1
|
Park K, Shin M, Natasha A, Kim J, Noh J, Kim SG, Kim B, Park J, Seo YR, Cho HK, Byun KS, Kim JH, Lee YS, Shim JO, Kim WK, Song JW. Novel human coronavirus in an infant patient with pneumonia, Republic of Korea. Emerg Microbes Infect 2025; 14:2466705. [PMID: 39945663 PMCID: PMC11849027 DOI: 10.1080/22221751.2025.2466705] [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: 10/17/2024] [Revised: 02/06/2025] [Accepted: 02/09/2025] [Indexed: 02/22/2025]
Abstract
Coronaviruses (CoVs) pose a significant threat to public health, causing a wide spectrum of clinical manifestations and outcomes. Beyond precipitating global outbreaks, Human CoVs (HCoVs) are frequently found among patients with respiratory infections. To date, limited attention has been directed towards alphacoronaviruses due to their low prevalence and fatality rates. Nasal swab and serum samples were collected from a paediatric patient, and an epidemiological survey was conducted. Retrospective surveillance investigated the molecular prevalence of CoV in 880 rodents collected in the Republic of Korea (ROK) from 2018 to 2022. Next-generation sequencing (NGS) and phylogenetic analyses characterized the novel HCoV and closely related CoVs harboured by Apodemus spp. On 15 December 2022, a 103-day-old infant was admitted with fever, cough, sputum production, and rhinorrhea, diagnosed with human parainfluenza virus 1 (HPIV-1) and rhinovirus co-infection. Elevated AST/ALT levels indicated transient liver dysfunction on the fourth day of hospitalization. Metagenomic NGS (mNGS) identified a novel HCoV in nasal swab and serum samples. Retrospective rodent surveillance and phylogenetic analyses showed the novel HCoV was closely related to alphacoronaviruses carried by Apodemus spp. in the ROK and China. This case highlights the potential of mNGS to identify emerging pathogens and raises awareness of possible extra-respiratory manifestations, such as transient liver dysfunction, associated with novel HCoVs. While the liver injury in this case may be attributable to the novel HCoV, further research is necessary to elucidate its clinical significance, epidemiological prevalence, and zoonotic origins.
Collapse
Affiliation(s)
- Kyungmin Park
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- Institute for Viral Diseases, Korea University College of Medicine, Seoul, Republic of Korea
| | - Minsoo Shin
- Department of Paediatrics, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Augustine Natasha
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jongwoo Kim
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Juyoung Noh
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seong-Gyu Kim
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Bohyeon Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jieun Park
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Ye-rin Seo
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hee-Kyung Cho
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kwan Soo Byun
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Korea University Medical Center, Seoul, Republic of Korea
| | - Ji Hoon Kim
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Korea University Medical Center, Seoul, Republic of Korea
| | - Young-Sun Lee
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Korea University Medical Center, Seoul, Republic of Korea
| | - Jung Ok Shim
- Department of Paediatrics, Korea University College of Medicine, Seoul, Republic of Korea
| | - Won-Keun Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
- Institute of Medical Research, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jin-Won Song
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Lian CY, Yao XY, Lv ZH, Zhang XL, Shao JW. Genetic diversity of canine coronavirus identified in dogs in yulin city, southern China. Virology 2025; 608:110528. [PMID: 40233446 DOI: 10.1016/j.virol.2025.110528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
The global outbreak of the novel coronavirus has renewed interest in related viral pathogens, including canine coronavirus (CCoV), which causes severe gastroenteritis, diarrhea, and vomiting in dogs worldwide. While cases of CCoV have been reported in China, specific instances in the Guangxi Zhuang Autonomous Region-a major center for dog breeding and consumption-have not been documented. In this study, we collected spleen tissue samples from dogs in Yulin city and conducted meta-transcriptomic sequencing. Bioinformatics analysis confirmed CCoV presence in these samples. Furthermore, virus screening and phylogenetic analyses identified the circulation of two CCoV genotypes within the dog population, revealing an overall prevalence of 14.2 %, with CCoV-IIb being the predominant genotype. Notably, two significant recombination events were detected among the analyzed strains. These findings provide valuable insights into the presence and genetic diversity of CCoV Yulin's dog populations, enhancing the understanding of its genetic variation and evolution.
Collapse
Affiliation(s)
- Chun-Yang Lian
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Xin-Yan Yao
- School of Animal Science and Technology, Foshan University, Foshan 528225, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Zhi-Hang Lv
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Xue-Lian Zhang
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Jian-Wei Shao
- School of Animal Science and Technology, Foshan University, Foshan 528225, China.
| |
Collapse
|
3
|
Anderson M, Lopez J, Wyr M, Ramirez PW. Defining diverse spike-receptor interactions involved in SARS-CoV-2 entry: Mechanisms and therapeutic opportunities. Virology 2025; 607:110507. [PMID: 40157321 DOI: 10.1016/j.virol.2025.110507] [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: 11/23/2024] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is an enveloped RNA virus that caused the Coronavirus Disease 2019 (COVID-19) pandemic. The SARS-CoV-2 Spike glycoprotein binds to angiotensin converting enzyme 2 (ACE2) on host cells to facilitate viral entry. However, the presence of SARS-CoV-2 in nearly all human organs - including those with little or no ACE2 expression - suggests the involvement of alternative receptors. Recent studies have identified several cellular proteins and molecules that influence SARS-CoV-2 entry through ACE2-dependent, ACE2-independent, or inhibitory mechanisms. In this review, we explore how these alternative receptors were identified, their expression patterns and roles in viral entry, and their impact on SARS-CoV-2 infection. Additionally, we discuss therapeutic strategies aimed at disrupting these virus-receptor interactions to mitigate COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Michael Anderson
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Julian Lopez
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Maya Wyr
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Peter W Ramirez
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA.
| |
Collapse
|
4
|
Wu H, Liu Z, Li Y. Intestinal microbiota and respiratory system diseases: Relationships with three common respiratory virus infections. Microb Pathog 2025; 203:107500. [PMID: 40139334 DOI: 10.1016/j.micpath.2025.107500] [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: 12/05/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
In recent years, the role of the intestinal microbiota in regulating host health and immune balance has attracted widespread attention. This study provides an in-depth analysis of the close relationship between the intestinal microbiota and respiratory system diseases, with a focus on three common respiratory virus infections, including respiratory syncytial virus (RSV), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and influenza virus. The research indicates that during RSV infection, there is a significant decrease in intestinal microbial diversity, suggesting the impact of the virus on the intestinal ecosystem. In SARS-CoV-2 infection, there are evident alterations in the intestinal microbiota, which are positively correlated with the severity of the disease. Similarly, influenza virus infection is associated with dysbiosis of the intestinal microbiota, and studies have shown that the application of specific probiotics exhibits beneficial effects against influenza virus infection. Further research indicates that the intestinal microbiota exerts a wide and profound impact on the occurrence and development of respiratory system diseases through various mechanisms, including modulation of the immune system and production of short-chain fatty acids (SCFAs). This article comprehensively analyzes these research advances, providing new perspectives and potential strategies for the prevention and treatment of future respiratory system diseases. This study not only deepens our understanding of the relationship between the intestinal microbiota and respiratory system diseases but also offers valuable insights for further exploring the role of host-microbiota interactions in the development of diseases.
Collapse
Affiliation(s)
- Haonan Wu
- Department of Respiratory, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; Clinical Research Center for Child Health, The First Hospital of Jilin University, Changchun, China
| | - Ziyu Liu
- The First Hospital of Jilin University, Changchun, China.
| | - Yanan Li
- Department of Respiratory, Children's Medical Center, The First Hospital of Jilin University, Changchun, China; Clinical Research Center for Child Health, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
5
|
Yasmin S, Ansari MY. A detailed examination of coronavirus disease 2019 (COVID-19): Covering past and future perspectives. Microb Pathog 2025; 203:107398. [PMID: 39986548 DOI: 10.1016/j.micpath.2025.107398] [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: 07/06/2024] [Revised: 01/07/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
The COVID-19 disease has spread rapidly across the world within just six months, affecting 169 million people and causing 3.5 million deaths globally (2021). The most affected countries include the USA, Brazil, India, and several European countries such as the UK and Russia. Healthcare professionals face new challenges in finding better ways to manage patients and save lives. In this regard, more comprehensive research is needed, including genomic and proteomic studies, personalized medicines and the design of suitable treatments. However, finding novel molecular entities (NME) using a standard or de novo strategy to drug development is a time-consuming and costly process. Another alternate strategy is discovering new therapeutic uses for old/existing/available medications, known as drug repurposing. There are a variety of computational repurposing methodologies, and some of them have been used to counter the coronavirus disease pandemic of 2019 (COVID-19). This review article compiles recently published data on the origin, transmission, pathogenesis, diagnosis, and management of the coronavirus by drug repurposing and vaccine development approach. We have attempted to screen probable drugs in clinical trials by using literature survey. This systematic review aims to create priorities for future research of drugs repurposed and vaccine development for COVID-19.
Collapse
Affiliation(s)
- Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India; Ibne Seena College of Pharmacy, Azmi Vidya Nagri Anjhi Shahabad, Hardoi, Uttar Pradesh (U.P.) 241124, India.
| |
Collapse
|
6
|
Singh S, Liu Y, Burke M, Rayaprolu V, Stein SE, Hasan SS. Production and cryo-electron microscopy structure of an internally tagged SARS-CoV-2 spike ecto-domain construct. J Struct Biol X 2025; 11:100123. [PMID: 40046771 PMCID: PMC11880631 DOI: 10.1016/j.yjsbx.2025.100123] [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: 11/01/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 03/09/2025] Open
Abstract
The SARS-CoV-2 spike protein is synthesized in the endoplasmic reticulum of host cells, from where it undergoes export to the Golgi and the plasma membrane or retrieval from the Golgi to the endoplasmic reticulum. Elucidating the fundamental principles of this bidirectional secretion are pivotal to understanding virus assembly and designing the next generation of spike genetic vaccine with enhanced export properties. However, the widely used strategy of C-terminal affinity tagging of the spike cytosolic tail interferes with proper bidirectional trafficking. Hence, the structural and biophysical investigations of spike protein trafficking have been hindered by a lack of appropriate spike constructs. Here we describe a strategy for the internal tagging of the spike protein. Using sequence analyses and AlphaFold modeling, we identified a site down-stream of the signal sequence for the insertion of a twin-strep-tag, which facilitates purification of an ecto-domain construct from the extra-cellular medium of mammalian Expi293F cells. Mass spectrometry analyses show that the internal tag has minimal impact on N-glycan modifications, which are pivotal for spike-host interactions. Single particle cryo-electron microscopy reconstructions of the spike ecto-domain reveal conformational states compatible for ACE2 receptor interactions, further solidifying the feasibility of the internal tagging strategy. Collectively, these results present a substantial advance towards reagent development for the investigations of spike protein trafficking during coronavirus infection and genetic vaccination.
Collapse
Affiliation(s)
- Suruchi Singh
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore MD 21201, USA
| | - Yi Liu
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg MD 20899, USA
| | - Meghan Burke
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg MD 20899, USA
| | - Vamseedhar Rayaprolu
- Pacific Northwest Cryo-EM Center, Oregon Health and Sciences University, Portland, OR 97201, USA
| | - Stephen E. Stein
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg MD 20899, USA
| | - S. Saif Hasan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore MD 21201, USA
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore MD 21201, USA
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Rockville MD 20850, USA
| |
Collapse
|
7
|
Zhang C, Gu H, Peng J, He B, Liu Y, Yan X, Feng J, Liu Y. Phylogenetic relationships and species composition of host community influence the transmission of coronaviruses in sympatric bats. Mol Phylogenet Evol 2025; 207:108343. [PMID: 40147782 DOI: 10.1016/j.ympev.2025.108343] [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: 10/19/2024] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Since the emergence of Severe Acute Respiratory Syndrome (SARS) in 2002, bats have been recognized as important reservoirs of diverse coronaviruses (CoVs). Despite extensive research on the broad geographic transmission of bat CoVs, there is a notable gap in understanding the transmission dynamics within sympatric bat communities. Using a phylogeographic Bayesian statistical framework, we examined CoV transmission patterns and their determinants in a region where four bat roosting caves coexist and CoVs circulate persistently. Our findings reveal that two subgenera of CoVs, α-CoVs and β-CoVs dominate different bat caves at varying times. Notably, β-CoVs show more frequent cross-species transmission events among the dominant reservoir hosts, bats of Rhinolophidae. Phylogenetic distance between host species emerges as the key influence factor of viral cross-species transmission, whereas cohabitation duration and the number of hosts sharing caves do not significantly influence viral transmission. In addition, we emphasize that the compositional similarity of species in the roosting caves is critical for the inter-cave transmission of bat-CoVs, rather than the distance between cave. These results provide novel insights into the complex transmission dynamics of bat CoVs within sympatric bat communities.
Collapse
Affiliation(s)
- Chen Zhang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China.
| | - Hao Gu
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China.
| | - Jie Peng
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China.
| | - Biao He
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China.
| | - Yuhang Liu
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China.
| | - Xiaomin Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, Jilin Province, China.
| | - Jiang Feng
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China; College of Life Science, Jilin Agricultural University, Changchun, China; Key Laboratory of Vegetation Ecology, School of Environment, Institute of Grassland Science, Northeast Normal University, Ministry of Education, Changchun, China.
| | - Ying Liu
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China; Key Laboratory of Vegetation Ecology, School of Environment, Institute of Grassland Science, Northeast Normal University, Ministry of Education, Changchun, China.
| |
Collapse
|
8
|
Maloney SM, Shaw TM, Nennig KM, Larsen MS, Shah A, Kumar A, Marcotrigiano J, Grove J, Snijder EJ, Kirchdoerfer RN, Bailey AL. CD81 is a receptor for equine arteritis virus (family: Arteriviridae). mBio 2025:e0062325. [PMID: 40422661 DOI: 10.1128/mbio.00623-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
Arteriviruses are a family of single-stranded, positive-sense RNA (+ssRNA) viruses that infect diverse animal hosts. Many arteriviruses are macrophage-tropic, consistent with their utilization of the macrophage-specific molecule CD163 as a receptor. However, the horse arterivirus (equine arteritis virus, EAV), which infects additional cell types beyond macrophages, does not utilize CD163 in its entry mechanism. Here, we use a genome-wide CRISPR knockout screen to identify alternative receptors that could explain this discrepancy in arterivirus receptor utilization and tropism, identifying the plasma membrane tetraspanin CD81 as a required host factor for EAV infection. Genetic knockout of CD81 or pre-incubation with soluble CD81 protected cells from infection with EAV, but had no impact on susceptibility to other arteriviruses. Bypassing the entry step of the viral life cycle by transfecting the EAV genome into CD81-knockout cells produced infectious EAV, implicating CD81 in the EAV entry process. Screening of CD81 orthologs from natural arterivirus hosts identified the brushtail possum CD81 as unsupportive of EAV entry, indicating that CD81 incompatibility can serve as a barrier to cross-species infection. Horse/possum CD81 chimeras were then used to map the structural domains of CD81 engaged by EAV, identifying alpha helix "D" on the large extracellular loop of CD81 as critical for EAV entry. This study identifies the first example of receptor switching in the Arteriviridae family and, given the broad tissue distribution of CD81 expression, suggests that the adoption of CD81 enabled an expansion of EAV tropism.IMPORTANCEArteriviruses are a family of diverse positive-sense RNA viruses that can infect a wide range of animal hosts, but many details regarding how arteriviruses gain entry into cells remain unclear. Most arteriviruses are thought to utilize the macrophage-specific molecule CD163 as a receptor; however, the horse arterivirus (equine arteritis virus, EAV) infects additional cell types beyond macrophages and does not utilize CD163. In this study, we identified the host factor CD81 as a significant player in EAV entry. Beyond the implications that this discovery holds for equine health, this study adds to the increasingly complex picture of arterivirus entry and demonstrates that these viruses are capable of adopting new host molecules as receptors, with consequences for the types of cells these viruses infect, the disease they cause, and their mode(s) of transmission.
Collapse
Affiliation(s)
- Sara M Maloney
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
- University of Wisconsin-Madison Cellular and Molecular Pathology Graduate Program, Madison, Wisconsin, USA
| | - Teressa M Shaw
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Kylie M Nennig
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Malorie S Larsen
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Aadit Shah
- Stanford University School of Medicine, Stanford, California, USA
| | - Ashish Kumar
- Structural Virology Section, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - Joseph Marcotrigiano
- Structural Virology Section, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - Joe Grove
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Eric J Snijder
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, South Holland, The Netherlands
| | - Robert N Kirchdoerfer
- Department of Biochemistry, Institute for Molecular Virology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Adam L Bailey
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
9
|
Peka M, Balatsky V. Binding of transmissible gastroenteritis virus and porcine respiratory coronavirus to human and porcine aminopeptidase N receptors as an indicator of cross-species transmission. PLoS One 2025; 20:e0325023. [PMID: 40424299 PMCID: PMC12111490 DOI: 10.1371/journal.pone.0325023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Coronaviruses have the ability to overcome interspecies barriers and adapt to new hosts, posing significant epidemic risks in cases of zoonotic transmission to humans. A critical factor in this process is the interaction between coronavirus spike proteins and host cell surface receptors, which plays an important role in infection and disease progression. This study focused on two representatives of coronaviruses: transmissible gastroenteritis virus (TGEV) and its mutant, porcine respiratory coronavirus (PRCV), both of which naturally cause disease in pigs. A phylogenetic analysis of previously identified strains of these viruses was performed, and the conservation of receptor-binding domain (RBD) sequences within their spike proteins was evaluated. In silico modeling was performed for complexes of the RBDs from 16 virus strains with porcine aminopeptidase N (APN), as well as for putative complexes with the human APN receptor. The binding free energy of these modeled complexes was evaluated, along with the impact of more than 500 theoretical mutations in the RBD. The computational results suggest that the TGEV 133 strain exhibits the highest affinity for both porcine and human receptors, with only two additional mutations required to further enhance this affinity. Molecular dynamics simulations were conducted for porcine and human APN complexes with known TGEV strains (Purdue and 133) as well as a theoretical mutated strain. These simulations reveal differences in the dynamic behavior of complexes with porcine and human receptors and support the hypothesis that mutagenesis at a few key amino acid residues in the RBD could enable TGEV to achieve affinity for human APN comparable to that of its natural host receptor. The findings underscore a theoretical risk of zoonotic transmission of these coronaviruses to humans, emphasizing the importance of further monitoring these pathogens.
Collapse
Affiliation(s)
- Mykyta Peka
- Department of Molecular Biology and Biotechnology, School of Biology, V. N. Karazin Kharkiv National University, Kharkiv, Ukraine
- Genetics Laboratory, Institute of Pig Breeding and Agroindustrial Production, National Academy of Agrarian Sciences of Ukraine, Poltava, Ukraine
| | - Viktor Balatsky
- Genetics Laboratory, Institute of Pig Breeding and Agroindustrial Production, National Academy of Agrarian Sciences of Ukraine, Poltava, Ukraine
| |
Collapse
|
10
|
Frasson I, Quarta S, Ruvoletto M, Biasiolo A, Chinellato M, Turato C, Maggi M, Cendron L, Richter SN, Pontisso P. SerpinB3/Protease Activated Receptor-2 Axis Is Essential for SARS CoV-2 Infection. ACS Infect Dis 2025. [PMID: 40408638 DOI: 10.1021/acsinfecdis.5c00145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Recent research has proposed several host factors required for SARS-CoV-2 infection and involved in the inflammatory response. Among these, members of the human serpin family and PAR2 have been suggested to play a relevant role. As it has been shown that one of the multiple activities of protease inhibitor SerpinB3 is the activation of PAR2, we have modulated the expression of these two molecules on both human bronchial and hepatic cells and assessed cell surface Spike binding and SARS-CoV-2 infectivity. Our findings indicate that both SerpinB3 and PAR2 play a pivotal role in viral infection and downregulate the expression of interferon-γ, a cytokine with a well-known antiviral effect. These results underscore the potential of the SerpinB3-PAR2 axis as a target for antiviral therapy and provide support for addressing serpins as targets for this purpose.
Collapse
Affiliation(s)
- Ilaria Frasson
- Department of Molecular Medicine, University of Padua, via Gabelli 63, 35128 Padua, Italy
| | - Santina Quarta
- Department of Medicine, University of Padua, via Giustiniani, 2, 35128 Padua, Italy
| | | | - Alessandra Biasiolo
- Department of Medicine, University of Padua, via Giustiniani, 2, 35128 Padua, Italy
| | - Monica Chinellato
- Department of Medicine, University of Padua, via Giustiniani, 2, 35128 Padua, Italy
| | - Cristian Turato
- Department of Molecular Medicine, University of Pavia, Viale Golgi 19, 27100 Pavia, Italy
| | - Maristella Maggi
- Department of Molecular Medicine, University of Pavia, Viale Golgi 19, 27100 Pavia, Italy
| | - Laura Cendron
- Department of Biology, University of Padua, via Bassi 58/B, 35121 Padua, Italy
| | - Sara N Richter
- Department of Molecular Medicine, University of Padua, via Gabelli 63, 35128 Padua, Italy
- Microbiology and Virology Unit, Padua University Hospital, Padua 35128, Italy
| | - Patrizia Pontisso
- Department of Medicine, University of Padua, via Giustiniani, 2, 35128 Padua, Italy
| |
Collapse
|
11
|
Lee I, Desai A, Patil A, Xu Y, Pozza-Adams K, Berdis AJ. Utilization of Flow Cytometry, Metabolomic Analyses and a Feline Infectious Peritonitis Case Study to Evaluate the Physiological Impact of Polyprenyl Immunostimulant. Cells 2025; 14:752. [PMID: 40422255 DOI: 10.3390/cells14100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/12/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Measles, hepatitis C, and COVID-19 are significant human diseases caused by RNA viruses. While vaccines exist to prevent infections, there are a small number of currently available therapeutic agents that can effectively treat these diseases after infection occurs. This study explores a new therapeutic strategy using a small molecule designated polyprenyl immunostimulant (PI) to increase innate immune responses and combat viral infections. Using a multi-disciplinary approach, this study quantifies the effects of PI in mice and THP-1 cells using flow cytometry to identify immune phenotypic markers and mass spectroscopy to monitor the metabolomic profiles of immune cells perturbed by PI treatment. The metabolomic studies identified that sphinganine and ceramide, which are precursors of sphingosine-1-phosphate (S1P), were the common metabolites upregulated in THP-1 and mice blood. Sphingosine-1-phosphate can mediate the trafficking of T cells, whereas ceramide can signal the activation and proliferation of T cells, thereby modulating the mammalian host's immunity. To demonstrate proof-of-principle, a case study was conducted to examine the benefit of administering PI to improve the outcomes of a feline co-infected with two distinct RNA viruses-feline leukemia virus and feline infectious peritonitis virus. Both viruses produce deadly symptoms that closely resemble RNA viruses that infect humans. The results identify quantifiable cellular and metabolic markers arising from PI treatment that can be used to establish a platform measuring the efficacy of PI in modulating the innate immune system.
Collapse
Affiliation(s)
- Irene Lee
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Amar Desai
- CASE Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Akshay Patil
- Department of Chemistry, Cleveland State University, Cleveland, OH 44115, USA
| | - Yan Xu
- Department of Chemistry, Cleveland State University, Cleveland, OH 44115, USA
| | | | - Anthony J Berdis
- Department of Chemistry, Cleveland State University, Cleveland, OH 44115, USA
| |
Collapse
|
12
|
Xiao X, Li S, Zheng Z, Ji Y, Du Q, Zuo Y, Miao Y, Yuan Y, Zheng H, Huang F, Wang J. Targeting USP22 to promote K63-linked ubiquitination and degradation of SARS-CoV-2 nucleocapsid protein. J Virol 2025; 99:e0223424. [PMID: 40183543 PMCID: PMC12090743 DOI: 10.1128/jvi.02234-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/13/2025] [Indexed: 04/05/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) generally hijacks the cellular machinery of host cells for survival. However, how SARS-CoV-2 employs the host's deubiquitinase to facilitate virus replication remains largely unknown. In this study, we identified the host deubiquitinase USP22 as a crucial regulator of the expression of SARS-CoV-2 nucleocapsid protein (SARS-CoV-2 NP), which is essential for SARS-CoV-2 replication. We demonstrated that SARS-CoV-2 NP proteins undergo ubiquitination-dependent degradation in host cells, while USP22 interacts with SARS-CoV-2 NP and downregulates K63-linked polyubiquitination of SARS-CoV-2 NP, thereby protecting SARS-CoV-2 NP from degradation. Importantly, we further revealed that sulbactam, an antibiotic, can reduce USP22 protein levels, eventually promoting the degradation of SARS-CoV-2 NP in vitro and in vivo. This study reveals the mechanism by which SARS-CoV-2-encoded NP protein employs host deubiquitinase for virus survival and provides a potential strategy to fight against SARS-CoV-2 infection.IMPORTANCESevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleocapsid protein (SARS-CoV-2 NP) plays a pivotal role in viral infection by binding to viral RNA, stabilizing the viral genome, and promoting replication. However, the interactions between SARS-CoV-2 NP and host intracellular proteins had not been elucidated. In this study, we provide evidence that SARS-CoV-2 NP interacts with the deubiquitinase USP22 in host cells, which downregulates SARS-CoV-2 NP ubiquitination. This reduction in ubiquitination effectively prevents intracellular degradation of SARS-CoV-2 NP, thereby enhancing its stability, marking USP22 as a potential target for antiviral strategies. Additionally, our findings indicate that sulbactam significantly decreases the protein levels of USP22, thereby reducing SARS-CoV-2 NP levels. This discovery suggests a novel therapeutic pathway in which sulbactam could be repurposed as an antiviral agent, demonstrating how certain antibiotics might contribute to antiviral treatment. This work thus opens avenues for drug repurposing and highlights the therapeutic potential of targeting host pathways to inhibit viral replication.
Collapse
Affiliation(s)
- Xin Xiao
- Department of Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shifeng Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhijin Zheng
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu, China.
| | - Yingying Ji
- Department of Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qian Du
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu, China.
| | - Yibo Zuo
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu, China.
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ying Miao
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yukang Yuan
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu, China.
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Hui Zheng
- Department of Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
- International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences (IBMS), Collaborative Innovation Center of Hematology, MOE Key Laboratory of Geriatric Disease and Immunology of Ministry of Education of China, School of Medicine, Soochow University, Suzhou, Jiangsu, China.
- Department of Laboratory Medicine, Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Fang Huang
- Department of Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
13
|
Zhou J, Liu Y, Gu T, Zhou J, Chen F, Li S. Investigating the gut bacteria structure and function of hibernating bats through 16S rRNA high-throughput sequencing and culturomics. mSystems 2025; 10:e0146324. [PMID: 40202348 PMCID: PMC12090805 DOI: 10.1128/msystems.01463-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
The gut microbiota of bats is vital for their roles in health and the ecosystem, yet studies on hibernating bats in southwest China, particularly in the unique karst landscape of Guizhou, are limited. We captured three hibernating bat species-Pipistrellus (PB), Rhinolophus (RB), and Myotis (MB)-in Liping County, collecting rectal samples for 16S rRNA amplicon sequencing. Data processing involved Trimmomatic, Flash, and Qiime2 for operational taxonomic unit (OTU) standardization and species annotation via the Greengenes database. Differential abundance was analyzed using LEfSe, and diversity metrics were assessed through alpha and beta diversity analyses. The RB group was predominantly composed of Proteobacteria (80.99%), while MB and PB exhibited diverse compositions with significant OTU richness (729 in MB). Notable genera included Hafnia and Yersinia in RB and Cosenzaea myxofaciens in MB. High proportions of unclassified taxa were observed, particularly in RB (83.81%). Functional predictions indicated metabolic pathways, with a significant representation of human diseases in PB. Culturomics revealed the successful cultivation of Huaxiibacter chinensis and Enterobacter chengduensis from bats for the first time and appears to have identified a new bacterium that is likely closely related to Clostridium paraputrificum.IMPORTANCEOur research reveals significant differences in the composition and diversity of the gut microbiota among three bat groups (PB, MB, and RB) from Guizhou. While Proteobacteria predominates in all groups, its abundance varies. Notably, the high richness of operational taxonomic units (OTUs) in the MB group suggests a more diverse microbial ecosystem, underscoring the complex interactions between species diversity, diet, gut microbiota, and overall ecological dynamics in bats. Furthermore, the substantial presence of unknown bacterial species in their intestines highlights the critical importance of cultivation-based approaches. The presence of specific taxa may have potential health implications for both bats and humans. These findings emphasize the need for further investigations into the functional roles of these microbiota and their contributions to host health. Future research should focus on longitudinal studies to elucidate these intricate interactions.
Collapse
Affiliation(s)
- Jian Zhou
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Center for Disease Control and Prevention, Guiyang, Key Laboratory of Microbio and Infectious Disease Prevention and Control in Guizhou Province, Guiyang, Guizhou, China
| | - Ying Liu
- Guizhou Center for Disease Control and Prevention, Guiyang, Key Laboratory of Microbio and Infectious Disease Prevention and Control in Guizhou Province, Guiyang, Guizhou, China
| | - Tao Gu
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jingzhu Zhou
- Guizhou Center for Disease Control and Prevention, Guiyang, Key Laboratory of Microbio and Infectious Disease Prevention and Control in Guizhou Province, Guiyang, Guizhou, China
| | - Fengming Chen
- Guizhou Center for Disease Control and Prevention, Guiyang, Key Laboratory of Microbio and Infectious Disease Prevention and Control in Guizhou Province, Guiyang, Guizhou, China
| | - Shijun Li
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Center for Disease Control and Prevention, Guiyang, Key Laboratory of Microbio and Infectious Disease Prevention and Control in Guizhou Province, Guiyang, Guizhou, China
| |
Collapse
|
14
|
Rudroff T. Climate crossroads: How global warming drives coronavirus emergence, the long COVID crisis of tomorrow, and AI's role in navigating our future. Infect Dis Now 2025; 55:105091. [PMID: 40389117 DOI: 10.1016/j.idnow.2025.105091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 05/01/2025] [Accepted: 05/14/2025] [Indexed: 05/21/2025]
Abstract
This narrative review examines the critical nexus between climate change, coronavirus emergence, and Long COVID-a triad that may shape public health outcomes for generations. Climate change disrupts ecological balances that have historically limited viral spillover events, creating novel interfaces between wildlife reservoirs and human populations. The coronavirus family presents particular concern due to its diversity, adaptability, and demonstrated capacity for cross-species transmission. With over 200 coronaviruses identified in bat populations alone, this vast reservoir of genetic diversity, combined with the family's propensity for recombination, creates substantial pandemic potential that climate disruption may further amplify. Long COVID has revealed another dimension of the coronavirus threat: the potential for significant chronic disease burden following acute infection. This complex multisystem condition affects a substantial portion of SARS-CoV-2 infected individuals, with mechanisms including viral persistence, autoimmunity, microclot formation, and mitochondrial dysfunction. Future projections suggest that climate change could increase global viral spillover risk by 30-45% by 2070, particularly in Southeast Asia, Central Africa, and parts of South America. Artificial intelligence offers promising tools for addressing these interconnected challenges through enhanced surveillance, accelerated therapeutic development, and optimized healthcare delivery. Understanding the climate-coronavirus-chronic illness nexus has become essential to the development of resilient health systems and effective planetary health policies face to an uncertain future.
Collapse
Affiliation(s)
- Thorsten Rudroff
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland.
| |
Collapse
|
15
|
Nguyen THV, Ferron F, Murakami K. Neurotoxic Implications of Human Coronaviruses in Neurodegenerative Diseases: A Perspective from Amyloid Aggregation. ACS Chem Biol 2025; 20:983-992. [PMID: 40272376 DOI: 10.1021/acschembio.5c00153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Human coronaviruses (HCoVs) include seven species: HCoV-229E, HCoV-NL63, HCoV-OC43, HCoV-HKU1, MERS-CoV, SARS-CoV-1, and SARS-CoV-2. The last three, classified as Betacoronaviruses, are highly transmissible and have caused severe pandemics. HCoV infections primarily affect the respiratory system, leading to symptoms such as dry cough, fever, and breath shortness, which can progress to acute respiratory failure and death. Beyond respiratory effects, increasing evidence links HCoVs to neurological dysfunction. However, distinguishing direct neural complications from preexisting disorders, particularly in the elderly, remains challenging. This study examines the association between HCoVs and neurodegenerative diseases like Alzheimer disease, Parkinson disease, Lewy body dementia, amyotrophic lateral sclerosis, and Creutzfeldt-Jakob disease. It also presents the long-term neurological effects of HCoV infections and their differential impact across age groups and sexes. A key aspect of this study is the investigation of the sequence and structural similarities between amyloidogenic and HCoV spike proteins, which can provide insights into potential neuropathomechanisms.
Collapse
Affiliation(s)
- Thi Hong Van Nguyen
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Francois Ferron
- Aix Marseille Univ, CNRS-Architecture et Fonction des Macromolécules Biologiques (AFMB) UMR7257, Marseille 13288, France
- European Virus Bioinformatics Center, Jena 07743, Germany
| | - Kazuma Murakami
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
16
|
Aliyari SR, Xie G, Xia X, Wang L, Zhou ZH, Cheng G. Infectivity and structure of SARS-CoV-2 after hydrogen peroxide treatment. mBio 2025; 16:e0399424. [PMID: 40257280 PMCID: PMC12077155 DOI: 10.1128/mbio.03994-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 02/18/2025] [Indexed: 04/22/2025] Open
Abstract
Hydrogen peroxide (H2O2) exhibits broad-spectrum antiviral activity and is commonly used as an over-the-counter disinfecting agent. However, its potential activities against SARS-CoV-2 have not been systematically evaluated, and mechanisms of action are not well understood. In this study, we investigate H2O2's antiviral activity against SARS-CoV-2 infection and its impact on the virion's structural integrity as compared to the commonly used fixative agent paraformaldehyde (PFA). We show that H2O2 rapidly and directly inactivates SARS-CoV-2 with a half-maximal inhibitory concentration (IC50) of 0.0015%. Cryogenic electron tomography (cryo-ET) with subtomogram averaging reveals that treatment with PFA induced the viral trimeric spike protein (S) to adopt a post-fusion conformation, and treatment of viral particles with H2O2 locked S in its pre-fusion conformation. Therefore, H2O2 treatment likely has induced modifications, such as oxidation of cysteine residues within the S subunits of the spike trimer that locked them in their pre-fusion conformation. Locking of the meta-stable pre-fusion trimer prevents its transition to the post-fusion conformation, a process essential for viral fusion with host cells and entry into host cells. Together, our cellular, biochemical, and structural studies established that hydrogen peroxide can inactivate SARS-CoV-2 in tissue culture and uncovered its underlying molecular mechanism.IMPORTANCEHydrogen peroxide (H2O2) is the commonly used, over-the-counter antiseptic solution available in pharmacies, but its effect against the SARS-CoV-2 virus has not been evaluated systematically. In this study, we show that H2O2 inactivates the SARS-CoV-2 infectivity and establish the effective concentration of this activity. Cryogenic electron tomography and sub-tomogram averaging reveal a detailed structural understanding of how H2O2 affects the SARS-CoV-2 spike in comparison with that of the commonly used fixative PFA under identical conditions. We found that PFA promoted a post-fusion conformation of the viral spike protein, while H2O2 could potentially lock the spike in its pre-fusion state. Our findings not only substantiate the disinfectant efficacy of H2O2 as a potent agent against SARS-CoV-2 but also lay the groundwork for future investigations into targeted antiviral therapies that may leverage the virus' structural susceptibilities. In addition, this study may have significant implications for developing new antiviral strategies and improving existing disinfection protocols.
Collapse
Affiliation(s)
- Saba R. Aliyari
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Guodong Xie
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
- California NanoSystems Institute, UCLA, Los Angeles, California, USA
| | - Xian Xia
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
- California NanoSystems Institute, UCLA, Los Angeles, California, USA
| | - Lulan Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Z. Hong Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
- California NanoSystems Institute, UCLA, Los Angeles, California, USA
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| |
Collapse
|
17
|
Hartmann S, Radochonski L, Ye C, Martinez-Sobrido L, Chen J. SARS-CoV-2 ORF3a drives dynamic dense body formation for optimal viral infectivity. Nat Commun 2025; 16:4393. [PMID: 40355429 PMCID: PMC12069715 DOI: 10.1038/s41467-025-59475-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
SARS-CoV-2 hijacks multiple organelles for virion assembly, of which the mechanisms have not been fully understood. Here, we identified a SARS-CoV-2-driven membrane structure named the 3a dense body (3DB). 3DBs are unusual electron-dense and dynamic structures driven by the accessory protein ORF3a via remodeling a specific subset of the trans-Golgi network (TGN) and early endosomal membrane. 3DB formation is conserved in related bat and pangolin coronaviruses but was lost during the evolution to SARS-CoV. During SARS-CoV-2 infection, 3DB recruits the viral structural proteins spike (S) and membrane (M) and undergoes dynamic fusion/fission to maintain the optimal unprocessed-to-processed ratio of S on assembled virions. Disruption of 3DB formation resulted in virions assembled with an abnormal S processing rate, leading to a dramatic reduction in viral entry efficiency. Our study uncovers the crucial role of 3DB in maintaining maximal SARS-CoV-2 infectivity and highlights its potential as a target for COVID-19 prophylactics and therapeutics.
Collapse
Affiliation(s)
- Stella Hartmann
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Lisa Radochonski
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Jueqi Chen
- Department of Microbiology, University of Chicago, Chicago, IL, USA.
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA.
| |
Collapse
|
18
|
Jafari AS, Mozaffari Nejad AS, Faraji H, Abdel-Moneim AS, Asgari S, Karami H, Kamali A, Kheirkhah Vakilabad AA, Habibi A, Faramarzpour M. Diagnostic Challenges in Fungal Coinfections Associated With Global COVID-19. SCIENTIFICA 2025; 2025:6840605. [PMID: 40370518 PMCID: PMC12077979 DOI: 10.1155/sci5/6840605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 03/11/2025] [Indexed: 05/16/2025]
Abstract
The early diagnosis of opportunistic infections is a critical concern for patient care worldwide, particularly in the context of the COVID-19 pandemic. This review examines the challenges and advancements in the management and early diagnosis of opportunistic fungal infections, which have become increasingly prominent during the pandemic. Using multiple sources, including curated databases such as PubMed and Scopus, as well as Google Scholar for broader literature searches, we systematically reviewed studies on COVID-19-associated fungal infections, with a focus on candidiasis, mucormycosis, and aspergillosis. The inclusion criteria encompassed peer-reviewed articles, clinical case reports, and cohort studies that discussed diagnostic methods, clinical outcomes, and treatment responses. Data were systematically extracted and analyzed to identify key trends and gaps in current diagnostic practices. Given the significance of opportunistic fungal infections-particularly the selected species-this review provides a comprehensive analysis of diagnostic challenges and advancements in the context of COVID-19 and beyond. Currently, there is no definitive strategy for effectively addressing these opportunistic pathogens, highlighting the need for continued research and innovation. Despite advancements in medical technology, opportunistic fungal infections continue to pose significant challenges to early and accurate diagnosis. The COVID-19 pandemic has exacerbated these challenges, with secondary fungal infections contributing to increased morbidity and mortality rates. This review highlights the complexities of diagnosing fungal coinfections and emphasizes the urgent need for improved diagnostic strategies. Enhancing the early and accurate detection of these infections is critical for effective patient management, particularly during viral pandemics. Addressing the challenges outlined in this review requires innovative diagnostic approaches to improve patient outcomes and reduce the burden of opportunistic infections on global healthcare systems.
Collapse
Affiliation(s)
- Ariyo Shahin Jafari
- Department of Medical Parasitology and Virology, Sechenov University, Moscow, Russia
| | - Amir Sasan Mozaffari Nejad
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
- Universal Scientific Education and Research Network (USERN) JMU Office, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Hossein Faraji
- Tropical and Communicable Diseases Research Center, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Ahmed S. Abdel-Moneim
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Saeme Asgari
- Department of Biochemistry and Biophysics, TeMS.C., Islamic Azad University, Tehran, Iran
| | - Hakime Karami
- Universal Scientific Education and Research Network (USERN) JMU Office, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Ali Kamali
- School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | | | - Ali Habibi
- Department of Accounting and Management, Islamic Azad University, Pardis Branch, Pardis, Iran
| | - Motahareh Faramarzpour
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran
| |
Collapse
|
19
|
Zhu H, Liu X, He J, Lei J, Zhao J. High-affinity, broad-spectrum, "centipede-like" multi-branched drug conjugates, anchored to the S protein, for blocking coronavirus infection. Eur J Med Chem 2025; 289:117450. [PMID: 40022880 DOI: 10.1016/j.ejmech.2025.117450] [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: 11/04/2024] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Over the past two decades, various coronaviruses have posed a severe threat to human life and health, with the spike protein (S protein) being a critical protein for infecting host cells. Glycyrrhizic acid (GA), as a natural drug, can inhibit the infection of coronaviruses by binding to the receptor-binding domain (RBD) of the S protein. However, issues like poor water solubility and weak binding affinity with the S protein have hindered its further application. Therefore, drawing inspiration from the biological structure of centipedes, a ROS-responsive multi-branched drug conjugate (ODPAG) was constructed through a "polymer-drug linkage" strategy using dextran as the backbone and GA as the active "claw". ODPAG exhibited drug loading of 22.0 ± 0.2% (OD40kPAG) and 19.7 ± 0.1% (OD450kPAG), showing ROS responsiveness with a half-life 6.4 times that of GA (OD40kPAG) and 5.4 times longer (OD450kPAG). In in vitro antiviral experiments, ODPAG exhibited an enhanced binding affinity to the S protein, with IC50 values of 1.33 μM (OD40kPAG) and 0.89 μM (OD450kPAG) against SARS-CoV-2 pseudovirus, demonstrating exceptional antiviral efficacy. These results collectively indicate that ODPAG can block coronavirus infection by binding to the S protein, exhibiting significant potential in addressing the current challenges posed by the novel coronavirus. Additionally, the "polymer-drug conjugate" strategy employed in this process is efficient, cost-effective, and offers new insights for combating future emergent coronaviruses.
Collapse
Affiliation(s)
- Huatai Zhu
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing, 100083, China; MOE Engineering Research Center of Forestry Biomass Materials and Bioenergy, Beijing Forestry University, Beijing, 100083, China
| | - Xuan Liu
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing, 100083, China; MOE Engineering Research Center of Forestry Biomass Materials and Bioenergy, Beijing Forestry University, Beijing, 100083, China
| | - Jing He
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing, 100083, China; MOE Engineering Research Center of Forestry Biomass Materials and Bioenergy, Beijing Forestry University, Beijing, 100083, China
| | - Jiandu Lei
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing, 100083, China; MOE Engineering Research Center of Forestry Biomass Materials and Bioenergy, Beijing Forestry University, Beijing, 100083, China.
| | - Jingyang Zhao
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery (CAS), Institute of Process Engineering, China Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
20
|
Safiriyu AA, Hussain A, Dewangan N, Kasle G, Shindler K, Pal D, Das Sarma J. The fusion peptide of the spike protein S2 domain may be a mimetic analog of β-coronaviruses and serve as a novel virus-host membrane fusion inhibitor. Antiviral Res 2025; 237:106144. [PMID: 40101846 DOI: 10.1016/j.antiviral.2025.106144] [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: 08/22/2024] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/20/2025]
Abstract
Coronavirus has garnered more attention recently, particularly in the aftermath of the 2019 pandemic. The β genus of the coronavirus family has demonstrated a significant threat to humanity. Current mitigation strategies involve the development of vaccines and repurposing drugs for symptomatic management of coronavirus infection, specifically SARS-Cov 2. Fusion inhibitors that are available as antiviral drugs for coronavirus have targeted the heptad repeat (HR) 1 and 2 in the S2 domain of the spike protein. The current study identified a fusion peptide (FP) upstream of HR1 as a potential target for developing membrane fusion inhibitors, and mimetic peptides analogous to the FP segment were tested for antiviral activity. Four mimetic fusion peptides (MFPs) (RSA59PP (MFP633), RSA59P (MFP634), RSMHV2P (MFP635), and RSMHV2PP (MFP636)) that are analogous to the FP of murine β coronavirus mouse hepatitis virus (MHV), MHV-A59/RSA59 (PP) and MHV-2/RSMHV2 (P) with central proline mutations, were tested. Results show the ability of MFPs to reduce cell-to-cell fusion and viral replication in vitro. MFP633, which contains a central double proline, exhibited the most potent inhibitory effect in spike protein-mediated membrane fusion assays. Biophysical experiments also demonstrated the strongest interactions between double-proline containing MFPs (MFP633 and MFP636) with biomimetic liposomes. In vivo studies using a liposome-mediated delivery system in mice confirmed the antiviral activity of MFP633. These findings suggest that targeting FPs could develop effective fusion inhibitors against coronaviruses. MFPs act on the host cell membrane by competing with the viral FP during the early stage of host-viral membrane fusion events. MFP633 is a promising peptide drug candidate that warrants future examination to assess whether this and other dual-proline containing peptides may exert similar anti-viral effects in other coronaviruses with conserved FP structures.
Collapse
Affiliation(s)
- Abass Alao Safiriyu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Haringhata, 741246, India
| | - Afaq Hussain
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Haringhata, 741246, India
| | - Nikesh Dewangan
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Haringhata, 741246, India
| | - Grishma Kasle
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Haringhata, 741246, India
| | - Kenneth Shindler
- Scheie Eye Institute and F. M. Kirby Center for Molecular Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bengaluru, 560012, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Haringhata, 741246, India; Department of Ophthalmology, University of Pennsylvania, USA.
| |
Collapse
|
21
|
Sacristán C, Esperón F, Sacristán I, Serra Cobo J, López Roig M, Lisón F, Millán J. Molecular survey of herpesviruses in bats from Chile and Spain reveals potentially novel species. Vet Res Commun 2025; 49:185. [PMID: 40310489 PMCID: PMC12045804 DOI: 10.1007/s11259-025-10747-3] [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: 02/09/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
Bats (order Chiroptera) are known as important hosts and reservoirs for several zoonotic viruses. To this date, most virology studies in bats have focused on RNA viruses; consequently, information about DNA viruses is more limited. Herein we surveyed the presence of herpesviruses in blood or spleen samples of three bat species of Spain (n = 31) and five bat species of Chile (n = 50) by using a broad-spectrum nested PCR. Overall, herpesvirus DNA was detected in 9.7% (3/31) bats of Spain and 10.0% (5/50) bats of Chile. Three gammaherpesvirus sequence types were found in bats from Spain, while sequence types of two betaherpesviruses, two gammaherpesviruses and one unclassified herpesvirus were detected in Chilean bats, two of which could represent novel herpesvirus species. The impact (if any) of these herpesviruses on the health of the studied species needs to be clarified. This study increases our knowledge of herpesvirus diversity in bats and expands their geographic range in South America. Future herpesvirus surveillance studies are warranted to test chiropteran families other than Vespertilionidae and Molossidae in Chile and Spain.
Collapse
Affiliation(s)
- Carlos Sacristán
- Centro de Investigación en Sanidad Animal (CISA-INIA), CSIC, Carretera Algete-El Casar de Talamanca, Km. 8,1, Valdeolmos, Madrid, 28130, Spain.
| | - Fernando Esperón
- Veterinary Department, School of Biomedical and Health Sciences, Universidad Europea de Madrid, C. Tajo, S/N, Villaviciosa de Odón, Madrid, 28670, Spain
| | - Irene Sacristán
- Centro de Investigación en Sanidad Animal (CISA-INIA), CSIC, Carretera Algete-El Casar de Talamanca, Km. 8,1, Valdeolmos, Madrid, 28130, Spain
| | - Jordi Serra Cobo
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Av. Diagonal 643, Barcelona, 08028, Spain
- Institut de Recerca de Biodiversitat (IRBio), Facultat de Biologia, Universitat de Barcelona, Av. Diagonal 643, Barcelona, 08028, Spain
| | - Marc López Roig
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Av. Diagonal 643, Barcelona, 08028, Spain
- Institut de Recerca de Biodiversitat (IRBio), Facultat de Biologia, Universitat de Barcelona, Av. Diagonal 643, Barcelona, 08028, Spain
| | - Fulgencio Lisón
- Wildlife Ecology and Conservation Lab, Zoology Department, Universidad de Concepción, Edmundo Larenas 219, Concepción, Bío Bío, 4070409, Chile
| | - Javier Millán
- Instituto Agroalimentario de Aragón-IA2 (Universidad de Zaragoza-CITA), Miguel Servet 177, Zaragoza, 50013, Spain
- Fundación ARAID, Avda. Ranillas 1, Zaragoza, 50018, Spain
- One Health Institute, Facultad de Ciencias de la Vida, Universidad Andres Bello, República 440, Santiago, 8370251, Chile
| |
Collapse
|
22
|
Wang C, Li M, Nan X, Deng Y, Fan S, Lan J. Molecular mechanisms of RaTG13 and SARS-CoV-2 RBD bound to Rhinolophus affinis bat ACE2. Protein Sci 2025; 34:e70117. [PMID: 40260962 PMCID: PMC12012733 DOI: 10.1002/pro.70117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/24/2025]
Abstract
The discovery of the RaTG13 coronavirus in Rhinolophus affinis bats in 2013, sharing 96.3% genome homology with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), suggested bats as the origin of SARS-CoV-2. Although both human angiotensin-converting enzyme 2 (hACE2) and Rhinolophus affinis bat ACE2 (bACE2-Ra, seven polymorphic variants named 01-07) are known to serve as entry receptors for these coronaviruses, the binding mechanism of RaTG13 receptor binding domain (RBD) bound to bACE2-Ra remains poorly understood. Here, we found that RaTG13 RBD bound to bACE2-Ra-07 with a weaker affinity (2.42 μM) compared with SARS-CoV-2 RBD (372 nM). Additional glycosylation at residue N370 of RaTG13 had little influence on bACE2-Ra-07 binding by RaTG13 RBD. Crystal structures of the SARS-CoV-2 and RaTG13 N370Q RBD bound to bACE2-Ra-07 were solved. Interface analysis and surface plasmon resonance (SPR) assay indicated that residue substitutions at 493, 498, 501, and 505 may play a more important role in the cross-species recognition of bACE2-Ra-07 by the SARS-CoV-2 RBD. Besides, the N370Q mutation enhanced the binding affinity between the RBD of pangolin coronavirus isolated from Guangxi (PCoV-GX) and the bACE2-Ra-07 receptor by over 10-fold. Furthermore, the recently prevalent SARS-CoV-2 variant RBDs extensively retained the interaction with the bACE2-Ra-07 receptor. Our findings give new lights on the cross-species evolution of SARS-CoV-2 and prompt the urgency to monitor the circulation of coronaviruses in bats to better prevent future spillover.
Collapse
Affiliation(s)
- Chenghai Wang
- School of Biomedical SciencesHunan UniversityChangshaChina
| | - Min Li
- Beijing Advanced Innovation Center for Structural Biology, School of Life SciencesTsinghua UniversityBeijingChina
| | - Xiaoyan Nan
- School of Biomedical SciencesHunan UniversityChangshaChina
| | - Yang Deng
- Hunan Provincial Key Laboratory of Anti‐Resistance Microbial DrugsThe Third Hospital of ChangshaChangshaChina
| | - Shilong Fan
- Beijing Advanced Innovation Center for Structural Biology, School of Life SciencesTsinghua UniversityBeijingChina
| | - Jun Lan
- School of Biomedical SciencesHunan UniversityChangshaChina
- Hunan Provincial Key Laboratory of Anti‐Resistance Microbial DrugsThe Third Hospital of ChangshaChangshaChina
| |
Collapse
|
23
|
Meseldžić N, Prnjavorac B, Dujić T, Malenica M, Prnjavorac L, Bedak O, Imamović-Kadrić S, Marjanović D, Bego T. Insights into biochemical, hematological, and coagulation parameters and their association with COVID-19 severity within four patients cohort from Bosnia and Herzegovina. Technol Health Care 2025:9287329251327481. [PMID: 40302487 DOI: 10.1177/09287329251327481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
IntroductionCOVID-19, caused by the SARS-CoV-2 virus, has resulted in a global public health crisis with a wide spectrum of clinical manifestations, ranging from asymptomatic infections to severe pneumonia. This study explores the association between various biomarkers and COVID-19 progression, aiming to identify early indicators of disease severity and enhance patient management.Materials and MethodsThe study included 750 confirmed COVID-19 patients categorized into four groups based on disease severity. Patients were recruited at the General Hospital in Tešanj, Bosnia and Herzegovina. All biochemical, hematological and coagulation parameters were analyzed using standard IFCC protocols.ResultsThe study identified significant differences in biochemical, hematological, and coagulation biomarkers across varying COVID-19 severities. Key markers such as C-reactive protein (CRP), D-dimer, lymphocyte count, erythrocyte sedimentation rate (ESR), and platelet count were analyzed. Elevated CRP and D-dimer were strongly linked to severe cases, while decreased lymphocyte count, elevated ESR, and platelet abnormalities were also associated with increased disease severity.ConclusionsOur study highlights the vital role of specific biochemical, hematological and coagulation parameters in predicting COVID-19 severity. Integrating these findings into clinical practice could enhance timely risk stratification, early intervention, and improved outcomes for affected individuals.
Collapse
Affiliation(s)
- Neven Meseldžić
- Department of Pharmaceutical biochemistry and laboratory diagnostics, University of Sarajevo, Faculty of Pharmacy, Sarajevo, Bosnia and Herzegovina
| | | | - Tanja Dujić
- Department of Pharmaceutical biochemistry and laboratory diagnostics, University of Sarajevo, Faculty of Pharmacy, Sarajevo, Bosnia and Herzegovina
| | - Maja Malenica
- Department of Pharmaceutical biochemistry and laboratory diagnostics, University of Sarajevo, Faculty of Pharmacy, Sarajevo, Bosnia and Herzegovina
| | | | - Omer Bedak
- General Hospital Tešanj, Tešanj, Bosnia and Herzegovina
| | - Selma Imamović-Kadrić
- Department of Pharmaceutical biochemistry and laboratory diagnostics, University of Sarajevo, Faculty of Pharmacy, Sarajevo, Bosnia and Herzegovina
| | - Damir Marjanović
- Institute for Anthropological Research, Zagreb, Croatia
- Department of Genetics and Bioengineering, International Burch University, Sarajevo, Bosnia and Herzegovina
- Faculty of Biotechnology and Drug Development, University of Rijeka, Rijeka, Croatia
| | - Tamer Bego
- Department of Pharmaceutical biochemistry and laboratory diagnostics, University of Sarajevo, Faculty of Pharmacy, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
24
|
Domanska-Blicharz K, Sajewicz-Krukowska J, Lisowska A, Opolska J, Tarasiuk K, Dziadek K. Genomic Alterations of the Infectious Bronchitis Virus (IBV) Strain of the GI-23 Lineage Induced by Passages in Chickens and Quails. Int J Mol Sci 2025; 26:4200. [PMID: 40362437 PMCID: PMC12071609 DOI: 10.3390/ijms26094200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/25/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Infectious bronchitis virus (IBV) of the GI-23 lineage, which first emerged in the Middle East in the late 1990s, has since spread worldwide. The factors driving its expansion, whether human involvement, wild bird migration, or the virus's biological traits, are still unclear. This study aimed to trace the genome evolution of GI-23 IBV in chickens and its adaptability to quails, which are susceptible to both gamma- and deltacoronaviruses. Thirty specific-pathogen-free (SPF) birds, aged between two and three weeks, were used. Initially, three birds were inoculated with the G052/2016 IBV via the oculo-nasal route. On the third day post-infection (dpi), oropharyngeal swabs were collected from the whole group, pooled, and subsequently used to infect three next birds. This process was repeated nine more times during consecutive IBV passages (P-I-P-X), and eventually, virus sequencing was performed using Next-Generation Sequencing (NGS). The obtained results showed that quails were not susceptible to the IBV GI-23 lineage, as the virus RNA was detected in low amounts only during the first passage (QP-I) with no further detections in later rounds of IBV passaging. In chickens, only mild diarrhea symptoms appeared in a few individuals. The NGS analysis identified sixty-two single nucleotide variants (SNVs), thirty of which caused amino acid changes, twenty-eight were synonymous, and one SNV introduced a stop codon. Three SNVs were found in untranslated regions. However, none of these SNVs lasted beyond seven passages, with forty-four being unique SNVs. The Shannon entropy values measured during passages varied for pol1a, pol1b, S, 5a, 5b, and N genes, with overall genome complexity peaking at CP-VI and CP-X. The highest complexity was observed in the pol1a (CP-X) and S genes (CP-IV, CP-VI, CP-VIII, and CP-X). Along with the S gene that was under positive selection, eight codons in pol1a were also positively selected. These findings suggest that even in an adapted host, IBV variability does not stabilize without immune pressure, indicating continuous molecular changes within its genome.
Collapse
Affiliation(s)
- Katarzyna Domanska-Blicharz
- Department of Virology and Viral Animal Diseases, National Veterinary Research Institute, al. Partyzantów 57, 24-100 Puławy, Poland; (J.S.-K.); (A.L.); (J.O.); (K.T.); (K.D.)
| | | | | | | | | | | |
Collapse
|
25
|
Medina-Gudiño J, López-Vidal Y, Pardo-Tovar JA, Velázquez-Salinas L, Basurto-Alcántara FJ. Detection of avian, murine, bovine, shrew, and bat coronaviruses in wild mammals from Mexico. Virol J 2025; 22:122. [PMID: 40287753 PMCID: PMC12034150 DOI: 10.1186/s12985-025-02724-8] [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: 11/13/2024] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Coronaviruses infect a wide range of animal and human hosts. Some human coronaviruses, such as SARS-CoV, MERS-CoV, and SARS-CoV-2, originated in animals, with bats often serving as ancestral hosts. This study analyzed samples from wild animals in three Mexican states, using an RT-PCR assay targeting the RdRp gene to detect and genotype coronaviruses, assessing their potential role as reservoirs. Phylogenetic analysis was conducted to determine the genetic relationships of the identified coronaviruses. Gammacoronavirus RNA was identified in fallow deer, llamas, spider monkeys, and mouflons; Betacoronavirus RNA in mouflons and dwarf goats; and Alphacoronavirus RNA in dwarf goats and ponies. The detected viral sequences exhibited high nucleotide identity with known coronaviruses, including Avian coronavirus (Gammacoronavirus), Murine coronavirus (Betacoronavirus), Betacoronavirus 1 (Betacoronavirus), Wénchéng shrew coronavirus (unclassified Alphacoronavirus), and Bat coronavirus HKU10 (Alphacoronavirus). These findings represent the first report of Avian coronavirus, Murine coronavirus, Wénchéng shrew coronavirus, and Bat coronavirus HKU10 in these species, as well as the first detection of Avian coronavirus in llamas, spider monkeys, and mouflons. This study provides valuable insights into the potential role of wildlife as coronavirus reservoirs, highlighting the importance of monitoring these viruses to mitigate future zoonotic transmission risks.
Collapse
Affiliation(s)
- Jocelyn Medina-Gudiño
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de Mexico, México
| | - Yolanda López-Vidal
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de Mexico, México
| | - J Adolfo Pardo-Tovar
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de Mexico, México
| | - Lauro Velázquez-Salinas
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, United States Department of Agriculture-Agricultural Research Service, Greenport, NY, USA
| | - Francisco Javier Basurto-Alcántara
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de Mexico, México.
| |
Collapse
|
26
|
Wang C, Nan X, Deng Y, Fan S, Li X, Lan J. Structural insights into the receptor-binding domain of bat coronavirus ZXC21. Structure 2025:S0969-2126(25)00138-8. [PMID: 40306273 DOI: 10.1016/j.str.2025.04.004] [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: 12/18/2024] [Revised: 03/05/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Bat coronaviruses ZXC21 and ZC45 were discovered before the COVID-19 outbreak and share approximately 86% genome homology with SARS-CoV-2. Earlier studies indicated that ZXC21 and ZC45 may be involved in the emergence of SARS-CoV-2. However, the cell invasion mechanisms of ZXC21 and ZC45 remain unclear. Here, we determined the crystal structure of the ZXC21 receptor-binding domain (RBD) and found that the core structure shared high similarity with SARS-CoV-2, MERS-CoV, human coronavirus (HCoV)-HKU1, SARS-CoV, and HCoV-OC43 RBDs, whereas the receptor-binding motifs (RBMs) differ. We demonstrated that the ZXC21 RBD had no interaction with the human coronavirus receptors angiotensin-converting enzyme 2 (ACE2), dipeptidylpeptidase 4 (DPP4), aminopeptidase N (APN), or transmembrane serine protease 2 (TMPRSS2) by surface plasmon resonance (SPR). Moreover, the P5S-3B11 Fab can bind to the ZXC21 RBD, indicating that this SARS-CoV-2 core-targeting antibody may retain neutralizing activity toward the ZXC21 coronavirus. Our results revealed the bat coronavirus ZXC21 RBD structure, which may provide further insights into the evolution of SARS-CoV-2 and the other human beta-coronaviruses.
Collapse
Affiliation(s)
- Chenghai Wang
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
| | - Xiaoyan Nan
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, China
| | - Yang Deng
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Shilong Fan
- Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xin Li
- Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, The Third Hospital of Changsha, Changsha, Hunan, China
| | - Jun Lan
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, China; Hunan Provincial Key Laboratory of Anti-Resistance Microbial Drugs, The Third Hospital of Changsha, Changsha, Hunan, China.
| |
Collapse
|
27
|
Jiang W, Huang C, Muyldermans S, Jia L. Small but Mighty: Nanobodies in the Fight Against Infectious Diseases. Biomolecules 2025; 15:610. [PMID: 40427503 PMCID: PMC12109223 DOI: 10.3390/biom15050610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/20/2025] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Infectious diseases, caused by pathogenic microorganisms and capable of spreading, pose a significant threat to global public health. Developing efficient and cost-effective techniques for treating infectious diseases is crucial in curbing their progression and reducing patients' morbidity and mortality. Nanobodies (Nbs), a novel class of affinity reagents derived from unique heavy chain-only antibodies in camelids, represent the smallest intact and fully functional antigen-binding fragments. Compared with conventional antibodies and their antigen binding fragments, Nbs offer numerous advantages, including high affinity, exceptional target specificity, cost-effective production, easy accessibility, and robust stability, demonstrating immense potential in infectious disease treatment. This review introduces Nbs and focuses on discussing their mechanisms and intervention strategies in the treatment of viral and bacterial infections.
Collapse
Affiliation(s)
- Wenning Jiang
- Department of Public Security Administration, Liaoning Police College, Dalian 116036, China
| | - Chundong Huang
- Dalian Kangyuan Medical Technology Co., Ltd., Dalian 116014, China
| | - Serge Muyldermans
- Dalian Kangyuan Medical Technology Co., Ltd., Dalian 116014, China
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Lingyun Jia
- The School of Bioengineering, Dalian University of Technology, Dalian 116036, China
| |
Collapse
|
28
|
Zhang S, Cao Y, Huang Y, Zhang X, Mou C, Qin T, Chen Z, Bao W. Abortive PDCoV infection triggers Wnt/β-catenin pathway activation, enhancing intestinal stem cell self-renewal and promoting chicken resistance. J Virol 2025; 99:e0013725. [PMID: 40135895 PMCID: PMC11998530 DOI: 10.1128/jvi.00137-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025] Open
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging coronavirus causing economic losses to swine industries worldwide. PDCoV can infect chickens under laboratory conditions, usually with no symptoms or mild symptoms, and may cause outbreaks in backyard poultry and wildfowl, posing a potential risk of significant economic loss to the commercial poultry industry. However, the reasons for such a subdued reaction after infection are not known. Here, using chicken intestinal organoid monolayers, we found that although PDCoV infects them nearly as well as porcine intestinal organoid monolayers, infection did not result in detectable amounts of progeny virus. In ex vivo and in vivo experiments using chickens, PDCoV infection failed to initiate interferon and inflammatory responses. Additionally, infection did not result in a disrupted intestinal barrier nor a reduced number of goblet cells and mucus secretion, as in pigs. In fact, the number of goblet cells increased as did the secreted mucus, thereby providing an enhanced protective barrier. Ex vivo PDCoV infection in chicken triggered activation of the Wnt/β-catenin pathway with the upregulation of Wnt/β-catenin pathway genes (Wnt3a, Lrp5, β-catenin, and TCF4) and Wnt target genes (Lgr5, cyclin D1, and C-myc). This activation stimulates the self-renewal of intestinal stem cells (ISCs), accelerating ISC-mediated epithelial regeneration by significant up-regulation of PCNA (transiently amplifying cells), BMI1 (ISCs), and Lyz (Paneth cells). Our data demonstrate that abortive infection of PDCoV in chicken cells activates the Wnt/β-catenin pathway, which facilitates the self-renewal and proliferation of ISCs, contributing to chickens' resistance to PDCoV infection.IMPORTANCEThe intestinal epithelium is the main target of PDCoV infection and serves as a physical barrier against pathogens. Additionally, ISCs are charged with tissue repair after injury, and promoting rapid self-renewal of intestinal epithelium will help to re-establish the physical barrier and maintain intestinal health. We found that PDCoV infection in chicken intestinal organoid monolayers resulted in abortive infection and failed to produce infectious virions, disrupt the intestinal barrier, reduce the number of goblet cells and mucus secretion, and induce innate immunity, but rather increased goblet cell numbers and mucus secretion. Abortive PDCoV infection activated the Wnt/β-catenin pathway, enhancing ISC renewal and accelerating the renewal and replenishment of shed PDCoV-infected intestinal epithelial cells, thereby enhancing chicken resistance to PDCoV infection. This study provides novel insights into the mechanisms underlying the mild or asymptomatic response to PDCoV infection in chickens, which is critical for understanding the virus's potential risks to the poultry industry.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanan Cao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Yanjie Huang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Xueli Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Chunxiao Mou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Tao Qin
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhenhai Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
29
|
Guillebaud J, Ou TP, Hul V, Hoem T, Meng C, Nuon S, Hoem S, Lim R, Khun L, Furey NM, Cappelle J, Duong V, Chevalier V. Study of coronavirus diversity in wildlife in Northern Cambodia suggests continuous circulation of SARS-CoV-2-related viruses in bats. Sci Rep 2025; 15:12628. [PMID: 40221475 PMCID: PMC11993651 DOI: 10.1038/s41598-025-92475-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: 09/20/2024] [Accepted: 02/27/2025] [Indexed: 04/14/2025] Open
Abstract
Since SARS-CoV-2's emergence, studies in Southeast Asia, including Cambodia, have identified related coronaviruses (CoVs) in rhinolophid bats. This pilot study investigates the prevalence and diversity of CoVs in wildlife from two Cambodian provinces known for wildlife trade and environmental changes, factors favoring zoonotic spillover risk. Samples were collected from 2020 to 2022 using active (capture and swabbing of bats and rodents) and non-invasive (collection of feces from bat caves and wildlife habitats) methods. RNA was screened for CoVs using conventional pan-CoVs and real-time Sarbecovirus-specific PCR systems. Positive samples were sequenced and phylogenetic analysis was performed on the partial RdRp gene. A total of 2608 samples were collected: 867 rectal swabs from bats, 159 from rodents, 41 from other wild animals, and 1541 fecal samples. The overall prevalence of CoVs was 2.0%, with a 3.3% positive rate in bats, 2.5% in rodents, and no CoVs detected in other wildlife species. Alpha-CoVs were exclusive to bats, while Beta-CoVs were found in both bats and rodents. Seven SARS-CoV-2-related viruses were identified in Rhinolophus shameli bats sampled in August 2020, March 2021, and December 2021. Our results highlight diverse CoVs in Cambodian bats and rodents and emphasize bats as significant reservoirs. They also suggest continuous circulation of bat SARS-CoV-2-related viruses may occur in a region where ecological and human factors could favor virus emergence. Continuous surveillance and integrated approaches are crucial to managing and mitigating emerging zoonotic diseases.
Collapse
Affiliation(s)
- Julia Guillebaud
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia.
- International Centre of Research in Agriculture for Development (CIRAD), UMR ASTRE, Montpellier, France.
| | - Tey Putita Ou
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Vibol Hul
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Thavry Hoem
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Chana Meng
- Department of Wildlife and Biodiversity, Forestry Administration, Ministry of Agriculture, Forestry and Fisheries, Phnom Penh, Cambodia
| | - Sithun Nuon
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Sreyleak Hoem
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Reaksa Lim
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Limmey Khun
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | | | - Julien Cappelle
- International Centre of Research in Agriculture for Development (CIRAD), UMR ASTRE, Montpellier, France
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Véronique Chevalier
- International Centre of Research in Agriculture for Development (CIRAD), UMR ASTRE, Montpellier, France
- Epidemiology and Clinical Research Unit, Institut Pasteur de Madagascar, Antananarivo, Madagascar
- CIRAD, UMR ASTRE, Antananarivo, Madagascar
| |
Collapse
|
30
|
Yang Q, Zong K, Zhao X, Zhang F, Li F, Li X. Design, Synthesis, Evaluation, and Molecular Dynamics Simulation of SARS-CoV-2 M pro Inhibitors. ACS Med Chem Lett 2025; 16:668-674. [PMID: 40236562 PMCID: PMC11995235 DOI: 10.1021/acsmedchemlett.5c00065] [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: 02/01/2025] [Revised: 03/05/2025] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
COVID-19, caused by SARS-CoV-2, is a highly contagious disease with significant transmissibility and pathogenicity. The main protease of SARS-CoV-2 (Mpro or 3CLpro) is crucial for viral replication, making it a key therapeutic target. Nirmatrelvir, a promising Mpro inhibitor, contains a trifluoroacetyl group in its P4 fragment, which presents opportunities for further optimization. This study aims to enhance the inhibitory activity of nirmatrelvir through structural modification of the P4 fragment. Using a computer-aided drug design (CADD) approach, 11 novel compounds were identified based on molecular docking scores, binding free energy, predicted ADMET properties, structural diversity, synthetic feasibility, and inhibitory activity. IC50 measurements and molecular dynamics (MD) simulations demonstrated significant inhibitory potential for most compounds, with IC50 values ranging from 0.0435-0.9989 μM. Notably, compounds 2-5a and 2-5f exhibited inhibitory activity against SARS-CoV-2 Mpro comparable to that of nirmatrelvir. These findings offer valuable insights for the development of anti-SARS-CoV-2 therapeutics.
Collapse
Affiliation(s)
- Qinghua Yang
- College of
Petrochemical Engineering, Liaoning Petrochemical
University, Liaoning 113001, China
- Beijing
Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China
| | - Keli Zong
- College
of
Chemistry and Life Science, Beijing University
of Technology, Beijing 100124, P. R. China
- Beijing
Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China
| | - Xu Zhao
- Department
of Hepatology, Fifth Medical Center of Chinese
PLA General Hospital, 100 West Fourth Ring Road, Beijing100071, China
| | - Fenghua Zhang
- College of
Petrochemical Engineering, Liaoning Petrochemical
University, Liaoning 113001, China
| | - Fei Li
- College of
Petrochemical Engineering, Liaoning Petrochemical
University, Liaoning 113001, China
| | - Xingzhou Li
- Beijing
Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China
| |
Collapse
|
31
|
Purushotham JN, Lutz HL, Parker E, Andersen KG. Immunological drivers of zoonotic virus emergence, evolution, and endemicity. Immunity 2025; 58:784-796. [PMID: 40168990 PMCID: PMC11981831 DOI: 10.1016/j.immuni.2025.03.014] [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: 10/10/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/03/2025]
Abstract
The disruption of natural ecosystems caused by climate change and human activity is amplifying the risk of zoonotic spillover, presenting a growing global health threat. In the past two decades, the emergence of multiple zoonotic viruses has exposed critical gaps in our ability to predict epidemic trajectories and implement effective interventions. RNA viruses, in particular, are challenging to control due to their high mutation rates and ability to adapt and evade immune defenses. To better prepare for future outbreaks, it is vital that we deepen our understanding of the factors driving viral emergence, transmission, and persistence in human populations. Specifically, deciphering the interactions between antibody-mediated immunity and viral evolution will be key. In this perspective, we explore these dynamic relationships and highlight research priorities that may guide the development of more effective strategies to mitigate the impact of emerging infectious diseases.
Collapse
Affiliation(s)
- Jyothi N Purushotham
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA
| | - Holly L Lutz
- Denver Museum of Nature and Science, Denver, CO, USA
| | - Edyth Parker
- The Institute of Genomics and Global Health (IGH), Redeemer's University, Ede, Osun, Nigeria
| | - Kristian G Andersen
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA; Scripps Research Translational Institute, La Jolla, CA, USA.
| |
Collapse
|
32
|
Bowers KH, De Angelis D, Birrell PJ. Modelling with SPEED: a Stochastic Predictor of Early Epidemic Detection. J Theor Biol 2025; 607:112120. [PMID: 40189138 DOI: 10.1016/j.jtbi.2025.112120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 04/20/2025]
Abstract
The frequency of emerging infectious disease outbreaks continues to rise, necessitating predictive frameworks for public health decision-making. This study introduces the Stochastic Predictor of Early Epidemic Detection (SPEED) model, an adaptation of the classic Susceptible-Infected-Recovered model, employing a Gillespie-like algorithm to simulate early-stage stochastic disease transmission. SPEED incorporates individual-level detection probabilities based on the infection time and the lag from GP consultation to lab confirmation. The model dynamically adjusts to public health responses by enhancing testing and reducing detection times once a single case has been identified. SPEED serves two key functionalities. First, as a statistical inference tool refining reproduction number estimates following the detection of a small number of cases. SPEED inference uses specified prior distributions for the reproduction number to provide reliable posterior estimates. Second, to simulate epidemic scenarios under specified values of the reproduction number in order to construct a distribution of the time to subsequent detections. The model is used to evaluate how second case timings can rule out higher values of the reproduction number. Comparisons with simulations under heightened surveillance scenarios demonstrate the model's utility in assessing response efficacy on the initial outbreak spread. Our results demonstrate SPEED applied to a single case of influenza A(H1N2)v, detected through routine flu surveillance on the 23rd November 2023.
Collapse
Affiliation(s)
| | - Daniela De Angelis
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK; Analysis & Intelligence Assessment Directorate, Chief Data Officer Group, UKHSA, UK
| | - Paul J Birrell
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK; Analysis & Intelligence Assessment Directorate, Chief Data Officer Group, UKHSA, UK.
| |
Collapse
|
33
|
Riccetti N, Signorelli S, Fanelli A, Massaro E, Bacco M, Szewczyk W, Ibarreta D, Ciscar JC, Cescatti A, Coecke S, Capua I. The urgency of addressing zoonotic diseases surveillance: Potential opportunities considering One Health approaches and common European Data Spaces. Data Brief 2025; 59:111332. [PMID: 40018645 PMCID: PMC11867294 DOI: 10.1016/j.dib.2025.111332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/22/2024] [Accepted: 01/21/2025] [Indexed: 03/01/2025] Open
Abstract
Currently, transdisciplinary data from animal surveillance that are available for One Health approaches to public health are scarce, negatively impacting our ability to anticipate and prepare for future public health threats, particularly those involving zoonotic diseases with pandemic or epidemic potential. In this article, we explore the potential of the common European Data Spaces framework to enhance the availability of animal surveillance data, in order to better address public health threats. We propose building upon and expanding existing initiatives, such as the European Data Spaces for Health, Agriculture, and Green Deal, to design innovative services. These services could enable the integration of different data sources to inform research and policymaking on public health interventions. An overarching layer, populated with data and generating integrative information, could support a One Health approach to research and policymaking for the preparedness and anticipation of zoonotic diseases. Consequently, this approach might foster data sharing from Member States by leveraging existing developments within data spaces in terms of, for example, data security. It could also support researchers and developers in accessing transdisciplinary, stratified, and quality-controlled data for their projects.
Collapse
Affiliation(s)
- Nicola Riccetti
- European Commission, Joint Research Centre (JRC). Via E. Fermi 2749, 21027 Ispra Italy
| | - Serena Signorelli
- European Commission, Joint Research Centre (JRC). Via E. Fermi 2749, 21027 Ispra Italy
| | - Angela Fanelli
- European Commission, Joint Research Centre (JRC). Via E. Fermi 2749, 21027 Ispra Italy
- European Commission, Joint Research Centre (JRC). C. Inca Garcilaso 3, 41092 Sevilla Spain
| | - Emanuele Massaro
- European Commission, Joint Research Centre (JRC). Via E. Fermi 2749, 21027 Ispra Italy
| | - Manlio Bacco
- European Commission, Joint Research Centre (JRC). Via E. Fermi 2749, 21027 Ispra Italy
| | - Wojciech Szewczyk
- European Commission, Joint Research Centre (JRC). C. Inca Garcilaso 3, 41092 Sevilla Spain
| | - Dolores Ibarreta
- European Commission, Joint Research Centre (JRC). C. Inca Garcilaso 3, 41092 Sevilla Spain
| | - Juan Carlos Ciscar
- European Commission, Joint Research Centre (JRC). C. Inca Garcilaso 3, 41092 Sevilla Spain
| | - Alessandro Cescatti
- European Commission, Joint Research Centre (JRC). Via E. Fermi 2749, 21027 Ispra Italy
| | - Sandra Coecke
- European Commission, Joint Research Centre (JRC). Via E. Fermi 2749, 21027 Ispra Italy
| | - Ilaria Capua
- Johns Hopkins University SAIS Europe. Via B. Andreatta 3, 40126 Bologna Italy
| |
Collapse
|
34
|
Li H, Song C, Li Y, Zhang T, Yang X, Wang H. Genome-wide CRISPR screen reveals host factors for gama- and delta-coronavirus infection in Huh7 cells. Int J Biol Macromol 2025; 304:140728. [PMID: 39920943 DOI: 10.1016/j.ijbiomac.2025.140728] [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: 11/21/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Genome-wide CRISPR screening has emerged as a powerful tool for identifying novel host factors involved in viral infections. In recent years, host factors for several Alpha- and Beta-coronaviruses have been systematically screened and characterized. However, knowledge regarding Gamma- and Delta-coronavirus infections remains limited. In this study, we conducted genome-scale CRISPR knockout (KO) screening in Huh7 cells infected with infectious bronchitis virus (IBV), a Gamma-coronavirus, and porcine deltacoronavirus (PDCoV), a Delta-coronavirus. We identified known host factors for PDCoV, including APN and TMEM41B. We confirmed that human APN does not serve as a critical host factor for IBV. Notably, SPPL3 was identified as a key factor involved in viral particle entry and S protein-induced syncytium formation through the modulation of cellular N-glycosylation. Furthermore, we performed a meta-analysis integrating all Huh7 cell-based genome-wide CRISPR screens across the four genera of coronaviruses (Alpha-, Beta-, Gamma-, and Delta-coronaviruses). Our analysis highlighted conserved host pathways, particularly those related to proteoglycans, glycoproteins, and vesicle trafficking. TMEM41B, SCAP, and FAM98A emerged as the most frequently targeted host genes. These findings provide valuable insights into the life cycles of IBV and PDCoV infections and facilitate the development of host-directed therapeutic strategies.
Collapse
Affiliation(s)
- Hao Li
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Cailiang Song
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Yuqing Li
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Tiejun Zhang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Xin Yang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China
| | - Hongning Wang
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu 610064, China; Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Chengdu 610064, China.
| |
Collapse
|
35
|
Singh A, Jangid K, Nehul S, Dhaka P, Rani R, Pareek A, Sharma GK, Kumar P, Tomar S. Structural and Mechanistic Insights into the Main Protease (Mpro) Dimer Interface Destabilization Inhibitor: Unveiling New Therapeutic Avenues against SARS-CoV-2. Biochemistry 2025; 64:1589-1605. [PMID: 39882595 DOI: 10.1021/acs.biochem.4c00535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
SARS-CoV-2 variant recurrence has emphasized the imperative prerequisite for effective antivirals. The main protease (Mpro) of SARS-CoV-2 is crucial for viral replication, making it one of the prime and promising antiviral targets. Mpro features several druggable sites, including active sites and allosteric sites near the dimerization interface, that regulate its catalytic activity. This study identified six highly efficacious antiviral SARS-CoV-2 compounds (WIN-62577, KT185, bexarotene, ledipasvir, diacerein, and simepervir) using structure-based virtual screening of compound libraries against Mpro. Using SPR and ITC, the binding of selected inhibitory compounds to the target Mpro was validated. The FRET-based protease assay demonstrated that the identified molecules effectively inhibit Mpro with IC50 values in the range from 0.64 to 11.98 μM. Additionally, in vitro cell-based antiviral assays showed high efficacy with EC50 values in the range of 1.51 to 18.92 μM. The crystal structure of the Mpro-minocycline complex detailed the possible inhibition mechanism of minocycline, an FDA-approved antibiotic. Minocycline binds to an allosteric site, revealing residues critical for the loss of protease activity due to destabilization of molecular interactions at the dimeric interface, which are crucial for the proteolytic activity of Mpro. The study suggests that the binding of minocycline to the allosteric site may play a role in Mpro dimer destabilization and direct the rational design of minocycline derivatives as antiviral drugs.
Collapse
Affiliation(s)
- Ankur Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| | - Kuldeep Jangid
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| | - Sanketkumar Nehul
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| | - Preeti Dhaka
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| | - Ruchi Rani
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| | - Akshay Pareek
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| | - Gaurav Kumar Sharma
- Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, Uttarakhand 247667, India
| |
Collapse
|
36
|
Abstract
For decades, scientists have relied on traditional animal models to study viral infection and the immune response. However, these models have limitations, and the search for more accurate and reliable ways to study the human-pathogen interphase has led to the development of humanized mouse systems. These revolutionary models have transformed how we understand viral infection and the human immune system's interactions with viruses to control or exacerbate disease. They are also paving the way for new treatments and therapies. In this article, we explore the history and development of humanized mouse systems and their advantages, limitations, and applications in viral immunology research. We describe the different types of humanized mouse models, including their generation and utility for studying human pathogens, with an emphasis on human-specific viruses. In addition, we discuss areas for further refinement and future applications.
Collapse
Affiliation(s)
- Angela Wahl
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA ; ,
| | - J Victor Garcia
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA ; ,
| |
Collapse
|
37
|
Sun S, He J, Liu L, Zhu Y, Zhang Q, Qiu Y, Han Y, Xue S, Peng X, Long Y, Lu T, Wu W, Xia A, Zhou Y, Yan Y, Gao Y, Lu L, Sun L, Xie M, Wang Q. Anti-S2 antibodies responsible for the SARS-CoV-2 infection-induced serological cross-reactivity against MERS-CoV and MERS-related coronaviruses. Front Immunol 2025; 16:1541269. [PMID: 40226608 PMCID: PMC11985752 DOI: 10.3389/fimmu.2025.1541269] [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: 12/07/2024] [Accepted: 03/06/2025] [Indexed: 04/15/2025] Open
Abstract
Sarbecoviruses, such as SARS-CoV-2, utilize angiotensin-converting enzyme 2 (ACE2) as the entry receptor; while merbecoviruses, such as MERS-CoV, use dipeptidyl peptidase 4 (DPP4) for viral entry. Recently, several MERS-related coronaviruses, NeoCoV and PDF-2180, were reported to use ACE2, the same receptor as SARS-CoV-2, to enter cells, raising the possibility of potential recombination between SARS-CoV-2 and MERS-related coronaviruses within the co-infected ACE2-expressing cells. However, facing this potential recombination risk, the serum and antibody cross-reactivity against MERS/MERS-related coronaviruses after SARS-CoV-2 vaccination and/or infection is still elusive. Here, in this study, we showed that the serological cross-reactivity against MERS/MERS-related S proteins could be induced by SARS-CoV-2 infection but not by inactivated SARS-CoV-2 vaccination. Further investigation revealed that this serum cross-reactivity is due to monoclonals recognizing relatively conserved S2 epitopes, such as fusion peptide and stem helix, but not by antibodies against the receptor-binding domain (RBD), N-terminal domain (NTD) or subdomain-1 (SD1). Some of these anti-S2 cross-reactive mAbs showed cross-neutralizing activity, while none of them exhibited antibody-dependent enhancement (ADE) effect of viral entry in vitro. Together, these results dissected the SARS-CoV-2 infection-induced serological cross-reactivity against MERS/MERS-related coronaviruses, and highlighted the significance of conserved S2 region for the design and development of pan-β-coronaviruses vaccines.
Collapse
Affiliation(s)
- Siyuan Sun
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiaying He
- Microbiological Testing Department, Baoshan District Center for Disease Control and Prevention, Shanghai, China
| | - Luotian Liu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuzhen Zhu
- Department of Gastroenterology, Jingan District Central Hospitals, Fudan University, Shanghai, China
| | - Qingsong Zhang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yinong Qiu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuru Han
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Song Xue
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaofang Peng
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yiming Long
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tianyu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wei Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Anqi Xia
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yunjiao Zhou
- Fundamental Research Center, Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Yan Yan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yidan Gao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lei Sun
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Minxiang Xie
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qiao Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Fifth People’s Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
38
|
de Souza JÂ, de Souza Gomes I, de Souza Fernandes L, Andrade LAF, de Souza LÂ, de Almeida Paiva V, Araujo SC, de Lima LHF, Dias RS, de Melo-Minardi RC, da Fonseca FG, de Paula SO, de Azevedo Silveira S. In vitro enzymatic and cell culture assays for SARS-CoV-2 main protease interaction with ambenonium. Sci Rep 2025; 15:10606. [PMID: 40148508 PMCID: PMC11950299 DOI: 10.1038/s41598-025-94283-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
The 2019 pandemic of coronavirus disease (COVID-19) caused by SARS-CoV-2 led to millions of deaths worldwide since its emergence. The viral genomic material can code structural and non-structural proteins including the main protease or 3CLpro, a cysteine protease that cleavages the viral polyprotein generating 11 proteins that participate in viral pre-replication. Thus, 3CLpro is a promising therapeutic target for SARS-CoV-2 inhibition by new drugs or drug repositioning because 3CLpro is dissimilar to human proteases. We conducted in vitro assays demonstrating the modulation activity of ambenonium, a drug already used in Myasthenia gravis that acts by inhibiting the action of acetylcholinesterase, and had its potential inhibitory activity against viral replication pointed out in a previous in silico study. In concentrations of 100 µM, 50 µM, 25 µM, 10 µM, and 1 µM there was no inhibition in the formation of lysis plates, with a slight increase in the genome copy number at the higher concentrations evaluated. However, in the concentrations of 0,1 µM and 0,01 µM, there was a reduction in the number of lysis plates. This behavior suggests that the ambenonium acts as a modulator of viral activity in vitro. To investigate potential conformational changes in the protein between dimeric and monomeric forms in the presence of the compound, a local docking analysis was performed. Results indicated this conformational shift is possible, though further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Juliana Ângelo de Souza
- Department of Computer Science, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil.
| | - Isabela de Souza Gomes
- Department of Computer Science, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | | | - Luis Adan Flores Andrade
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luciana Ângelo de Souza
- Department of Biochemistry and Molecular Biology, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | | | - Sheila Cruz Araujo
- Department of Bioinformatics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
- Department of Exact and Biological Sciences, Universidade Federal de São João del-Rei, Sete Lagoas Campus, Sete Lagoas, Minas Gerais, Brazil
| | - Leonardo Henrique Franca de Lima
- Department of Exact and Biological Sciences, Universidade Federal de São João del-Rei, Sete Lagoas Campus, Sete Lagoas, Minas Gerais, Brazil
| | - Roberto Sousa Dias
- Department of General Biology, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | | | | | | | - Sabrina de Azevedo Silveira
- Department of Computer Science, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
- IDATA - Institute of Artificial Intelligence and Computational Science, Viçosa, Minas Gerais, Brazil
| |
Collapse
|
39
|
Nappi F. Myocarditis and Inflammatory Cardiomyopathy in Dilated Heart Failure. Viruses 2025; 17:484. [PMID: 40284927 PMCID: PMC12031395 DOI: 10.3390/v17040484] [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: 02/01/2025] [Revised: 03/16/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Inflammatory cardiomyopathy is a condition that is characterised by the presence of inflammatory cells in the myocardium, which can lead to a significant deterioration in cardiac function. The etiology of this condition involves multiple factors, both infectious and non-infectious causes. While it is primarily associated with viral infections, other potential causes include bacterial, protozoal, or fungal infections, as well as a wide variety of toxic substances and drugs, and systemic immune-mediated pathological conditions. In spite of comprehensive investigation, the presence of inflammatory cardiomyopathy accompanied by left ventricular dysfunction, heart failure or arrhythmia is indicative of an unfavourable outcome. The reasons for the occurrence of either favourable outcomes, characterised by the absence of residual myocardial injury, or unfavourable outcomes, marked by the development of dilated cardiomyopathy, in patients afflicted by the condition remain to be elucidated. The relative contributions of pathogenic agents, genomic profiles of the host, and environmental factors in disease progression and resolution remain subjects of ongoing discourse. This includes the determination of which viruses function as active inducers and which merely play a bystander role. It remains unknown which changes in the host immune profile are critical in determining the outcome of myocarditis caused by various viruses, including coxsackievirus B3 (CVB3), adenoviruses, parvoviruses B19 and SARS-CoV-2. The objective of this review is unambiguous: to provide a concise summary and comprehensive assessment of the extant evidence on the pathogenesis, diagnosis and treatment of myocarditis and inflammatory cardiomyopathy. Its focus is exclusively on virus-induced and virus-associated myocarditis. In addition, the extant lacunae of knowledge in this field are identified and the extant experimental models are evaluated, with the aim of proposing future directions for the research domain. This includes differential gene expression that regulates iron and lipid and metabolic remodelling. Furthermore, the current state of knowledge regarding the cardiovascular implications of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is also discussed, along with the open questions that remain to be addressed.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France
| |
Collapse
|
40
|
Wang J, Liu Q, Xu C, Fu F, Shao Q, Fu Y, Wang Z, Ma J, Wang H, Yan Y, Sun J, Cheng Y. Conserved function of bat IRF7 in activating antiviral innate immunity: insights into the innate immune response in bats. Vet Res 2025; 56:59. [PMID: 40108733 PMCID: PMC11921751 DOI: 10.1186/s13567-025-01490-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 03/24/2024] [Indexed: 03/22/2025] Open
Abstract
Bats are natural hosts for various highly pathogenic viruses, which pose a considerable threat to humans and animals. However, they rarely display signs of disease infection from these viruses. The expression of IRF7-induced IFN-β plays a crucial role in preventing viral infections. However, the role of bat IRF7 during viral infection remains unclear. In this study, we cloned Tadarida brasiliensis IRF7 and discovered that its amino acid sequence was poorly conserved among species. Next, we investigated the expression of bat IRF7 mRNA in Tadarida brasiliensis lung cells (TB 1 Lu) infected with RNA viruses such as Newcastle disease virus (NDV), avian influenza virus (AIV), vesicular stomatitis virus (VSV), and the double-stranded RNA (dsRNA) analogue poly (I:C) and demonstrated that these viral infections significantly upregulated the mRNA expression of bat IRF7. Furthermore, the overexpression of IRF7 in TB1 Lu cells activated the expression of bat innate immune-related genes and inhibited virus replication. Importantly, we observed that bat IRF7 function is highly conserved in avian and mammalian species. Structurally, we revealed that the IRF domain of bat IRF7 is essential for activating IFN-β. In summary, our findings indicate that bat IRF7 has a conserved ability to activate bat antiviral innate immunity. This study provides a theoretical foundation for further understanding the innate immune response in bats.
Collapse
Affiliation(s)
- Jie Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Qiuju Liu
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Caixia Xu
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Feiyu Fu
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Shao
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Yapeng Fu
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaofei Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Jingjiao Ma
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Hengan Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Yaxian Yan
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Jianhe Sun
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China.
| | - Yuqiang Cheng
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
41
|
Li K, Jadhav P, Wen Y, Tan H, Wang J. Development of a Fluorescence Polarization Assay for the SARS-CoV-2 Papain-like Protease. ACS Pharmacol Transl Sci 2025; 8:774-784. [PMID: 40109744 PMCID: PMC11915184 DOI: 10.1021/acsptsci.4c00642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 03/22/2025]
Abstract
The COVID-19 pandemic has caused significant losses to the global community. Although effective vaccination and antiviral therapeutics provide primary defense, SARS-CoV-2 remains a public health threat, given the emerging resistant variants. The SARS-CoV-2 papain-like protease (PLpro) is essential for viral replication and is a promising drug target. We recently designed a series of biarylphenyl PLpro inhibitors with a representative lead Jun12682 showing potent antiviral efficacy in a SARS-CoV-2 infection mouse model. In this study, we designed a fluorescein-labeled biarylphenyl probe Jun12781 and used it to optimize a fluorescence polarization (FP) assay. The FP assay is suitable for high-throughput screening with a Z' factor of 0.69. In addition, we found a positive correlation between the FP binding affinity and the enzymatic inhibitory potency of PLpro inhibitors, suggesting that the FP assay is valid in characterizing the binding affinity of PLpro inhibitors.
Collapse
Affiliation(s)
- Kan Li
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Prakash Jadhav
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Yu Wen
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Haozhou Tan
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Jun Wang
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
42
|
Liu C, Park YJ, Ma CB, Stuart C, Gen R, Sun YC, Yang X, Lin MY, Xiong Q, Si JY, Liu P, Veesler D, Yan H. ACE2 utilization of HKU25 clade MERS-related coronaviruses with broad geographic distribution. RESEARCH SQUARE 2025:rs.3.rs-6097445. [PMID: 40162213 PMCID: PMC11952669 DOI: 10.21203/rs.3.rs-6097445/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Dipeptidyl peptidase-4 (DPP4) is a well-established receptor for several MERS-related coronaviruses (MERSr-CoVs) isolated from humans, camels, pangolins, and bats 1-6. However, the receptor usage of many genetically diverse bat MERSr-CoVs with broad geographical distributions remains poorly understood. Recent studies have identified angiotensin-converting enzyme 2 (ACE2) as an entry receptor for multiple merbecovirus clades. Here, using viral antigen and pseudovirus-based functional assays, we demonstrate that several bat merbecoviruses from the HKU25 clade previously thought to utilize DPP4 7, employ ACE2 as their functional receptor. Cryo-electron microscopy analysis revealed that HsItaly2011 and VsCoV-a7 recognize ACE2 with a binding mode sharing similarity with that of HKU5 but involving remodeled interfaces and distinct ortholog selectivity, suggesting a common evolutionary origin of ACE2 utilization for these two clades of viruses. EjCoV-3, a strain closely related to the DPP4-using MERSr-CoV BtCoV-422, exhibited relatively broad ACE2 ortholog tropism and could utilize human ACE2 albeit suboptimally. Despite differences in entry mechanisms and spike proteolytic activation compared to MERS-CoV, these viruses remain sensitive to several broadly neutralizing antibodies and entry inhibitors. These findings redefine our understanding of the evolution of receptor usage among MERSr-CoVs and highlight the versatility of ACE2 as a functional receptor for diverse coronaviruses.
Collapse
Affiliation(s)
- Chen Liu
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Young-Jun Park
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington; Seattle, WA 98195, USA
| | - Cheng-Bao Ma
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Cameron Stuart
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Risako Gen
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
| | - Yu-Cheng Sun
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Xiao Yang
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Mei-Yi Lin
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Qing Xiong
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Jun-Yu Si
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - Peng Liu
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| | - David Veesler
- Department of Biochemistry, University of Washington; Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington; Seattle, WA 98195, USA
| | - Huan Yan
- State Key Laboratory of Virology and Biosafety, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University; Wuhan, Hubei, 430072, China
| |
Collapse
|
43
|
Barrera Martínez Y, Boillat Oriani GA, Vega Montes P, Martínez Moreno E, Pérez Pérez A, Casajuana Pérez RJ, Muñoz Cobos F. [Long-term persistence of post-COVID-19 symptoms: A two-year follow-up of a Primary Care cohort]. An Sist Sanit Navar 2025; 48:e1101. [PMID: 40079454 PMCID: PMC11927030 DOI: 10.23938/assn.1101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/09/2024] [Accepted: 11/22/2024] [Indexed: 03/15/2025]
Abstract
BACKGROUND This study aims to determine the prevalence of long COVID two years after initial infection, identify prognostic factors, and assess its impact on quality of life. METHODOLOGY An ambispective cohort study was conducted with patients aged = 18 years from two health centers in Málaga, Spain, who tested positive for SARS-CoV-2 between October 2020 and May 2021. Systematic random sampling was performed in October 2022, with 5% precision, 5% alpha error, and 25% expected losses. The cohort was followed until May 2023. Dependent variables included long COVID (= 1 symptom lasting =8 weeks), symptom count, quality of life (EuroQol 5-D), and global health perception (EQ-EVA). Independent variables included age, sex, severity of initial infection severity, vaccination status, comorbidities, and reinfection. RESULTS Of 914 eligible individuals, 173 patients were sampled (mean age 47 years, 58.4% female). Long COVID was observed in 32.36% of participants, with 23% affected two years after infection. Fatigue and anosmia/dysgeusia were the most frequent symptoms. Prognostic factors for long COVID included higher initial infection severity, younger age, and reinfection. For symptom count, factors were depression, initial infection severity, and reinfection. Health perception was eight points lower in long COVID patients (77.72: SD=17.10 vs 86.15; SD=16.25; p<0.001). Female sex, older age, fewer comorbidities, and more vaccine doses were associated with better quality of life. CONCLUSIONS Thirty-two percent of patients experienced long COVID, mainly fatigue and anosmia/dysgeusia. It associates with higher initial severity, younger age, and reinfection, negatively impacting quality of life.
Collapse
Affiliation(s)
| | - Gerardo Andrés Boillat Oriani
- Servicio Andaluz de Salud. Atención Primaria de Salud. Centro de Salud Rincón de la Victoria. Rincón de la Victoria. Málaga. España.
| | - Pedro Vega Montes
- Servicio Andaluz de Salud. Atención Primaria de Salud. Centro de Salud El Palo. Málaga. España.
| | - Elena Martínez Moreno
- Servicio Andaluz de Salud. Atención Primaria de Salud. Centro de Salud El Palo. Málaga. España.
| | - Alejandro Pérez Pérez
- Servicio Andaluz de Salud. Atención Primaria de Salud. Centro de Salud El Palo. Málaga. España.
| | | | - Francisca Muñoz Cobos
- Servicio Andaluz de Salud. Atención Primaria de Salud. Centro de Salud El Palo. Málaga. España.
| |
Collapse
|
44
|
Borrego-Ruiz A, Borrego JJ. Involvement of virus infections and antiviral agents in schizophrenia. Psychol Med 2025; 55:e73. [PMID: 40059820 PMCID: PMC12055031 DOI: 10.1017/s0033291725000467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Schizophrenia is a chronic and complex mental disorder resulting from interactions between cumulative and synergistic genetic and environmental factors. Viral infection during the prenatal stage constitutes one of the most relevant risk factors for the development of schizophrenia later in adulthood. METHODS A narrative review was conducted to explore the link between viral infections and schizophrenia, as well as the neuropsychiatric effects of antiviral drugs, particularly in the context of this specific mental condition. Literature searches were performed using the PubMed, Scopus, and Web of Science databases. RESULTS Several viral infections, such as herpesviruses, influenza virus, Borna disease virus, and coronaviruses, can directly or indirectly disrupt normal fetal brain development by modifying gene expression in the maternal immune system, thereby contributing to the pathophysiological symptoms of schizophrenia. In addition, neuropsychiatric effects caused by antiviral drugs are frequent and represent significant adverse outcomes for viral treatment. CONCLUSIONS Epidemiological evidence suggests a potential relationship between viruses and schizophrenia. Increases in inflammatory cytokine levels and changes in the expression of key genes observed in several viral infections may constitute potential links between these viral infections and schizophrenia. Furthermore, antivirals may affect the central nervous system, although for most drugs, their mechanisms of action are still unclear, and a strong relationship between antivirals and schizophrenia has not yet been established.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Juan J. Borrego
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Plataforma BIONAND, Málaga, Spain
| |
Collapse
|
45
|
Chen J, Wang J, Zhao H, Tan X, Yan S, Zhang H, Wang T, Tang X. Molecular breeding of pigs in the genome editing era. Genet Sel Evol 2025; 57:12. [PMID: 40065264 PMCID: PMC11892312 DOI: 10.1186/s12711-025-00961-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND To address the increasing demand for high-quality pork protein, it is essential to implement strategies that enhance diets and produce pigs with excellent production traits. Selective breeding and crossbreeding are the primary methods used for genetic improvement in modern agriculture. However, these methods face challenges due to long breeding cycles and the necessity for beneficial genetic variation associated with high-quality traits within the population. This limitation restricts the transfer of desirable alleles across different genera and species. This article systematically reviews past and current research advancements in porcine molecular breeding. It discusses the screening of clustered regularly interspaced short palindromic repeats (CRISPR) to identify resistance loci in swine and the challenges and future applications of genetically modified pigs. MAIN BODY The emergence of transgenic and gene editing technologies has prompted researchers to apply these methods to pig breeding. These advancements allow for alterations in the pig genome through various techniques, ranging from random integration into the genome to site-specific insertion and from target gene knockout (KO) to precise base and prime editing. As a result, numerous desirable traits, such as disease resistance, high meat yield, improved feed efficiency, reduced fat deposition, and lower environmental waste, can be achieved easily and effectively by genetic modification. These traits can serve as valuable resources to enhance swine breeding programmes. CONCLUSION In the era of genome editing, molecular breeding of pigs is critical to the future of agriculture. Long-term and multidomain analyses of genetically modified pigs by researchers, related policy development by regulatory agencies, and public awareness and acceptance of their safety are the keys to realizing the transition of genetically modified products from the laboratory to the market.
Collapse
Affiliation(s)
- Jiahuan Chen
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Jiaqi Wang
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Haoran Zhao
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Xiao Tan
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Shihan Yan
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Huanyu Zhang
- College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Tiefeng Wang
- College of Life Science, Baicheng Normal University, Baicheng, 137000, China.
| | - Xiaochun Tang
- College of Animal Sciences, Jilin University, Changchun, 130062, China.
| |
Collapse
|
46
|
Anderson LN, Hoyt CT, Zucker JD, McNaughton AD, Teuton JR, Karis K, Arokium-Christian NN, Warley JT, Stromberg ZR, Gyori BM, Kumar N. Computational tools and data integration to accelerate vaccine development: challenges, opportunities, and future directions. Front Immunol 2025; 16:1502484. [PMID: 40124369 PMCID: PMC11925797 DOI: 10.3389/fimmu.2025.1502484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/23/2025] [Indexed: 03/25/2025] Open
Abstract
The development of effective vaccines is crucial for combating current and emerging pathogens. Despite significant advances in the field of vaccine development there remain numerous challenges including the lack of standardized data reporting and curation practices, making it difficult to determine correlates of protection from experimental and clinical studies. Significant gaps in data and knowledge integration can hinder vaccine development which relies on a comprehensive understanding of the interplay between pathogens and the host immune system. In this review, we explore the current landscape of vaccine development, highlighting the computational challenges, limitations, and opportunities associated with integrating diverse data types for leveraging artificial intelligence (AI) and machine learning (ML) techniques in vaccine design. We discuss the role of natural language processing, semantic integration, and causal inference in extracting valuable insights from published literature and unstructured data sources, as well as the computational modeling of immune responses. Furthermore, we highlight specific challenges associated with uncertainty quantification in vaccine development and emphasize the importance of establishing standardized data formats and ontologies to facilitate the integration and analysis of heterogeneous data. Through data harmonization and integration, the development of safe and effective vaccines can be accelerated to improve public health outcomes. Looking to the future, we highlight the need for collaborative efforts among researchers, data scientists, and public health experts to realize the full potential of AI-assisted vaccine design and streamline the vaccine development process.
Collapse
Affiliation(s)
| | - Charles Tapley Hoyt
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, United States
| | - Jeremy D. Zucker
- Pacific Northwest National Laboratory (DOE), Richland, WA, United States
| | | | - Jeremy R. Teuton
- Pacific Northwest National Laboratory (DOE), Richland, WA, United States
| | - Klas Karis
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, United States
| | | | - Jackson T. Warley
- Pacific Northwest National Laboratory (DOE), Richland, WA, United States
| | | | - Benjamin M. Gyori
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, United States
- Department of Bioengineering, College of Engineering, Northeastern University, Boston, MA, United States
| | - Neeraj Kumar
- Pacific Northwest National Laboratory (DOE), Richland, WA, United States
| |
Collapse
|
47
|
Xu Y, Han Y, Xu P, Zhou S, Zhao P, Wang Y, Hu J, Ma M, Li Z, Bo S, Zhao C, Ji L, Yuan Y, Zhao W, Wang J, Jin Q, He G, Wu Z. Avian Migration-Mediated Transmission and Recombination Driving the Diversity of Gammacoronaviruses and Deltacoronaviruses. Mol Biol Evol 2025; 42:msaf045. [PMID: 39963938 PMCID: PMC11886833 DOI: 10.1093/molbev/msaf045] [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: 11/16/2024] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 03/08/2025] Open
Abstract
In the wake of pandemics like COVID-19, which have zoonotic origins, the role of wildlife as reservoirs for emerging infectious diseases has garnered heightened attention. Migratory birds, traversing continents, represent a potent but under-researched vector for the spread of infectious diseases, including novel coronaviruses. This study delves into the genetic diversity and transmission dynamics of coronaviruses in migratory birds, presenting pivotal findings. From April 2019 to April 2023, we screened 5,263 migratory bird samples collected from Shanghai, China, identifying 372 coronavirus-positive samples belonging to five avian-related coronavirus subgenera and subsequently obtaining 120 complete genome sequences. To facilitate further research with a global perspective, the study curated all available 19,000 avian-associated coronaviruses and expanded the original 12 species to 16, including three novel coronavirus species identified in our study and one re-classified species from the public domain. The study illuminates the intricate genetic evolution and transmission dynamics of birds-related coronaviruses on a global scale. A notable aspect of our research is the identification of complex recombination patterns within the spike protein across different virus species and subgenera, highlighting migratory birds as a reservoir of coronavirus. Notably, the coronaviruses found in migratory birds, predominantly from the orders Anseriformes, Charadriiformes, and Pelecaniformes, with domestic ducks from Anseriformes playing a key role in bridging the transmission of coronaviruses between migratory and non-migratory birds. These findings reveal the genetic and recombination characteristics of coronaviruses in migratory birds, emphasizing the critical role of ecologically pivotal bird species in coronavirus transmission and genetic diversity shaping.
Collapse
Affiliation(s)
- Yuting Xu
- School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Yelin Han
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Panpan Xu
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Siyu Zhou
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Peng Zhao
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Yuyang Wang
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Jie Hu
- School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Min Ma
- School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Zirong Li
- Shanghai Forestry Station, Shanghai, PR China
| | - Shunqi Bo
- Shanghai Landscaping & City Appearance Administrative Bureau, Shanghai, PR China
| | | | - Lei Ji
- Shanghai Forestry Station, Shanghai, PR China
| | - Yue Yuan
- Shanghai Chongming Dongtan Nature Reserve Administration Center, Shanghai, PR China
| | - Wenliang Zhao
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Jianwei Wang
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Qi Jin
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Guimei He
- School of Life Sciences, East China Normal University, Shanghai, PR China
- Institute of Eco-Chongming (IEC), East China Normal University, Shanghai, PR China
- Shanghai Institute of Wildlife Epidemics, East China Normal University, Shanghai, PR China
| | - Zhiqiang Wu
- NHC Key Laboratory of Systems Biology of Pathogens, State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| |
Collapse
|
48
|
Dolgova NV, Qureshi M, Latimer M, Grishin A, Cygler M, Vogt LI, Cotelesage JJH, Sokaras D, Kroll T, Pickering IJ, George GN. Structural Changes at the Zinc Active Site of ACE2 on Binding the SARS-CoV-2 Spike Protein Receptor Binding Domain. Inorg Chem 2025; 64:3831-3841. [PMID: 39962897 DOI: 10.1021/acs.inorgchem.4c04974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
The causative agent of Covid-19 is the SARS-CoV-2 virus. Initiation of cell entry by SARS-CoV-2 is critically dependent upon binding of the SARS-CoV-2 spike protein to angiotensin-converting enzyme 2 (ACE2, EC 3.4.17.23). The mechanism of binding of the SARS-CoV-2 spike receptor binding domain to ACE2 is among the most intensively studied infection mechanisms of any pathogen, including a very large number of structural studies. ACE2 is a membrane-associated zinc carboxypeptidase, comprising three domains, the protease domain, a neck domain, and a membrane-spanning α-helical domain. In addition to its role as a carboxypeptidase, ACE2 is also a chaperone for a Na+-amino acid cotransporter called B0AT1, and in the presence of B0AT1, full-length ACE2 forms dimers. Most studies to date related to Covid-19 have employed just the ACE2 protease domain and have neglected any possible roles of the Zn2+-containing ACE2 active site. We show here that ACE2, including the neck domain in addition to the protease domain (and in the absence of B0AT1), is dimeric and shows distinctive allostery in its catalytic activity. In contrast, the intensively studied protease domain is monomeric and shows no allostery. Binding of the spike receptor binding domain (RBD) to dimeric ACE2 eliminates its allostery. X-ray absorption spectroscopy of Zn2+ ACE2 shows distinctive changes in the active site structure upon binding of spike RBD but only in the dimeric form. Taken together, our results indicate that the Zn2+-containing active site exhibits a notable level of flexibility and that the dimeric form of ACE2, including both protease and neck domains, likely presents a superior model for the study of ACE2-spike interactions than the monomeric ACE2.
Collapse
Affiliation(s)
- Natalia V Dolgova
- Calibr─California Institute for Biomedical Research, Scripps Research, 11119 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Muhammad Qureshi
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Matthew Latimer
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Andrey Grishin
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Miroslaw Cygler
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Linda I Vogt
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Julien J H Cotelesage
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - Dimosthenis Sokaras
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Thomas Kroll
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Ingrid J Pickering
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Toxicology Centre, University of Saskatchewan, 44 Campus Drive, Saskatoon, Saskatchewan S7N 5B3, Canada
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Graham N George
- Department of Geological Sciences, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Toxicology Centre, University of Saskatchewan, 44 Campus Drive, Saskatoon, Saskatchewan S7N 5B3, Canada
- Department of Chemistry, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5C9, Canada
| |
Collapse
|
49
|
Frigori RB. Evolution and pathogenicity of SARS-CoVs: A microcanonical analysis of receptor-binding motifs. Phys Rev E 2025; 111:034401. [PMID: 40247563 DOI: 10.1103/physreve.111.034401] [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: 09/10/2024] [Accepted: 02/07/2025] [Indexed: 04/19/2025]
Abstract
The rapid evolution and global impact of coronaviruses, notably SARS-CoV-1 and SARS-CoV-2, underscore the importance of understanding their molecular mechanisms in detail. This study focuses on the receptor-binding motif (RBM) within the spike protein of these viruses, a critical element for viral entry through interaction with the ACE2 receptor. We investigate the sequence variations in the RBM across SARS-CoV-1, SARS-CoV-2, and its early variants of concern (VOCs). Utilizing multicanonical simulations and microcanonical analysis, we examine how these variations influence the folding dynamics, thermostability, and solubility of the RBMs. Our methodology includes calculating the density of states (DoS) to identify structural phase transitions and assess thermodynamic properties. Furthermore, we solve the Poisson-Boltzmann equation to model the solubility of the RBMs in aqueous environments. This methodology is expected to elucidate structural and functional differences in viral evolution and pathogenicity, likely improving targeted treatments and vaccines.
Collapse
Affiliation(s)
- Rafael B Frigori
- Universidade Tecnológica Federal do Paraná, Rua Cristo Rei 19, CEP 85902-490, Toledo (PR), Brazil
| |
Collapse
|
50
|
Jiang Y, Xu L, Zheng X, Shi H. Recent advances in nutritional metabolism studies on SARS-CoV-2 infection. INFECTIOUS MEDICINE 2025; 4:100162. [PMID: 39936106 PMCID: PMC11810712 DOI: 10.1016/j.imj.2025.100162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 02/13/2025]
Abstract
In the context of the coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), metabolic research has become crucial for in-depth exploration of viral infection mechanisms and in searching for therapeutic strategies. This paper summarizes the interrelationships between carbohydrate, lipid, and amino acid metabolism and COVID-19 infection, discussing their roles in infection progression. SARS-CoV-2 infection leads to insulin resistance and increased glycolysis, reducing glucose utilization and shifting metabolism to use fat as an energy source. Fat is crucial for viral replication, and imbalances in amino acid metabolism may interfere with immune regulation. Consequently, metabolic changes such as hyperglycemia, hypolipidemia, and deficiency of certain amino acids following SARS-CoV-2 infection can contribute to progression toward severe conditions. These metabolic pathways not only have potential value in prediction and diagnosis but also provide new perspectives for the development of therapeutic strategies. By monitoring metabolic changes, infection severity can be predicted early, and modulating these metabolic pathways may help reduce inflammatory responses, improve immune responses, and reduce the risk of thrombosis. Research on the relationship between metabolism and SARS-CoV-2 infection provides an important scientific basis for addressing the global challenge posed by COVID-19, however, further studies are needed to validate these findings and provide more effective strategies for disease control.
Collapse
Affiliation(s)
- Yufen Jiang
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Linle Xu
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Xuexing Zheng
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Hongbo Shi
- Beijing Municipal Key Laboratory of Liver Failure and Artificial Liver Treatment Research, Fourth Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| |
Collapse
|