.F.); (L.C.d.S.J.); (P.C.R.); (M.M.M.S.)
| | - Maria Ogrzewalska
- Fundação Oswaldo Cruz, IOC, Laboratório de Vírus Respiratórios, Exantemáticos, Enterovírus e Emergências Virais, Rio de Janeiro 21040-900, RJ, Brazil; (T.F.); (L.C.d.S.J.); (P.C.R.); (M.M.M.S.)
| |
Collapse
|
139
|
Wemhöner L, Brandts C, Dinse H, Skoda EM, Jansen S, Teufel M, Rohn H, Dodel R. Consequences of COVID-19 for geriatric patients during a pandemic. Sci Rep 2025; 15:3136. [PMID: 39856128 PMCID: PMC11759943 DOI: 10.1038/s41598-024-84379-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
To investigate the outcomes of geriatric COVID-19 patients in a German academic setting during the pandemic. This study included 468 consecutive geriatric patients (≥ 70 years) who tested positive for SARS-CoV-2 and were treated at the University of Duisburg-Essen from 2/2020 to 3/2021. 74 patients were transferred to a geriatric hospital and a 12-month follow-up (prospective study) was performed in 51 patients. Clinical assessments evaluated depression (GDS), apathy (AES), cognitive status (MMST), mobility (TUG), health status (EQ-5D-5 L), and daily living activities (Barthel Index). Demographic and clinical data were also analyzed. Results showed that the mortality in this vulnerable group was 52% (n = 209). Long-term survival was higher in patients who received comprehensive geriatric treatment (74.3% vs. 51.8%). The duration of inpatient stay at the primary hospital was 13.3 ± 3.6 days, with 28.8% (n = 135) requiring intensive care. At the 12-month mark more patients with geriatric treatment lived in nursing homes. Barthel-Index/Timed-Up-and-Go-Test/MMST/AES/GDS, and EQ-5D-5 L indicated worse outcomes in the group who received geriatric treatment. Specialized geriatric care may improve survival in geriatric COVID-19 patients despite decreased long-term outcomes. Further research, including international studies like NAPKON, are encouraged to confirm these findings and explore potential interventions for improved outcomes in this vulnerable population.
Collapse
Affiliation(s)
- Ludwig Wemhöner
- Department of Geriatric Medicine, University Duisburg-Essen, Essen, Germany
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Charlotte Brandts
- Department of Geriatric Medicine, University Duisburg-Essen, Essen, Germany
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Hannah Dinse
- Clinic for Psychosomatic Medicine and Psychotherapy, LVR-University Hospital, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University of Duisburg-Essen, Essen, Germany
| | - Eva-Maria Skoda
- Clinic for Psychosomatic Medicine and Psychotherapy, LVR-University Hospital, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University of Duisburg-Essen, Essen, Germany
| | - Sarah Jansen
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Martin Teufel
- Clinic for Psychosomatic Medicine and Psychotherapy, LVR-University Hospital, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University of Duisburg-Essen, Essen, Germany
| | - Hana Rohn
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Medicine Essen University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Richard Dodel
- Department of Geriatric Medicine, University Duisburg-Essen, Essen, Germany.
- Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University of Duisburg-Essen, Essen, Germany.
- Chair of Geriatric Medicine, University Duisburg-Essen, Virchowstrasse 171, 45356, Essen, Germany.
| |
Collapse
|
140
|
Scarpa F, Branda F, Ceccarelli G, Romano C, Locci C, Pascale N, Azzena I, Fiori PL, Casu M, Pascarella S, Quaranta M, Benvenuto D, Cauda R, Ciccozzi M, Sanna D. SARS-CoV-2 XEC: A Genome-Based Survey. Microorganisms 2025; 13:253. [PMID: 40005620 PMCID: PMC11857677 DOI: 10.3390/microorganisms13020253] [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: 12/28/2024] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Recombination, a process of genetic exchange between distinct organisms, has played a critical role in the emergence of SARS-CoV-2 variants such as the XEC recombinant. This study provides a detailed genomic and structural characterization of XEC, derived from the recombination of lineages KP.3.3 (donor) and KS.1.1 (acceptor). Phylogenomic analyses reveal that XEC and its descendant XEC.1 form a monophyletic clade with close evolutionary ties to KP.3.3. The genomic breakpoint, spanning nucleotide positions 22,363-22,463, marks the shift from KS.1.1 to KP.3.3 within the spike protein gene. Mutational analysis highlights shared traits with its parental lineages, including mutations associated with immune evasion, receptor affinity, and fusogenicity. Notable changes, such as Q493E and L455S, may confer unique immunogenic properties, though XEC's overall immune escape potential is limited by the absence of new mutations in conserved epitopes. Despite these mutations, XEC demonstrates restricted geographical spread, low genetic variability, and an evolutionary trajectory indicative of an evolutionary dead-end. Bayesian Skyline Plot analysis corroborates this, showing stable but declining population size. These findings underscore the need for ongoing genomic surveillance to monitor recombinant variants' characteristics and public health impact. This study contributes to understanding viral evolution and highlights the importance of distinguishing variants of concern from those with minimal epidemiological significance.
Collapse
Affiliation(s)
- Fabio Scarpa
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.L.); (P.L.F.); (D.S.)
| | - Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (C.R.); (M.C.)
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, 00161 Rome, Italy;
| | - Chiara Romano
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (C.R.); (M.C.)
| | - Chiara Locci
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.L.); (P.L.F.); (D.S.)
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, 07100 Sassari, Italy; (N.P.); (I.A.); (M.C.)
| | - Noemi Pascale
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, 07100 Sassari, Italy; (N.P.); (I.A.); (M.C.)
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Via Vienna 2, 07100 Sassari, Italy
| | - Ilenia Azzena
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, 07100 Sassari, Italy; (N.P.); (I.A.); (M.C.)
| | - Pier Luigi Fiori
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.L.); (P.L.F.); (D.S.)
- Azienda Ospedaliera Universitaria (AOU) Sassari, 07100 Sassari, Italy
| | - Marco Casu
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, 07100 Sassari, Italy; (N.P.); (I.A.); (M.C.)
| | - Stefano Pascarella
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza, 00185 Rome, Italy; (S.P.); (M.Q.)
| | - Miriana Quaranta
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza, 00185 Rome, Italy; (S.P.); (M.Q.)
| | - Domenico Benvenuto
- Dipartimento di Sicurezza e Bioetica, Sezione di Malattie Infettive, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (D.B.); (R.C.)
| | - Roberto Cauda
- Dipartimento di Sicurezza e Bioetica, Sezione di Malattie Infettive, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (D.B.); (R.C.)
| | - Massimo Ciccozzi
- Unit of Medical Statistics and Molecular Epidemiology, Università Campus Bio-Medico di Roma, 00128 Rome, Italy; (C.R.); (M.C.)
| | - Daria Sanna
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (C.L.); (P.L.F.); (D.S.)
| |
Collapse
|
141
|
Moharram FA, Ibrahim RR, Mahgoub S, Abdel-Aziz MS, Said AM, Huang HC, Chen LY, Lai KH, Hashad N, Mady MS. Secondary metabolites of Alternaria alternate appraisal of their SARS-CoV-2 inhibitory and anti-inflammatory potentials. PLoS One 2025; 20:e0313616. [PMID: 39854441 PMCID: PMC11760621 DOI: 10.1371/journal.pone.0313616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/28/2024] [Indexed: 01/26/2025] Open
Abstract
This study identifies the secondary metabolites from Alternaria alternate and evaluates their ACE-2: Spike RBD (SARS-CoV-2) inhibitory activity confirmed via immunoblotting in human lung microvascular endothelial cells. In addition, their in vitro anti-inflammatory potential was assessed using a cell-based assay in LPS-treated RAW 264.7 macrophage cells. Two novel compounds, altenuline (1), phthalic acid bis (7'/7'' pentyloxy) isohexyl ester (2), along with 1-deoxyrubralactone (3) alternariol-5-O-methyl ether (4) and alternariol (5) were identified. Molecular docking and in vitro studies showed that compounds 2 and 4 were promising to counteract SARS-CoV-2 attachment to human ACE-2. Thus, they are considered promising natural anti-viral agents. SwissADME in silico analysis was conducted to predict the drug-like potential. Immunoblotting analysis confirmed that the tested compounds (1-4) demonstrated downregulation of ACE-2 expression in the endothelial cells from the lungs with variable degrees. Furthermore, the tested compounds (1-4) showed promising anti-inflammatory activities through TNF-α: TNFR2 inhibitory activity and their inhibitory effect on the proinflammatory cytokines (TNF-α and IL-6) in LPS-stimulated monocytes. In conclusion, our study, for the first time, provides beneficial experimental confirmation for the efficiency of the A. alternate secondary metabolites for the treatment of COVID-19 as they hinder SARS-CoV-2 infection and lower inflammatory responses initiated by SARS-CoV-2. A. alternate and its metabolites are considered in developing preventative and therapeutic tactics for COVID-19.
Collapse
Affiliation(s)
- Fatma A. Moharram
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Reham R. Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Shahenda Mahgoub
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohamed S. Abdel-Aziz
- Genetic Engineering and Biotechnology Division, Microbial Chemistry Department, National Research Centre, Giza, Egypt
| | - Ahmed M. Said
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Hui-Chi Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Lo-Yun Chen
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Kuei-Hung Lai
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Nashwa Hashad
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohamed S. Mady
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| |
Collapse
|
142
|
Kumar M, Baig MS, Bhardwaj K. Advancements in the development of antivirals against SARS-Coronavirus. Front Cell Infect Microbiol 2025; 15:1520811. [PMID: 39917633 PMCID: PMC11798951 DOI: 10.3389/fcimb.2025.1520811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/02/2025] [Indexed: 02/09/2025] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) caused an outbreak in 2002-2003, spreading to 29 countries with a mortality rate of about 10%. Strict quarantine and infection control methods quickly stopped the spread of the disease. Later research showed that SARS-CoV came from animals (zoonosis) and stressed the possibility of a similar spread from host to human, which was clearly shown by the COVID-19 outbreak. The COVID-19 pandemic, instigated by SARS-CoV-2, has affected 776 million confirmed cases and more than seven million deaths globally as of Sept 15, 2024. The existence of animal reservoirs of coronaviruses continues to pose a risk of re-emergence with improved fitness and virulence. Given the high death rate (up to 70 percent) and the high rate of severe sickness (up to 68.7 percent in long-COVID patients), it is even more critical to identify new therapies as soon as possible. This study combines research on antivirals that target SARS coronaviruses that have been conducted over the course of more than twenty years. It is a beneficial resource that might be useful in directing future studies.
Collapse
Affiliation(s)
- Mrityunjay Kumar
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, India
| | - Mirza Sarwar Baig
- Centre for Virology, School of Interdisciplinary Science and Technology, Jamia Hamdard, New Delhi, India
| | - Kanchan Bhardwaj
- Department of Biotechnology, School of Engineering and Technology, Manav Rachna International Institute of Research and Studies, Faridabad, India
- Regional Centre for Biotechnology, NCR Biotech Science Cluster, Faridabad, India
| |
Collapse
|
143
|
Varghese A, Liu J, Liu B, Guo W, Dong F, Patterson TA, Hong H. Analysis of Structures of SARS-CoV-2 Papain-like Protease Bound with Ligands Unveils Structural Features for Inhibiting the Enzyme. Molecules 2025; 30:491. [PMID: 39942596 PMCID: PMC11820935 DOI: 10.3390/molecules30030491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
The COVID-19 pandemic, driven by the novel coronavirus SARS-CoV-2, has drastically reshaped global health and socioeconomic landscapes. The papain-like protease (PLpro) plays a critical role in viral polyprotein cleavage and immune evasion, making it a prime target for therapeutic intervention. Numerous compounds have been identified as inhibitors of SARS-CoV-2 PLpro, with many characterized through crystallographic studies. To date, over 70 three-dimensional (3D) structures of PLpro complexed ligands have been deposited in the Protein Data Bank, offering valuable insight into ligand-binding features that could aid the discovery and development of effective COVID-19 treatments targeting PLpro. In this study, we reviewed and analyzed these 3D structures, focusing on the key residues involved in ligand interactions. Our analysis revealed that most inhibitors bind to PLpro's substrate recognition sites S3/S4 and SUb2. While these sites are highly attractive and have been extensively explored, other potential binding regions, such as SUb1 and the Zn(II) domain, are less explored and may hold untapped potential for future COVID-19 drug discovery and development. Our structural analysis provides insights into the molecular features of PLpro that could accelerate the development of novel therapeutics targeting this essential viral enzyme.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huixiao Hong
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (A.V.); (J.L.); (B.L.); (W.G.); (F.D.); (T.A.P.)
| |
Collapse
|
144
|
Jara R, Santos D, Reyes NS, Hermida E, Seoane A, Ypas M, Andres G, Echavarría M. Endemic coronavirus in children and adults with acute respiratory infection before the COVID-19 pandemic. Rev Argent Microbiol 2025:S0325-7541(24)00135-4. [PMID: 39848815 DOI: 10.1016/j.ram.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 10/18/2024] [Accepted: 11/13/2024] [Indexed: 01/25/2025] Open
Abstract
Acute respiratory infection (ARI) is one of the principal causes of morbidity worldwide, with respiratory viruses being common etiological agents. Among them, endemic human coronaviruses (hCoVs) including CoV-229E, CoV-OC43, CoV-NL63, and CoV-HKU1 can cause mild ARI but are usually not evaluated in the clinical setting. The aim of this work was to determine the prevalence of all respiratory pathogens, with the focus placed on endemic hCoVs in the pre-pandemic period. Circulating species, clinical associations and coinfections with other respiratory pathogens were evaluated in 510 immunocompetent patients (children and adults) with ARI using the FilmArray® Respiratory Panel (BioFire/bioMérieux). A total of 399 children (252 outpatients and 147 hospitalized) and 111 adult outpatients were enrolled in the pre-pandemic period (2008-2010 and 2016). Endemic hCoVs were the third and fifth more frequently detected viruses among adults and outpatient children, respectively, with an overall frequency close to 10%. The most prevalent species were CoV-OC43 (42.8%) and CoV-HKU1 (40.5%), followed by CoV-NL63 (19.0%) and CoV-229E (4.8%). Tachypnea, wheezing and chest indrawing were more frequent in hospitalized children compared to outpatients. All adult patients presented with symptoms of a common cold. Endemic hCoVs were detected year-round, primarily between June and November. Our results highlight their clinical relevance, and the need to include endemic hCoVs in routine screening. In the post-pandemic period, further long-term surveillance is needed for understanding the epidemiology of endemic hCoVs and their evolution, as a tool to anticipate the possible emergence of new species.
Collapse
Affiliation(s)
- Raquel Jara
- Virology Unit, Centro de Educación Médica e Investigaciones Clínicas (CEMIC) University Hospital, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Dafne Santos
- Virology Unit, Centro de Educación Médica e Investigaciones Clínicas (CEMIC) University Hospital, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Noelia Soledad Reyes
- Virology Unit, Centro de Educación Médica e Investigaciones Clínicas (CEMIC) University Hospital, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Eliana Hermida
- Virology Unit, Centro de Educación Médica e Investigaciones Clínicas (CEMIC) University Hospital, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | | | - Martin Ypas
- Emergency Service, CEMIC University Hospital, Argentina
| | - Gisela Andres
- Emergency Service, CEMIC University Hospital, Argentina
| | - Marcela Echavarría
- Virology Unit, Centro de Educación Médica e Investigaciones Clínicas (CEMIC) University Hospital, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Virology Laboratory, CEMIC University Hospital, Argentina.
| |
Collapse
|
145
|
Wang BH, Yu KY, Zhang XN, Sun XH, Tang LL, Shi XL. Fu Tu Sheng Jin Rehabilitation Formula Mitigate Airway Inflammation, Mucus Secretion and Immune Dysfunction Induced by SARS-CoV-2 Spike Protein. J Inflamm Res 2025; 18:1053-1065. [PMID: 39871960 PMCID: PMC11771161 DOI: 10.2147/jir.s480112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/27/2024] [Indexed: 01/29/2025] Open
Abstract
Objective To evaluate the effects of Fu Tu Sheng Jin Rehabilitation Formula (FTSJRF) on airway inflammation, mucus secretion, and immunoreaction in a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein-induced mouse model. Methods Forty-two mice were randomly divided into seven groups: normal, D1, D3, D10, D10H, D10M and D10L, according to the days of modeling and different dosages of FTSJRF. D1, D3, D10, D10H, D10M and D10L group mice were intratracheally administered with 15 µg SARS-CoV-2 spike protein; mice in the D10H, D10M, and D10L groups were intragastrically administered FTSJRF (46, 23 and 11.5 g/kg, respectively). Observe the pathological changes in lung tissues, expression of inflammatory factors, and mucins in different groups of mice using HE and PAS staining methods, as well as ELISA and RT-qPCR. Flow cytometry was used to detect T helper 17 (Th17)/regulatory T (Treg) cells and T helper 1(Th1)/T helper 2 (Th2) lymphocyte ratios and the proportions of conventional myeloid dendritic cells (cDCs), plasma cell-like DCs, CD80 and CD86 cells in mouse spleens. Results HE and PAS staining showed that, compared to that in the normal group, the lung tissue of the D1 group mice showed a significant inflammatory damage response, whereas the D3 and D10 groups showed a gradual recovery trend. Groups D1 and D3 showed mild mucus secretion, whereas the D10 group had excessive mucus secretion. The D10 group of mice displayed increased levels of IL-4, TNF-α, IL-33 and mucin genes such as MUC1, MUC4, etc, and FTSJRF inhibited the expression of these molecules, mucus secretion and lung damage in SARS-CoV-2 spike protein-induced mouse model. Flow cytometry results showed a decrease in the number of cDCs and an abnormal recovery of DC mature cells in the D10 group. FTSJRF increased the number of cDCs and promoted DC maturation. A higher Th17/Treg ratio was observed in the D3 and D10 groups than in the normal group, whereas this ratio decreases under the effect of FTSJRF. D10 had significantly lower Th1/Th2 ratio than normal, D1 and D3 groups, and high doses of FTSJRF increased it. Conclusion FTSJRF mitigates airway inflammation and mucus secretion induced by SARS-CoV-2 spike protein. Additionally, FTSJRF regulates immune functions by promoting DC maturation and Th17/Treg and Th1/Th2 cell homeostasis.
Collapse
Affiliation(s)
- Bo-Han Wang
- NanJing JiangNing Hospital of Chinese Medicine/Affiliated jiangning Hospital of Chinese Medicine, China Pharmaceutical University, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Ke-Yao Yu
- Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Xiao-Na Zhang
- Nanjing Hospital of Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Xian-Hong Sun
- Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
| | - Ling-Ling Tang
- School of Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Xiao-Lu Shi
- Affiliated Hospital of Nanjing University of Chinese Medicine/ Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, 210029, People’s Republic of China
| |
Collapse
|
146
|
Dong S, Fang H, Zhu J, Wu Z, Liu Y, Zhu J, Ma B, Chen Q, Yang Y. Inhalable siRNA Targeting IL-11 Nanoparticles Significantly Inhibit Bleomycin-Induced Pulmonary Fibrosis. ACS NANO 2025; 19:2742-2758. [PMID: 39791575 DOI: 10.1021/acsnano.4c15130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
For idiopathic pulmonary fibrosis (IPF), interleukin 11 (IL-11) is a pivotal cytokine that stimulates the transformation of fibroblasts into myofibroblasts, thus accelerating the progression of pulmonary fibrosis. Here, we develop an innovative inhalable small interfering RNA (siRNA) delivery system termed PEI-GBZA, which demonstrates impressive efficiency in loading siIL-11 targeting IL-11 (siIL-11) and substantially suppresses the differentiation of fibroblasts into myofibroblasts and epithelial-mesenchymal transition (EMT), reduces neutrophil and macrophage recruitment, and ultimately relieves the established fibrotic lesions in the IPF model. PEI-GBZA is prepared by modifying low-molecular-weight polyethylenimine (PEI) with 4-guanidinobenzoic acid (GBZA). The resulting PEI-GBZA may effectively encapsulate siIL-11 through a variety of interactions such as hydrophobic, hydrogen bonding, and electrostatic interactions, creating stable carrier/siIL-11 nanoparticles (PEI-GBZA/siIL-11 NPs). Upon inhalation, PEI-GBZA/siIL-11 NPs demonstrate effective retention in fibrotic lesions, leading to a marked mitigation of disease progression in a bleomycin-induced pulmonary fibrosis model. Impressively, this inhalation therapy exhibits negligible systemic toxicity. This work provides a universal and noninvasive RNA therapeutic delivery platform that holds significant promise for respiratory diseases. The potential for clinical application of this platform is substantial, offering a frontier for the treatment of IPF and potentially other pulmonary disorders.
Collapse
Affiliation(s)
- Shengting Dong
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Huapan Fang
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Junjie Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Zhiqiang Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Yi Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Jiafei Zhu
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Benting Ma
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qian Chen
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
- School of Materials Science and Engineering, Tongji University, Shanghai 201804, China
| |
Collapse
|
147
|
Bista S, Syangtan G, Darlami K, Chand AB, Bista S, Siddiqui MA, Pokhrel LR, Dawadi P, Joshi DR. Robotic versus manual disinfection of global priority pathogens at COVID-19-dedicated hospitals. Am J Infect Control 2025:S0196-6553(25)00041-0. [PMID: 39848288 DOI: 10.1016/j.ajic.2025.01.013] [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: 09/08/2024] [Revised: 01/11/2025] [Accepted: 01/12/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Twelve bacterial families identified as global priority pathogens (GPPs) pose the greatest threat to human health due to declining antibiotic efficacy. Robotics, a swift and contactless tool for disinfecting hospital surfaces, was sought to compare with manual disinfection. METHODS The disinfection efficacy of a robot was compared with manual disinfection for multiple clinical surfaces and inanimate objects at two hospitals in Nepal using bleach (NaOCl). Surfaces were swabbed pre- and post-disinfection and total heterotrophic plate count evaluated, and bacterial pathogens identified using Gram's staining and biochemical characteristics. Disinfection outcomes were reported as log reduction (log10 CFU/inch2) of heterotrophic count and presence or absence of GPPs: Staphylococcus aureus, Escherichia coli, Acinetobacter spp., and Klebsiella pneumoniae, among others. RESULTS Both robotic and manual disinfection significantly reduced the microbial load (log 2.3 to log 5.8) on hospital surfaces. No pathogens were detected post-disinfection using the robot. Robotic disinfection was more effective, significantly reducing the bacterial load (log 5.8) compared to manual disinfection (log 3.95). CONCLUSIONS Our results showed better efficacy of robotic disinfection over manual disinfection of hospital surfaces, and thus contactless robotic disinfection is recommended for disinfecting surfaces in the hospital and clinical settings as it favors patient safety against GPPs.
Collapse
Affiliation(s)
- Sayara Bista
- Central Department of Microbiology, Tribhuvan University, Kathmandu, Nepal
| | - Gopiram Syangtan
- Shi-Gan International College of Science and Technology, Tribhuvan University, Kathmandu, Nepal
| | - Kamal Darlami
- Institute of Engineering, Pulchowk Campus, Tribhuvan University, Lalitpur, Nepal
| | - Arun Bahadur Chand
- Department of Clinical Laboratory, KIST Medical College & Teaching Hospital, Lalitpur, Nepal
| | - Shrijana Bista
- Central Department of Microbiology, Tribhuvan University, Kathmandu, Nepal
| | | | - Lok R Pokhrel
- Department of Public Health, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Prabin Dawadi
- Central Department of Microbiology, Tribhuvan University, Kathmandu, Nepal; Department of Biology, The University of Mississippi, University City, MS, USA.
| | - Dev Raj Joshi
- Central Department of Microbiology, Tribhuvan University, Kathmandu, Nepal.
| |
Collapse
|
148
|
Bdeir N, Lüddecke T, Maaß H, Schmelz S, Rand U, Jacobsen H, Metzdorf K, Kulkarni U, Cossmann A, Stankov MV, Hoffmann M, Pöhlmann S, Blankenfeldt W, Dopfer-Jablonka A, Behrens GMN, Čičin-Šain L. Reverse mutational scanning of SARS-CoV-2 spike BA.2.86 identifies epitopes contributing to immune escape from polyclonal sera. Nat Commun 2025; 16:809. [PMID: 39827186 PMCID: PMC11743207 DOI: 10.1038/s41467-025-55871-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
The recently detected Omicron BA.2.86 lineage contains more than 30 amino acid mutations relative to BA.2. BA.2.86 and its JN.1 derivative evade neutralization by serum antibodies of fully vaccinated individuals. In this study, we elucidate epitopes driving the immune escape of BA.2.86 and JN.1 via pseudovirus neutralization. Here we generate 33 BA.2.86 mutants, each reverting a single mutation back to BA.2. We use this library in an approach that we call reverse mutational scanning to define distinct neutralization titers against each epitope. Mutations within the receptor binding domain at K356T, V483Δ, and to a lesser extent N460K, A484K, and F486P enhance immune escape. Interestingly, 16insMPLF within the spike N-terminal domain and P621S within S1/S2 also significantly contribute to antibody escape of BA.2.86. Upon XBB.1.5 booster vaccination, neutralization titers against JN.1 and BA.2.86 improve considerably, and residual immune escape is driven by 16insMPLF, N460K, E554K, and to a lesser extent P621S, and A484K.
Collapse
Affiliation(s)
- Najat Bdeir
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Tatjana Lüddecke
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Henrike Maaß
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefan Schmelz
- Department Structure and Function of Proteins, Helmholtz Centre for Infection Research Braunschweig, Braunschweig, Germany
| | - Ulfert Rand
- DSMZ- German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Henning Jacobsen
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Kristin Metzdorf
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Upasana Kulkarni
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Anne Cossmann
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Metodi V Stankov
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Wulf Blankenfeldt
- Department Structure and Function of Proteins, Helmholtz Centre for Infection Research Braunschweig, Braunschweig, Germany
- Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Alexandra Dopfer-Jablonka
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, partner site Hannover-Braunschweig, Hannover, Germany
| | - Georg M N Behrens
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research, partner site Hannover-Braunschweig, Hannover, Germany
- Centre for Individualized Infection Medicine, a joint venture of HZI and Hannover Medical School, Hannover, Germany
| | - Luka Čičin-Šain
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.
- German Center for Infection Research, partner site Hannover-Braunschweig, Hannover, Germany.
- Centre for Individualized Infection Medicine, a joint venture of HZI and Hannover Medical School, Hannover, Germany.
| |
Collapse
|
149
|
Chau LF, Lillico S, Opriessnig T, Blake R, Tardy L, Lee CH, Maxwell S, Warren C, Thornton E, Mclaughlin CL, McLachlan G, Tait-Burkard C, Fletcher S, Anderson S, Brown S, Gibbard L, Tzelos T, MacMillan-Christensen D, Baillie JK, Dorward DA, Griffiths DJ, Grey F. Human ACE2 transgenic pigs are susceptible to SARS-CoV-2 and develop COVID-19-like disease. Nat Commun 2025; 16:766. [PMID: 39824810 PMCID: PMC11742018 DOI: 10.1038/s41467-024-54615-1] [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: 04/03/2024] [Accepted: 11/18/2024] [Indexed: 01/20/2025] Open
Abstract
Animal models that accurately reflect COVID-19 are vital for understanding mechanisms of disease and advancing development of improved vaccines and therapeutics. Pigs are increasingly recognized as valuable models for human disease due to their genetic, anatomical, physiological, and immunological similarities to humans, and they present a more ethically viable alternative to non-human primates. However, pigs are not susceptible to SARS-CoV-2 infection which limits their utility as a model. To address this, we have developed transgenic pigs expressing human ACE2 that are susceptible to SARS-CoV-2 infection. Following challenge, clinical signs consistent with COVID-19, including fever, coughing and respiratory distress were observed, with virus replication detected in the nasal turbinates, trachea and lungs up to the study endpoint, seven days post-infection. Notably, examination of tissues revealed immunopathology in the lungs consistent with histological changes observed in fatal human COVID-19 cases. This study establishes human ACE2 transgenic pigs as a large animal model that accurately reflects many aspects of COVID-19 disease.
Collapse
Affiliation(s)
- Long Fung Chau
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Simon Lillico
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | | | - Rosemary Blake
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Luc Tardy
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Chen-Hsuin Lee
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Scott Maxwell
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Claire Warren
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Elizabeth Thornton
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Catherine L Mclaughlin
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Gerry McLachlan
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Christine Tait-Burkard
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Sarah Fletcher
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | | - J Kenneth Baillie
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
- Baillie Gifford Pandemic Science Hub, University of Edinburgh, Edinburgh, UK
| | - David A Dorward
- Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh, UK
- Department of Pathology, Royal Infirmary, Edinburgh, UK
| | | | - Finn Grey
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
150
|
Liang QZ, Ji CM, Wang B, Chen W, Cong F, Huang Y, Huang YW. Deltacoronavirus HKU11, HKU13, PDCoV (HKU15) and HKU17 spike pseudoviruses enter avian DF-1 cells via clathrin-mediated endocytosis in a Rab5-, Rab7- and pH-dependent manner. Vet Res 2025; 56:15. [PMID: 39825424 PMCID: PMC11740469 DOI: 10.1186/s13567-024-01442-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/14/2024] [Indexed: 01/20/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV), also known as HKU15, is a swine enteropathogenic virus that is believed to have originated in birds. PDCoV belongs to the genus Deltacoronavirus (DCoV), the members of which have mostly been identified in diverse avian species. We recently reported that chicken or porcine aminopeptidase N (APN), the major cellular receptor for PDCoV, can mediate cellular entry via three pseudotyped retroviruses displaying spike proteins from three avian DCoVs (HKU11, HKU13, and HKU17). In the present work, to better understand how avian-origin CoVs may be transmitted to pigs, we investigated the unknown DCoV entry pathway in avian cells. We show that clathrin-mediated endocytosis is involved in the entry of these DCoV pseudoviruses into chicken-origin DF-1 cells. Pseudovirus entry was suppressed by means of pharmacological inhibitors, dominant-negative mutants, and siRNAs targeting various cellular proteins and signalling molecules, suggesting that PDCoV and avian DCoV pseudovirus entry into DF-1 cells depends on clathrin, dynamin-2, cathepsins and a low-pH environment but is independent of caveolae and macropinocytosis. Furthermore, we found that DCoV pseudovirus entry was linked to Rab5- and Rab7-dependent pathways. This is the first report demonstrating that these DCoVs utilize clathrin-mediated endocytosis pathways to enter avian-origin cells, providing new insights into interspecies transmission of DCoVs.
Collapse
Affiliation(s)
- Qi-Zhang Liang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China
| | - Chun-Miao Ji
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Bin Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| | - Wei Chen
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China
| | - Feng Cong
- Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China.
| | - Yu Huang
- Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou, China.
| | - Yao-Wei Huang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China.
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|