1
|
Bayram H, Konyalilar N, Elci MA, Rajabi H, Aksoy GT, Mortazavi D, Kayalar Ö, Dikensoy Ö, Taborda-Barata L, Viegi G. Issue 4 - Impact of air pollution on COVID-19 mortality and morbidity: An epidemiological and mechanistic review. Pulmonology 2025; 31:2416829. [PMID: 38755091 DOI: 10.1016/j.pulmoe.2024.04.005] [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: 03/28/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 05/18/2024] Open
Abstract
Air pollution is a major global environment and health concern. Recent studies have suggested an association between air pollution and COVID-19 mortality and morbidity. In this context, a close association between increased levels of air pollutants such as particulate matter ≤2.5 to 10 µM, ozone and nitrogen dioxide and SARS-CoV-2 infection, hospital admissions and mortality due to COVID 19 has been reported. Air pollutants can make individuals more susceptible to SARS-CoV-2 infection by inducing the expression of proteins such as angiotensin converting enzyme (ACE)2 and transmembrane protease, serine 2 (TMPRSS2) that are required for viral entry into the host cell, while causing impairment in the host defence system by damaging the epithelial barrier, muco-ciliary clearance, inhibiting the antiviral response and causing immune dysregulation. The aim of this review is to report the epidemiological evidence on impact of air pollutants on COVID 19 in an up-to-date manner, as well as to provide insights on in vivo and in vitro mechanisms.
Collapse
Affiliation(s)
- Hasan Bayram
- Koç University Research Centre for Translational Medicine (KUTTAM), Zeytinburnu, Istanbul, Turkey
- Department of Pulmonary Medicine, School of Medicine, Koç University, Zeytinburnu, Istanbul, Turkey
| | - Nur Konyalilar
- Koç University Research Centre for Translational Medicine (KUTTAM), Zeytinburnu, Istanbul, Turkey
| | | | - Hadi Rajabi
- Koç University Research Centre for Translational Medicine (KUTTAM), Zeytinburnu, Istanbul, Turkey
| | - G Tuşe Aksoy
- Koç University Research Centre for Translational Medicine (KUTTAM), Zeytinburnu, Istanbul, Turkey
| | - Deniz Mortazavi
- Koç University Research Centre for Translational Medicine (KUTTAM), Zeytinburnu, Istanbul, Turkey
| | - Özgecan Kayalar
- Koç University Research Centre for Translational Medicine (KUTTAM), Zeytinburnu, Istanbul, Turkey
| | - Öner Dikensoy
- Department of Pulmonary Medicine, School of Medicine, Koç University, Zeytinburnu, Istanbul, Turkey
| | - Luis Taborda-Barata
- UBIAir - Clinical and Experimental Lung Centre UBIMedical, University of Beira Interior, Covilhã, Portugal
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | | |
Collapse
|
2
|
Barua S, Iduu NV, Murillo DFB, Tarannum A, Dimino H, Barua S, Shu Y, Johnson C, Miller MR, Chenoweth K, Christopherson P, Huber L, Wood T, Turner K, Wang C. Nationwide seroprevalence of SARS-CoV-2 Delta variant and five Omicron sublineages in companion cats and dogs in the USA: insights into their role in COVID-19 epidemiology. Emerg Microbes Infect 2025; 14:2437246. [PMID: 39635731 PMCID: PMC11636146 DOI: 10.1080/22221751.2024.2437246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Understanding SARS-CoV-2 epidemiology in companion animals is critical for evaluating their role in viral transmission and their potential as sentinels for human infections. This large-scale serosurvey analyzed serum samples from 706 cats and 2,396 dogs collected across the USA in 2023 using a surrogate virus neutralization test (sVNT) to detect SARS-CoV-2 antibodies. Overall, 5.7% of cats and 4.7% of dogs tested positive for antibodies, with younger animals (under 12 months) showing significantly lower seropositivity rates (p = 0.0048). Additionally, we analyzed 153 positive samples for variant-specific antibody responses using six sVNT kits targeting the Delta variant and five Omicron sublineages. Among cats, 67.5% showed antibodies to Delta, with positivity rates for Omicron sublineages as follows: BA.1 (62.5%), BA.2 (42.5%), BA.4/BA.5 (77.5%), XBB (52.5%), and XBB.1.5 (45.0%). In dogs, 55.8% were positive for Delta, and Omicron sublineage rates were BA.1 (46.0%), BA.4/BA.5 (37.2%), XBB (58.4%), BA.2 (13.3%), and XBB.1.5 (9.7%). Given the close contact between companion animals and humans, and the persistence of antibodies against various SARS-CoV-2 variants and sublineages, our findings suggest that seroprevalence in cats and dogs may serve as valuable tool for tracking COVID-19 epidemiology.
Collapse
Affiliation(s)
- Subarna Barua
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Nneka Vivian Iduu
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | | | - Asfiha Tarannum
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Hill Dimino
- College of Sciences and Mathematics, Auburn University, Auburn, AL, USA
| | - Suchita Barua
- College of Sciences and Mathematics, Auburn University, Auburn, AL, USA
| | - Yue Shu
- College of Sciences and Mathematics, Auburn University, Auburn, AL, USA
| | - Calvin Johnson
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Megan R. Miller
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Laurel, MD, USA
| | - Kelly Chenoweth
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Peter Christopherson
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Laura Huber
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Theresa Wood
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Kelley Turner
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Chengming Wang
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| |
Collapse
|
3
|
Kim MC, Jang SS, Van Lo T, Noh JY, Lim HA, Kim HY, Mun DY, Kim K, Lee TW, Choi YG, Yoon SW, Jeong DG, Kim SS, Kim HK. Circulation characteristics of bat coronaviruses linked to bat ecological factors in Korea, 2021-2022. Virulence 2025; 16:2502551. [PMID: 40336345 PMCID: PMC12077446 DOI: 10.1080/21505594.2025.2502551] [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: 08/12/2024] [Revised: 11/25/2024] [Accepted: 01/26/2025] [Indexed: 05/09/2025] Open
Abstract
Considering that bat ecology alterations may be linked with pathogen spillover, research on bat coronaviruses, particularly on the infection and transmission pattern among bats in relation with their ecology, is essential. We captured bats distributed in Korea from 2021 to 2022, examined coronaviruses in oral swabs, feces, urine, and ectoparasites, and were able to detect alphacoronavirus. We investigated coronaviruses, but noted no substantial differences in the body condition index in the coronavirus-positive bats. Binary logistic regression analysis revealed that bat ecological factors that were significantly associated with coronavirus-positive were roost type, sample type, and bat species. Coronavirus-positive ectoparasite cases suggested additional study on the potential role of them as the viral transmission vectors or fomites. Reinfection of a different coronavirus in recaptured bats was evident, suggesting the possibility that coronavirus circulation can evade the potential protective immunity acquired from previous coronavirus infections. The present findings provide comprehensive information on the coronaviruses transmission dynamics within bat populations linked with bat ecology.
Collapse
Affiliation(s)
- Min Chan Kim
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Seong Sik Jang
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Thi Van Lo
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Ji Yeong Noh
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Hyun A. Lim
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Ha Yeon Kim
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Da Young Mun
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| | - Kihyun Kim
- Ecological Technology Research Team, Division of Ecological Application, National Institute of Ecology, Seocheon, Republic of Korea
| | - Taek-Woo Lee
- Ecological Technology Research Team, Division of Ecological Application, National Institute of Ecology, Seocheon, Republic of Korea
| | - Yong Gun Choi
- The Korean Institute of Biospeleology, Daejeon, Korea
| | - Sun-Woo Yoon
- Department of Biological Sciences and Biotechnology, Andong National University, Andong, Republic of Korea
| | - Dae Gwin Jeong
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
- Bio-Analytical Science Division, Korea University of Science and Technology (UST), Daejeon, Korea
| | - Sun-Sook Kim
- Ecological Technology Research Team, Division of Ecological Application, National Institute of Ecology, Seocheon, Republic of Korea
| | - Hye Kwon Kim
- Department of Biological Sciences and Biotechnology, College of Natural Sciences, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
4
|
Alizon S, Sofonea MT. SARS-CoV-2 epidemiology, kinetics, and evolution: A narrative review. Virulence 2025; 16:2480633. [PMID: 40197159 PMCID: PMC11988222 DOI: 10.1080/21505594.2025.2480633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 11/26/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Since winter 2019, SARS-CoV-2 has emerged, spread, and evolved all around the globe. We explore 4 y of evolutionary epidemiology of this virus, ranging from the applied public health challenges to the more conceptual evolutionary biology perspectives. Through this review, we first present the spread and lethality of the infections it causes, starting from its emergence in Wuhan (China) from the initial epidemics all around the world, compare the virus to other betacoronaviruses, focus on its airborne transmission, compare containment strategies ("zero-COVID" vs. "herd immunity"), explain its phylogeographical tracking, underline the importance of natural selection on the epidemics, mention its within-host population dynamics. Finally, we discuss how the pandemic has transformed (or should transform) the surveillance and prevention of viral respiratory infections and identify perspectives for the research on epidemiology of COVID-19.
Collapse
Affiliation(s)
- Samuel Alizon
- CIRB, CNRS, INSERM, Collège de France, Université PSL, Paris, France
| | - Mircea T. Sofonea
- PCCEI, University Montpellier, INSERM, Montpellier, France
- Department of Anesthesiology, Critical Care, Intensive Care, Pain and Emergency Medicine, CHU Nîmes, Nîmes, France
| |
Collapse
|
5
|
Zhou S, Hui X, Wang W, Zhao C, Jin M, Qin Y, Chen M. SARS-CoV-2 and HCoV-OC43 regulate host m6A modification via activation of the mTORC1 signalling pathway to facilitate viral replication. Emerg Microbes Infect 2025; 14:2447620. [PMID: 39745173 PMCID: PMC11852242 DOI: 10.1080/22221751.2024.2447620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/08/2024] [Accepted: 12/22/2024] [Indexed: 02/25/2025]
Abstract
N6-methyladenosine (m6A) is the most prevalent post-transcriptional modification in eukaryotic RNA and is also present in various viral RNAs, where it plays a crucial role in regulating the viral life cycle. However, the molecular mechanisms through which viruses regulate host RNA m6A methylation are not fully understood. In this study, we reveal that SARS-CoV-2 and HCoV-OC43 infection enhance host m6A modification by activating the mTORC1 signalling pathway. Specifically, the viral non-structural protein nsp14 upregulates the expression of S-adenosylmethionine synthase MAT2A in an mTORC1-dependent manner. This mTORC1-MAT2A axis subsequently stimulates the synthesis of S-adenosylmethionine (SAM). The increase of SAM then enhances the m6A methylation of host RNA and facilitates viral replication. Our findings uncover a molecular mechanism by which viruses regulate host m6A methylation and provide insights into how SARS-CoV-2 hijacks host cellular epitranscriptomic modifications to promote its replication.
Collapse
Affiliation(s)
- Shixiong Zhou
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
| | - Xianfeng Hui
- National key laboratory of agricultural microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Weiwei Wang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
| | - Chunbei Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
| | - Meilin Jin
- National key laboratory of agricultural microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
| | - Yali Qin
- School of Life Sciences, Hubei University, Wuhan, People’s Republic of China
| | - Mingzhou Chen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, People’s Republic of China
- School of Life Sciences, Hubei University, Wuhan, People’s Republic of China
| |
Collapse
|
6
|
Liu M, Zhao L, Huang X, Tang Z, Zhong Y, Yan M, Liu S, Wang S, Sun Z, Rao Z, Yu L, Fang Y, Zhang W, Zhang H, Peng W. Identification of broad-spectrum M pro inhibitors: a focus on high-risk coronaviruses and conserved interactions. J Enzyme Inhib Med Chem 2025; 40:2503961. [PMID: 40396609 PMCID: PMC12096674 DOI: 10.1080/14756366.2025.2503961] [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: 03/19/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025] Open
Abstract
The COVID-19 pandemic underscores the urgent need to develop broad-spectrum antivirals against coronaviruses (CoVs) to prepare for future outbreaks. In this study, we presented a systematic approach to developing broad-spectrum Mpro inhibitors, with a focus on high-risk CoVs. We optimised S-217622 as a lead compound, with the goal of enhancing conserved interactions within the S1, S2, and S3/S4 pockets of Mpro, leading to significantly improved inhibitory potency against representative CoVs. Compound 25 exhibited submicromolar activity across all ten CoVs, with IC50 values below 0.1 μM for six of them. The X-ray co-crystal structure of SARS-CoV-2 Mpro in complex with compound 25 revealed the structural basis of conserved interactions contributing to its broad-spectrum activity. This study demonstrates the feasibility of reinforcing conserved interactions to develop Mpro inhibitors with broad-spectrum activity and provides valuable strategies for combating future pandemics caused by unknown CoVs.
Collapse
Affiliation(s)
- Man Liu
- Department of Infectious Diseases, The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital, Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, China
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
| | - Li Zhao
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
- Guangzhou Medical University, Guangzhou, China
| | - Xupeng Huang
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
| | - Zhenhao Tang
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
| | - Yihang Zhong
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
| | - Mengrong Yan
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
| | - Shun Liu
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
- University of South China, Hengyang, China
| | - Shunjing Wang
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
- University of South China, Hengyang, China
| | - Zeyun Sun
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
- Guangzhou Medical University, Guangzhou, China
| | - Zixuan Rao
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
- University of South China, Hengyang, China
| | - Linyi Yu
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
- University of South China, Hengyang, China
| | - Yuying Fang
- Department of Infectious Diseases, The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital, Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, China
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
| | - Wei Zhang
- Department of Infectious Diseases, The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital, Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, China
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
- University of South China, Hengyang, China
| | - Hongbo Zhang
- Beijing StoneWise Technology Co. Ltd, Beijing, China
| | - Wei Peng
- Department of Infectious Diseases, The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital, Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, China
- Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, China, China
- Guangzhou Medical University, Guangzhou, China
- University of South China, Hengyang, China
- Lead contact
| |
Collapse
|
7
|
Chen SC, Xu CT, Chang CF, Yang CS, Lin PH, Liu WM, Chen Y, Yu CH. Characterization of the binding features between SARS-CoV-2 5'-proximal transcripts of genomic RNA and nucleocapsid proteins. RNA Biol 2025; 22:1-16. [PMID: 40077853 PMCID: PMC11913385 DOI: 10.1080/15476286.2025.2471643] [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] [Revised: 02/01/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Packaging signals (PSs) of coronaviruses (CoVs) are specific RNA elements recognized by nucleocapsid (N) proteins that direct the selective packaging of genomic RNAs (gRNAs). These signals have been identified in the coding regions of the nonstructural protein 15 (Nsp 15) in CoVs classified under Embecovirus, a subgenus of betacoronaviruses (beta-CoVs). The PSs in other alpha- and beta-CoVs have been proposed to reside in the 5'-proximal regions of gRNAs, supported by comprehensive phylogenetic evidence. However, experimental data remain limited. In this study, we investigated the interactions between Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) 5'-proximal gRNA transcripts and N proteins using electrophoretic mobility shift assays (EMSAs). Our findings revealed that the in vitro synthesized 5'-proximal gRNA transcripts of CoVs can shift from a major conformation to alternative conformations. We also observed that the conformer comprising multiple stem-loops (SLs) is preferentially bound by N proteins. Deletions of the 5'-proximal structural elements of CoV gRNA transcripts, SL1 and SL5a/b/c in particular, were found to promote the formation of alternative conformations. Furthermore, we identified RNA-binding peptides from a pool derived from SARS-CoV N protein. These RNA-interacting peptides were shown to preferentially bind to wild-type SL5a RNA. In addition, our observations of N protein condensate formation in vitro demonstrated that liquid-liquid phase separation (LLPS) of N proteins with CoV-5'-UTR transcripts was influenced by the presence of SL5a/b/c. In conclusion, these results collectively reveal previously uncharacterized binding features between the 5'-proximal transcripts of CoV gRNAs and N proteins.
Collapse
Affiliation(s)
- Shih-Cheng Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan City, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Cui-Ting Xu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Chuan-Fu Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Chia-Shin Yang
- Institute of Translational Medicine and New Drug Development, College of Medicine, China Medical University, Taichung City, Taiwan
| | - Pin-Han Lin
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wei-Min Liu
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yeh Chen
- Institute of Translational Medicine and New Drug Development, College of Medicine, China Medical University, Taichung City, Taiwan
| | - Chien-Hung Yu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| |
Collapse
|
8
|
Hu S, Zhong Q, Xie X, Zhang S, Wang J, Liu H, Dai W. Research progress on critical viral protease inhibitors for coronaviruses and enteroviruses. Bioorg Med Chem Lett 2025; 122:130168. [PMID: 40074013 DOI: 10.1016/j.bmcl.2025.130168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
Viral infectious diseases have been seriously affecting human life and health. SARS-CoV-2 was the pathogen that caused Coronavirus Disease 2019 (COVID-19), and the impact of COVID-19 is still existing. Enterovirus 71 (EV71) is the primary pathogen of hand, foot, and mouth disease (HFMD), and no effective direct-acting antiviral drugs targeting EV71 has been approved yet. Innate antiviral strategies play an important role in preventing virus infections depending on the powerful immune regulatory system of body, while viruses have evolved to exploit diverse methods to overcome immune response. Viral proteases, which are known in cleaving viral polyproteins, have also been found to modulate the innate immunity of host cells, thereby promoting viral proliferation. Herein, we reviewed the current development of SARS-CoV-2 3CLpro, PLpro, and EV71 3Cpro and 2Apro, mainly including structure, function, modulation of immune response, and inhibitors of these four proteases, to further deepen the understanding of viral pathogenesis and provide a new perspective for subsequent corresponding drug development.
Collapse
Affiliation(s)
- Shulei Hu
- China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Qiuyu Zhong
- China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Xiong Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Shurui Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Jinlin Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China
| | - Hong Liu
- China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China.
| | - Wenhao Dai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China.
| |
Collapse
|
9
|
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
|
10
|
Chakraborty C, Lo YH, Bhattacharya M, Das A, Wen ZH. Looking beyond the origin of SARS-CoV-2: Significant strategic aspects during the five-year journey of COVID-19 vaccine development. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102527. [PMID: 40291378 PMCID: PMC12032352 DOI: 10.1016/j.omtn.2025.102527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
It has been five years since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and we are also approaching the five-year mark of the COVID-19 pandemic. The vaccine is a significant weapon in combating infectious diseases like SARS-CoV-2. Several vaccines were developed against SARS-CoV-2, and they demonstrated efficacy and safety during these five years. The rapid development of multiple next-generation vaccine candidates in different platforms with very little time is the success story of the vaccine development endeavor. This remarkable success of rapid vaccine development is a new paradigm for fast vaccine development that might help develop infectious diseases and fight against the pandemic. With the completion of five years since the beginning of SARS-CoV-2 origin, we are looking back on the five years and reviewing the milestones, vaccine platforms, animal models, clinical trials, successful collaborations, vaccine safety, real-world effectiveness, and challenges. Lessons learned during these five years will help us respond to public health emergencies and to fight the battle against future pandemics.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Yi-Hao Lo
- Department of Family Medicine, Zuoying Armed Forces General Hospital, Kaohsiung 81342, Taiwan
- Department of Nursing, Meiho University, Neipu Township, Pingtung County 91200, Taiwan
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, Odisha 756020, India
| | - Arpita Das
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, #70 Lien-Hai Road, Kaohsiung 804201, Taiwan
- National Museum of Marine Biology & Aquarium, # 2 Houwan Road, Checheng, Pingtung 94450, Taiwan
| |
Collapse
|
11
|
Dalla Pietà A, Genova B, Penna A, Sinigaglia A, Vogiatzis S, Barzon L, Pagliari M, Bonfante F, Torrigiani F, Sofia T, Verin R, Tosi A, Carpanese D, Sommaggio R, Barbieri V, Dalla Santa S, Zuccolotto G, Grigoletto A, Pasut G, Rosato A. On the adjuvanticity of hyaluronan: The case of a SARS-CoV-2 vaccine. J Control Release 2025; 382:113674. [PMID: 40164435 DOI: 10.1016/j.jconrel.2025.113674] [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: 07/21/2024] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Vaccines based on mRNA have been fundamental in facing the COVID-19 pandemic, however, they still raise concerns about stability and long-term efficacy. Thus, protein-based vaccines remain valid options and hence the study of effective adjuvants is crucial. Here, we developed a COVID-19 vaccine based on the receptor-binding domain (RBD) of SARS-CoV-2 Spike protein, which is covalently conjugated to the natural polymer hyaluronan (HA) that acts as an immunological adjuvant. Vaccination of K18-hACE2 mice with HA-RBD was well tolerated, and elicited high and sustained titres of RBD-binding antibodies and SARS-CoV-2-neutralizing antibodies, without the addition of other immunostimulatory compounds. Most importantly, HA-RBD vaccination conferred long-term protection to K18-hACE2 mice after challenge with SARS-CoV-2, also in the case of two consequent infections driven by different variants. These findings demonstrate the efficacy of HA-based vaccination against COVID-19 disease, and support the promising use of HA as an efficient and well tolerated adjuvant.
Collapse
Affiliation(s)
- Anna Dalla Pietà
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128 Padua, Italy
| | - Beatrice Genova
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128 Padua, Italy
| | - Alessandro Penna
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy
| | - Alessandro Sinigaglia
- Department of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy
| | - Stefania Vogiatzis
- Department of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy
| | - Luisa Barzon
- Department of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy
| | - Matteo Pagliari
- Department of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell'Università 10, 35020 Legnaro, PD, Italy
| | - Francesco Bonfante
- Department of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell'Università 10, 35020 Legnaro, PD, Italy
| | - Filippo Torrigiani
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, 35020 Legnaro, PD, Italy
| | - Tomasoni Sofia
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, 35020 Legnaro, PD, Italy
| | - Ranieri Verin
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell'Università 16, 35020 Legnaro, PD, Italy
| | - Anna Tosi
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy
| | - Debora Carpanese
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy
| | - Roberta Sommaggio
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128 Padua, Italy; Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy
| | - Vito Barbieri
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128 Padua, Italy
| | - Silvia Dalla Santa
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128 Padua, Italy
| | - Gaia Zuccolotto
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy
| | - Antonella Grigoletto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via Francesco Marzolo 5, 35131 Padua, Italy
| | - Gianfranco Pasut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via Francesco Marzolo 5, 35131 Padua, Italy.
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128 Padua, Italy; Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128 Padua, Italy.
| |
Collapse
|
12
|
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
|
13
|
Fathali F, Shokri Saravi M, Zarei Vanajemi P, Tafrihi M. Phlomis genus: bridging tradition and science in medicinal Research-a review of phytochemistry and pharmacological properties. Nat Prod Res 2025; 39:3281-3294. [PMID: 39616620 DOI: 10.1080/14786419.2024.2423029] [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: 12/27/2023] [Revised: 10/04/2024] [Accepted: 10/23/2024] [Indexed: 05/31/2025]
Abstract
The genus Phlomis contains more than 100 species distributed in North Africa and the Mediterranean region. This review highlights Phlomis-derived compounds' biological and pharmacological properties and their essential oils, with a special emphasis on anticancer activities. Relevant data was collected from scientific sources including Google Scholar, Science Direct, PubMed, and Springer Link by using the keyword "Phlomis". Also, the latest version of the plants' names was checked by www.worldfloraonline.org. Important Phytochemicals compounds like flavonoids, iridoids, and alkaloids, which were isolated from different Phlomis species showed significant biological activity, and inhibitory effects on various cancer cells (MCF-7, A549, HepG2, HT-29, etc.) via different mechanisms. The collected data strongly underpins the viewpoint that species belonging to the Phlomis genus have diverse biological and pharmaceutical activities to treat various diseases, including cancer, and their remarkable antimicrobial properties with no or minimal health and environmental hazards.
Collapse
Affiliation(s)
- Fatemeh Fathali
- Department of Cellular and Molecular Biology, University of Mazandaran, Mazandaran, Babolsar, Iran
| | - Manijeh Shokri Saravi
- Department of Cellular and Molecular Biology, University of Mazandaran, Mazandaran, Babolsar, Iran
| | - Paria Zarei Vanajemi
- TUM School of life sciences, Technische Universität München, Weihenstephan, Freising, Germany
| | - Majid Tafrihi
- Department of Cellular and Molecular Biology, University of Mazandaran, Mazandaran, Babolsar, Iran
| |
Collapse
|
14
|
Liu J, Chen X, Chen C, Wu J, Xie F, Li J, Han H, Zhao Y, Yang Y. Nonclinical safety and biodistribution evaluation of HC009 mRNA vaccine against COVID-19 in rat. Toxicology 2025; 514:154107. [PMID: 40064458 DOI: 10.1016/j.tox.2025.154107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 04/03/2025]
Abstract
mRNA-based technology has been evaluated in clinical trials for rapid control and prevention of emergencies and diseases. HC009, a mRNA vaccine encoding the full-length SARS-CoV-2 spike protein delivered via the QTsome platform, was tested in rats for immunogenicity, toxicity, and biodistribution. For immunogenicity and toxicity, rats received three intramuscular injections of HC009 at 3-week intervals followed by a 4-week observation period. In the biodistribution study, rats received a single intramuscular injection, with mRNA levels measured in organs at various time points. Results showed that HC009 elicited effective, long-lasting humoral immunity and Th1-biased cellular responses. The mRNA primarily localized to the injection site and spleen, with no observed vaccine-related toxicological reactions. These findings support HC009's potential for inducing an effective immune response with a favorable safety profile, warranting further clinical investigation.
Collapse
MESH Headings
- Animals
- COVID-19 Vaccines/pharmacokinetics
- COVID-19 Vaccines/toxicity
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Rats
- COVID-19/prevention & control
- COVID-19/immunology
- Tissue Distribution
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- SARS-CoV-2/immunology
- Male
- RNA, Messenger
- mRNA Vaccines
- Injections, Intramuscular
- Rats, Sprague-Dawley
- Vaccines, Synthetic/toxicity
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Female
- Immunity, Humoral/drug effects
- Antibodies, Viral/blood
Collapse
Affiliation(s)
- Juan Liu
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang 310020, China.
| | - Xicheng Chen
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang 310020, China
| | - Chuanqian Chen
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang 310020, China
| | - Jie Wu
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang 310020, China
| | - Fengyang Xie
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang 310020, China
| | - Jing Li
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang 310020, China
| | - Huafeng Han
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang 310020, China
| | - Yingying Zhao
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang 310020, China
| | - Yongsheng Yang
- Nucleic Acid Medicine Innovation Center, Zhejiang Haichang Biotech Co., Ltd., Hangzhou, Zhejiang 310020, China.
| |
Collapse
|
15
|
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
|
16
|
Sala C, Ninu A, Balducci V, Allegro G, Montalbano A, Lulli M, Boccitto ML, Guzzolino E, Spinelli V, Arcangeli A, Sartiani L, Cerbai E. Stable expression of SARS-CoV-2 envelope viroporin promotes intracellular calcium depletion in human cells: relevance for endoplasmic reticulum stress, cell proliferation, pluripotency and lineage differentiation. Cell Calcium 2025; 128:103032. [PMID: 40286431 DOI: 10.1016/j.ceca.2025.103032] [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: 11/27/2024] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
SARS-CoV-2 infection affects the respiratory system but also many tissues and organs that may be adversely compromised. Accordingly, recent evidence has assessed virus ability to infect different cell phenotypes, translate viral proteins and promote virus replication. Among them, Envelope (E) proteins sustain virus replication, promote inflammatory processes and remodelling of host cells. However, despite advances on structure and sequence, E-protein specific location and effects in human host cells are still controversial and poorly investigated. Using lentiviral vectors, we established HEK293 and hiPS cell lines stably expressing E-protein. Immunocytochemistry showed E-protein mainly locates within the endoplasmic reticulum, the ERGIC and the Golgi compartments, while only HEK293 cells display some protein staining in cell periphery suggesting a possible insertion into the plasmalemma. Electrophysiological recordings in HEK293 cells revealed E-protein self-assembles in the plasma membrane to mediate a cation efflux pore that is sensitive to amantadine blockade. Calcium fluorescence imaging in HEK293 and hiPS cells demonstrated E-protein expression induces a marked depletion of thapsigargin-sensitive intracellular calcium stores. The altered calcium homeostasis associates to reduced cell metabolic activity, mitochondrial potential, proliferation rate and promotes ER stress. Finally, trilineage differentiation of hiPS cells indicated E-protein expression preserves cell pluripotency while selectively impairs mesodermal differentiation. These results unveil a critical role of stable E-viroporin expression that through alteration of ER Ca²⁺ homeostasis, metabolic activity and induction of ER stress affects important cellular functions, including the differentiative process from pluripotent to mesodermal progenitors, a critical cell population in self-repair and homeostasis of most human tissue and organs.
Collapse
Affiliation(s)
- Cesare Sala
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Andrea Ninu
- Department of Neurofarba, University of Florence, Florence, Italy
| | | | - Giada Allegro
- Department of Neurofarba, University of Florence, Florence, Italy
| | - Alberto Montalbano
- G.E.A. Green Economy and Agriculture Centro per la Ricerca s.r.l, Pistoia, Italy
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | | | - Elena Guzzolino
- Department of Neurofarba, University of Florence, Florence, Italy
| | | | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Sartiani
- Department of Neurofarba, University of Florence, Florence, Italy.
| | | |
Collapse
|
17
|
Arunsiripate TT, Groeltz-Thrush J, Saeng-Chuto K, Guo B, Michael A, Siepker C, Derscheid RJ, Rahe MC, Zhang J, Burrough E, Piñeyro PE. Diagnostic investigation of porcine hemagglutinating encephalomyelitis virus as potential pathogen associated with respiratory clinical signs and pulmonary lesions in pigs. Microb Pathog 2025; 203:107493. [PMID: 40120700 DOI: 10.1016/j.micpath.2025.107493] [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: 09/12/2024] [Revised: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Porcine hemagglutinating encephalomyelitis virus (PHEV) is a member of the genus Betacoronavirus, known for its impact on the central and peripheral nervous systems in pigs. Traditionally associated with vomiting and wasting disease (VWD) and encephalomyelitis, PHEV was first reported in Canada in the late 1950s and has since been identified in numerous countries. Although serologic studies indicate global dissemination, the prevalence of PHEV remains unclear due to sporadic reporting and lack of active surveillance. Neonatal pigs are particularly vulnerable, with outbreaks resulting in high morbidity and mortality. Histopathological findings typically include non-suppurative encephalomyelitis and lymphoplasmacytic perivascular cuffs, gliosis, and neuronal degeneration. Recent observations have suggested a potential role for PHEV in respiratory disease, a hypothesis prompted by cases of influenza-like symptoms in pigs in Michigan in 2015 and corroborated by subsequent reports. This study aims to explore this possibility through a combination of clinical outbreak analysis and retrospective investigation. PHEV was confirmed via qPCR in 83.33 % of pigs examined for respiratory disease, with histological lesions such as necrotizing bronchitis and bronchiolitis. In-situ hybridization (ISH) confirmed the presence of PHEV mRNA in respiratory epithelium, and immunohistochemical analysis revealed significant macrophage infiltration in affected lung. Phylogenetic analysis indicated that PHEV strains from respiratory cases cluster closely with historical respiratory strains, though distinct from neurologic strains. This genetic differentiation suggests possible phenotypic variation contributing to respiratory tropism. The retrospective study identified PHEV in 7.62 % of cases with necrotizing bronchitis or bronchiolitis, reinforcing the virus's potential role in respiratory disease. Notably, PHEV co-infection with other respiratory pathogens such as PRRSV was observed, suggesting it may contribute to the porcine respiratory disease complex (PRDC). These findings suggest that PHEV is a significant respiratory pathogen in swine, warranting its inclusion in the differential diagnosis for respiratory disease in nursery pigs. Future research should focus on elucidating the pathogenesis of PHEV in respiratory disease, host-virus interactions, and the virus's impact on immune response and secondary infections. Understanding these factors will be crucial in developing effective preventive and therapeutic strategies against PHEV in swine.
Collapse
Affiliation(s)
- Trevor T Arunsiripate
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Jennifer Groeltz-Thrush
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Kepalee Saeng-Chuto
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Baoqing Guo
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Alyona Michael
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA; Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL, USA
| | - Christopher Siepker
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Rachel J Derscheid
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Michael C Rahe
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA; Department of Population Health and Pathobiology, North Caroline State University, NC, USA
| | - Jianqiang Zhang
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Eric Burrough
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA
| | - Pablo E Piñeyro
- Department of Veterinary Diagnostic & Production Animal Medicine, Iowa State University, IA, USA.
| |
Collapse
|
18
|
Chen R, Hao Z, Ye J, Zhao X, Hu S, Luo J, Li J, Wu H, Liang X, Shen C, Deng M, Zhang W, Zhu Z, Qin Y, Hu G, Zhang L, Cao F, Liu Y, Liu R, Sun Q, Wei H, Wang Z. Decoding post-mortem infection dynamics of SARS-CoV-2, IAV and RSV: New insights for public health and emerging infectious diseases management. J Infect 2025; 90:106489. [PMID: 40268146 DOI: 10.1016/j.jinf.2025.106489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/25/2025] [Accepted: 04/12/2025] [Indexed: 04/25/2025]
Abstract
OBJECTIVES The persistence and infectivity of respiratory viruses in cadavers remain poorly characterized, posing significant biosafety risks for forensic and healthcare professionals. This study systematically evaluates the post-mortem stability and transmission potential of SARS-CoV-2, influenza A virus (IAV), and respiratory syncytial virus (RSV) under varying environmental conditions, providing critical insights into viral kinetics. METHODS To assess the post-mortem stability of SARS-CoV-2, tissue samples were collected from infected cadavers at 4 ℃, room temperature (RT, 20-22 ℃), and 37 ℃ over a predetermined timeframe. Viral kinetics were analyzed using quantitative assays, while histopathology and immunohistochemistry characterized tissue-specific distribution. Additionally, comparative analyses were conducted both in vitro and in cadaveric tissues to characterize the survival dynamics of IAV and RSV under identical conditions. RESULTS SARS-CoV-2 exhibited prolonged post-mortem infectivity, persisting for up to 5 days at RT and 37 ℃ and over 7 days at 4 ℃, with the highest risk of transmission occurring within the first 72 h at RT and 24 h at 37 ℃. In contrast, RSV remained viable for 1-2 days, while IAV persisted for only a few hours post-mortem. Viral decay rates were temperature-dependent and varied across tissues, demonstrating distinct post-mortem survival kinetics. CONCLUSIONS This study presents the first comprehensive analysis of viral persistence in cadavers, revealing prolonged SARS-CoV-2 stability compared to IAV and RSV. These findings underscore the need for enhanced post-mortem biosafety protocols to mitigate occupational exposure risks in forensic and clinical settings. By elucidating viral decay dynamics across environmental conditions, this research establishes a critical foundation for infection control strategies, informing biosafety policies for emerging respiratory pathogens.
Collapse
Affiliation(s)
- Run Chen
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zeyi Hao
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jian Ye
- Institute of Forensic Science, Ministry of Public Security, Beijing 100038, China
| | - Xingchun Zhao
- Institute of Forensic Science, Ministry of Public Security, Beijing 100038, China
| | - Sheng Hu
- Institute of Forensic Science, Ministry of Public Security, Beijing 100038, China
| | - Jianliang Luo
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Junhua Li
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan, Hubei 430023, China
| | - Hao Wu
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - XingGong Liang
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chen Shen
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Mingyan Deng
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wanqing Zhang
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zhengyang Zhu
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yudong Qin
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Gengwang Hu
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Letong Zhang
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Fan Cao
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yuzhao Liu
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Ruina Liu
- Center for Translational Medicine, Shaanxi Belt and Road Joint Laboratory of Precision Medicine in Psychiatry, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi 710061, China
| | - Qinru Sun
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Hongping Wei
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan, Hubei 430023, China.
| | - Zhenyuan Wang
- Department of Forensic Pathology, College of Forensic Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
19
|
Parra RG, Komives EA, Wolynes PG, Ferreiro DU. Frustration in physiology and molecular medicine. Mol Aspects Med 2025; 103:101362. [PMID: 40273505 DOI: 10.1016/j.mam.2025.101362] [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: 01/30/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
Molecules provide the ultimate language in terms of which physiology and pathology must be understood. Myriads of proteins participate in elaborate networks of interactions and perform chemical activities coordinating the life of cells. To perform these often amazing tasks, proteins must move and we must think of them as dynamic ensembles of three dimensional structures formed first by folding the polypeptide chains so as to minimize the conflicts between the interactions of their constituent amino acids. It is apparent however that, even when completely folded, not all conflicting interactions have been resolved so the structure remains 'locally frustrated'. Over the last decades it has become clearer that this local frustration is not just a random accident but plays an essential part of the inner workings of protein molecules. We will review here the physical origins of the frustration concept and review evidence that local frustration is important for protein physiology, protein-protein recognition, catalysis and allostery. Also, we highlight examples showing how alterations in the local frustration patterns can be linked to distinct pathologies. Finally we explore the extensions of the impact of frustration in higher order levels of organization of systems including gene regulatory networks and the neural networks of the brain.
Collapse
Affiliation(s)
- R Gonzalo Parra
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Spain
| | | | - Peter G Wolynes
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA.
| | - Diego U Ferreiro
- Protein Physiology Lab, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina.
| |
Collapse
|
20
|
Zabiegala A, Kim Y, Chang KO. Host susceptibilities and entry processes of SARS-CoV-2 Omicron variants using pseudotyped viruses carrying spike protein. BMC Vet Res 2025; 21:377. [PMID: 40426227 PMCID: PMC12108000 DOI: 10.1186/s12917-025-04822-9] [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: 02/17/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
The zoonotic potential has been well studied for SARS-CoV-2 and its earlier variants, but the information for Omicron variants and SARS-CoV is lacking. In this study, we generated lentivirus-based pseudoviruses carrying spike protein (S) of SARS-CoV-2, parental and Omicron variants including BA.1.1, BA.4/5, XBB.1 and JN.1 to assess the entry into cells expressing human or animal ACE2 including dogs, cats and white-tailed deer. Using these pseudoviruses, along with pseudoviruses carrying S of MERS-CoV and SARS-CoV, we assessed the protease processing of these various S through western blotting, entry/inhibition assays, and fusion assays. The results showed that overall, pseudotyped viruses carrying each S of SARS-CoV-2 Omicron strains efficiently entered cells expressing human or animal ACE2 comparably (BA.1.1 and JN.1) or better (BA.4/5 and XBB.1) than those with parental strain. In addition, the entries of pseudotyped viruses carrying S of SARS-CoV were also efficient the cells expressing human or animal ACE2. The presence of TMPRSS2 significantly increased the entry of all tested pseudoviruses including those with S of MERS-CoV, SARS-CoV and SARS-CoV-2, with BA.1.1, JN1, and XBB.1 Omicron having the largest fold increase. When cathepsin inhibitors were examined to assess their inhibitory effects on entry of parental and Omicron variants, they were significantly less effective in the entry of Omicron variants compared to parent strain, suggesting Omicron strains do not depend on the endosomal route compared to parental strain.
Collapse
Affiliation(s)
- Alexandria Zabiegala
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, 1800 Denison Avenue, Manhattan, KS, 66506, USA
| | - Yunjeong Kim
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, 1800 Denison Avenue, Manhattan, KS, 66506, USA
| | - Kyeong-Ok Chang
- Department of Diagnostic Medicine and Pathobiology, College of Veterinary Medicine, Kansas State University, 1800 Denison Avenue, Manhattan, KS, 66506, USA.
| |
Collapse
|
21
|
Tang H, Zhuo Y, Chen J, Zhang R, Zheng M, Huang X, Chen Y, Huang M, Zeng Z, Huang X, Han C, Huang Y. Immune evasion, infectivity, and membrane fusion of the SARS-CoV-2 JN.1 variant. Virol J 2025; 22:162. [PMID: 40413500 PMCID: PMC12103042 DOI: 10.1186/s12985-025-02737-3] [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: 02/19/2025] [Accepted: 04/10/2025] [Indexed: 05/27/2025] Open
Abstract
SARS-CoV-2 undergoes continuous mutations during transmission, resulting in a variety of Omicron subvariants. Currently, SARS-CoV-2 BA.2.86 and its descendants JN.1, KP.2, KP.1.1 have been identified as the primary variants spreading globally. These emerging Omicron variants have increased transmissibility, potentially elevating the risk of viral reinfection in the population. However, the biological characteristics of newly-emerged Omicron subvariants in infecting host cells remain unclear. In this study, we assessed the neutralization effect of BA.2.86 and its descendant JN.1, as well as D614G, BA.2, BA.4/5, XBB.1.5, EG.5.1, HV.1, HK.3, JD.1.1 and JG.3 on convalescent sera obtained from individuals infected with BA.5 or XBB.1.5 strain. We evaluated the biological characteristics of variants spike proteins by measuring viral infectivity, affinity for receptors, and membrane fusion. Compared to XBB-related subvariants, BA.2.86 exhibited a diminished immune escape response, but JN.1 displayed a markedly augmented immune escape capability, which was closely related to its rapid transmission. BA.2.86 was less infectious in susceptible cells, while the JN.1 variant exhibited relatively high infectivity. Notably, BA.2.86 and JN.1 exhibited low fusion activity in 293 T-ACE2 cells, but relatively high fusogenicity in transmembrane protease serine 2 (TMPRSS2) overexpression cells. This study explored the evolutionary characteristics of emerging Omicron subvariants in host adaptation, and provided new strategies for the prevention and treatment of coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Haijun Tang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Yanhang Zhuo
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Jianlin Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Rongzhao Zhang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Miao Zheng
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350001, China
| | - Xinghua Huang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Yisheng Chen
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Minjian Huang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Zhaonan Zeng
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China
| | - Xueping Huang
- Department of Gastroenterology, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Department of Gastroenterology, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China.
| | - Chenfeng Han
- Department of Blood Transfusion, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215000, China.
| | - Yi Huang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, China.
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China.
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China.
- Central Laboratory, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, China.
- Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Key Laboratory of Critical Care Medicine, Fuzhou, Fujian, 350001, China.
| |
Collapse
|
22
|
Frausto-Avila M, León-Montiel RDJ, Quiroz-Juárez MA, U'Ren AB. Prospective study using artificial neural networks for identification of high-risk COVID-19 patients. Sci Rep 2025; 15:18005. [PMID: 40410212 PMCID: PMC12102217 DOI: 10.1038/s41598-025-00925-3] [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: 11/27/2024] [Accepted: 05/02/2025] [Indexed: 05/25/2025] Open
Abstract
The COVID-19 pandemic caused a major public health crisis, with severe impacts on global health and the economy. Machine learning (ML) has been crucial in developing new technologies to address challenges posed by the pandemic, particularly in identifying high-risk COVID-19 patients. This identification is vital for efficiently allocating hospital resources and controlling the virus's spread. Comprehensive validation of these intelligent approaches is necessary to confirm their clinical usefulness and help create future strategies for managing viral outbreaks. Here we present a prospective study to evaluate the performance of state-of-the-art ML models designed to identify high-risk COVID-19 patients across four clinical stages. Using artificial neural networks trained with historical patient data from Mexico, we assess the models' accuracy across six epidemiological waves without retraining them. We then compare their performance against neural networks trained with cumulative historical data up to the end of each wave. The findings reveal that models trained on early data can effectively predict high-risk patients in later waves, despite changes in vaccination rates, viral strains, and treatments. These results suggest that artificial intelligence-based patient classification methods could be robust tools for future pandemics, aiding in predicting clinical outcomes under evolving conditions.
Collapse
Affiliation(s)
- Mateo Frausto-Avila
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, 76230, Querétaro, Mexico
| | - Roberto de J León-Montiel
- Instituto de Ciencias Nucleares, Universidad Nacional Autónoma de México, Apartado Postal 70-543, 04510, Mexico, CDMX, Mexico
| | - Mario A Quiroz-Juárez
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Boulevard Juriquilla 3001, 76230, Querétaro, Mexico.
| | - Alfred B U'Ren
- Instituto de Ciencias Nucleares, Universidad Nacional Autónoma de México, Apartado Postal 70-543, 04510, Mexico, CDMX, Mexico
| |
Collapse
|
23
|
Kumar N, Segovia D, Kumar P, Atti HB, Kumar S, Mishra J. Mucosal implications of oral Jak3-targeted drugs in COVID patients. Mol Med 2025; 31:203. [PMID: 40410684 PMCID: PMC12100796 DOI: 10.1186/s10020-025-01260-z] [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: 03/18/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025] Open
Abstract
The JAK family, particularly JAK3, plays a crucial role in immune signaling and inflammatory responses. Dysregulated JAK3 activation in SARS-CoV-2 infections has been associated with severe inflammation and respiratory complications, making JAK inhibitors a viable therapeutic option. However, their use raises concerns regarding immunosuppression, which could increase susceptibility to secondary infections. While long-term adverse effects are less of a concern in acute COVID-19 treatment, patient selection and monitoring remain critical. Furthermore, adverse effects associated with oral JAK3 inhibitors necessitate the exploration of alternative strategies to optimize therapeutic efficacy while minimizing risks. This review highlights the role of JAK3 in immune and epithelial cells, examines the adverse effects of oral JAK3 inhibitors in COVID-19 and other treatments, and discusses alternative therapeutic strategies for improving patient outcomes.
Collapse
Affiliation(s)
- Narendra Kumar
- ILR-College of Pharmacy, Texas A&M University Health Science Center, Kingsville, TX, USA.
| | - Daniel Segovia
- ILR-College of Pharmacy, Texas A&M University Health Science Center, Kingsville, TX, USA
| | - Priyam Kumar
- University of Pennsylvania, Philadelphia, PA, USA
| | - Hima Bindu Atti
- ILR-College of Pharmacy, Texas A&M University Health Science Center, Kingsville, TX, USA
| | - Soaham Kumar
- Veterans Memorial High School, Corpus Christi, TX, USA
| | - Jayshree Mishra
- ILR-College of Pharmacy, Texas A&M University Health Science Center, Kingsville, TX, USA.
| |
Collapse
|
24
|
Ghimire R, Shrestha R, Amaradhi R, Liu L, More S, Ganesh T, Ford AK, Channappanavar R. Toll-like receptor 7 (TLR7)-mediated antiviral response protects mice from lethal SARS-CoV-2 infection. J Virol 2025; 99:e0166824. [PMID: 40162785 PMCID: PMC12090760 DOI: 10.1128/jvi.01668-24] [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: 09/23/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced impaired antiviral immunity and excessive inflammatory responses cause lethal pneumonia. However, the in vivo roles of key pattern recognition receptors that elicit protective antiviral and fatal inflammatory responses, specifically in the lungs, are not well described. Coronaviruses possess single-stranded RNA genome that activates TLR7/8 to induce an antiviral interferon (IFN) and robust inflammatory cytokine response. Here, using wild-type and TLR7-deficient (TLR7-/-) mice infected with mouse-adapted SARS-CoV-2 (MA-CoV-2), we examined the role of TLR7 in the lung antiviral and inflammatory response and severe pneumonia. We showed that TLR7 deficiency significantly increased lung virus loads and morbidity/mortality, which correlated with reduced levels of type I IFNs (Ifna/b), type III IFNs (Ifnl), and IFN-stimulated genes (ISGs) in the lungs. A detailed evaluation of MA-CoV-2-infected lungs revealed increased neutrophil accumulation and lung pathology in TLR7-/- mice. We further showed that blocking type I IFN receptor (IFNAR) signaling enhanced SARS-CoV-2 replication in the lungs and caused severe lung pathology, leading to 100% mortality compared to infected control mice. Moreover, immunohistochemical assessment of the lungs revealed increased numbers of SARS-CoV-2 antigen-positive macrophages, pneumocytes, and bronchial epithelial cells in TLR7-/- and IFNAR-deficient mice compared to control mice. In summary, we conclusively demonstrated that despite TLR7-induced robust lung inflammation, TLR7-induced IFN/ISG responses suppress lung virus replication and pathology and provide protection against SARS-CoV-2-induced fatal pneumonia. Additionally, given the similar disease outcomes in control, TLR7-/-, and IFNAR-deficient MA-CoV-2-infected mice and coronavirus disease 2019 (COVID-19) patients, we propose that MA-CoV-2-infected mice constitute an excellent model for studying COVID-19.IMPORTANCESevere coronavirus disease 2019 (COVID-19) is caused by a delicate balance between a strong antiviral and an exuberant inflammatory response. A robust antiviral immunity and regulated inflammation are protective, while a weak antiviral response and excessive inflammation are detrimental. However, the key host immune sensors that elicit protective antiviral and inflammatory responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge are poorly defined. Here, we examined the role of viral RNA-mediated TLR7 activation in the lung antiviral and inflammatory responses in SARS-CoV-2-infected mice. We demonstrate that TLR7 deficiency led to a high rate of morbidity and mortality, which correlated with an impaired antiviral interferon (IFN)-I/III response, enhanced lung virus replication, and severe lung pathology. Furthermore, we show that blocking IFN-I signaling using anti-IFN receptor antibody promoted SARS-CoV-2 replication in the lungs and caused severe disease. These results provide conclusive evidence that TLR7 and IFN-I receptor deficiencies lead to severe disease in mice, replicating clinical features observed in COVID-19 patients.
Collapse
Affiliation(s)
- Roshan Ghimire
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Rakshya Shrestha
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Radhika Amaradhi
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lin Liu
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Sunil More
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Alexandra K. Ford
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| | - Rudragouda Channappanavar
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma, USA
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, Oklahoma, USA
| |
Collapse
|
25
|
Sun H, Yang Q, Zhang Y, Cui S, Zhou Z, Zhang P, Jia L, Zhang M, Wang Y, Chen X, Pei R. Syntaxin-6 restricts SARS-CoV-2 infection by facilitating virus trafficking to autophagosomes. J Virol 2025; 99:e0000225. [PMID: 40277356 PMCID: PMC12090716 DOI: 10.1128/jvi.00002-25] [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: 01/07/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Despite the diminishing global impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus continues to circulate and undergo mutations, posing ongoing challenges for public health. A comprehensive understanding of virus entry mechanisms is crucial for managing new epidemic strains. However, the cellular processes post-endocytosis remain largely unexplored. This study employs proximity labeling to examine proteins near ACE2 post-viral infection and identified syntaxin-6 (STX6) as a factor that inhibits SARS-CoV-2 infection by impeding the endocytic release of the virus. SARS-CoV-2 infection enhances early endosome recruitment of STX6. STX6 appears to hinder the maturation of viral particles-laden early endosomes into late endosomes, from which the virus could escape. Instead, it promotes the trafficking of the virus toward the autophagy-lysosomal degradation pathway. STX6 exhibits a broad-spectrum effect against various SARS-CoV-2 variants and several other viruses that enter via endocytosis. We report for the first time the function of STX6 as a restrictive factor in viral infection.IMPORTANCEVirus entry is the first step of the virus life cycle, and the exploitation of the endo-lysosome pathway for cellular entry by viruses has been well documented. Meanwhile, the intrinsic defense present within cells interferes with virus entry. We identified STX6 as a host restriction factor for viral entry by facilitating the virus trafficking to the autophagy-lysosomal degradation pathway. Notably, STX6 exhibits broad-spectrum antiviral activity against diverse severe acute respiratory syndrome coronavirus 2 variants and other viruses employing endocytosis for entry.
Collapse
Affiliation(s)
- Hao Sun
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qi Yang
- Guangzhou Laboratory, Guangzhou, China
| | - Yecheng Zhang
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Saisai Cui
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhe Zhou
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Peilu Zhang
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lijia Jia
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Mingxia Zhang
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yun Wang
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xinwen Chen
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Guangzhou Laboratory, Guangzhou, China
| | - Rongjuan Pei
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
26
|
Zhou J, Liu X, Xu Y, Wang J, Qian T, Sang X, Hasan MN, Warshel A, An J, Saha A, Huang Z. Computational and Experimental Study of the Conformational Variation of the Catalytic Residue His41 of the SARS-CoV-2 Main Protease. J Phys Chem B 2025. [PMID: 40387138 DOI: 10.1021/acs.jpcb.5c01718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
The main protease (Mpro) is essential for the replication of SARS-CoV-2, making it one of the major therapeutic targets for COVID-19 treatment. Here, we explored the conformational dynamics and energetics of the catalytic residue His41 in Mpro, as revealed by a rare conformational shift observed in the cocrystal structures of Mpro bound by certain inhibitors. Using steered molecular dynamics combined with umbrella sampling, we demonstrated that π-cation interactions between these inhibitors and the ionized catalytic dyad significantly reduced the energy barrier for the conformational flip of the His41 side chain. To further investigate the structure-activity relationship linked to this conformational change, we designed and synthesized a series of covalent inhibitors that control His41 flipping. Among these, compound H102-7 exhibited remarkable inhibitory activity with an IC50 of 5 nM. Drug resistance studies revealed that these inhibitors displayed improved resistance profiles compared to the clinically approved Mpro covalent inhibitor, Nirmatrelvir. This study integrates computational simulations, medicinal chemistry, and molecular biology to uncover an interesting allosteric effect of a key catalytic residue of SARS-CoV-2 Mpro and yields new promising molecules for the further development of Mpro-targeted therapeutic intervention.
Collapse
Affiliation(s)
- Jiao Zhou
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, Chinese University of Hong Kong, Shenzhen 518172, China
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xiang Liu
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China
- Department of Medicine, Division of Infectious Diseases and Global Public Health, School of Medicine, University of California at San Diego, La Jolla, California 92037, United States
| | - Yan Xu
- Department of Medicine, Division of Infectious Diseases and Global Public Health, School of Medicine, University of California at San Diego, La Jolla, California 92037, United States
| | - Juan Wang
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Tingli Qian
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, Chinese University of Hong Kong, Shenzhen 518172, China
| | - Xiaohong Sang
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, Chinese University of Hong Kong, Shenzhen 518172, China
| | - Md Nazmul Hasan
- Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53213, United States
| | - Arieh Warshel
- Department of Chemistry, University of Southern California, Los Angeles, California 90089, United States
| | - Jing An
- Department of Medicine, Division of Infectious Diseases and Global Public Health, School of Medicine, University of California at San Diego, La Jolla, California 92037, United States
| | - Arjun Saha
- Department of Chemistry & Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53213, United States
| | - Ziwei Huang
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, Chinese University of Hong Kong, Shenzhen 518172, China
- Department of Medicine, Division of Infectious Diseases and Global Public Health, School of Medicine, University of California at San Diego, La Jolla, California 92037, United States
| |
Collapse
|
27
|
Zhan J, Zhong F, Dai L, Ma J, Chai Y, Zhao X, Chang L, Zhang Y, Wang J, Tang Y, Zhong WZ, Zhang G, Li L, Zhu Q, Chen Z, Xia X, Peng L, Wu J, Li R, Li D, Zhu Y, Zhou X, Wu Y, Chen R, Li J, Li Y, Shu H. Perioperative SARS-CoV-2 infection and postoperative complications: a single-centre retrospective cohort study in China. BMJ Open 2025; 15:e093044. [PMID: 40389317 PMCID: PMC12090866 DOI: 10.1136/bmjopen-2024-093044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 04/15/2025] [Indexed: 05/21/2025] Open
Abstract
OBJECTIVE To explore the association between perioperative SARS-CoV-2 infection and the postoperative complications during the breakout of the Omicron epidemic wave. DESIGN Observational retrospective cohort study. Multivariable logistic regression was performed to explore the association between the duration from surgery to COVID-19 diagnosis and the likelihood of postoperative complications. SETTING A general hospital in China. PARTICIPANTS 7927 patients aged 18 years and older who underwent surgical treatment between 1 December 2022 and 28 February 2023. PRIMARY OUTCOME MEASURES The outcome was a composite of postoperative adverse events that occurred within the initial 30 postoperative days. RESULTS Of all patients, 420 (11.76%) experienced postoperative complications. Compared with No COVID-19, preoperative COVID-19 within 1 week (pre-1w) exhibited a high risk of postoperative complications (adjusted OR (aOR), 2.67; 95% CI 1.50 to 4.78), followed by patients with pre-2w (aOR, 2.14; 95% CI 1.20 to 3.80). For patients with postoperative COVID-19 within 1 week (post-1w), the aOR was 2.48 (95% CI 1.48 to 4.13), followed by patients with post-2w (aOR 1.95; 95% CI 1.10 to 3.45), and those with post-3w (aOR 2.25; 95% CI 1.27 to 3.98). The risks of postoperative complications decreased roughly with the increase of the time interval between the surgery date and SARS-CoV-2 infection. Stratification analyses suggested that perioperative COVID-19 increased the risk of postoperative complications in older patients, smokers, those with comorbidities or experiencing moderate or severe COVID-19 symptoms. CONCLUSIONS Our findings reveal a significant time-dependent relationship between perioperative COVID-19 and postoperative complications, highlighting the importance of tailored preoperative risk evaluations, enhanced postoperative surveillance, and the implementation of effective postoperative COVID-19 prevention measures. TRIAL REGISTRATION NUMBER ChiCTR2300072473.
Collapse
Affiliation(s)
- Jia Zhan
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Fei Zhong
- Global Health Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - LingYan Dai
- Global Health Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jue Ma
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - YunFei Chai
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou, China
| | - XiRui Zhao
- Global Health Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lu Chang
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - YiDan Zhang
- Global Health Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - JunJiang Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yong Tang
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Guangyan Zhang
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Le Li
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qiang Zhu
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - ZhiHao Chen
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xin Xia
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - LiShan Peng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jing Wu
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - RuiYun Li
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - DanYang Li
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yan Zhu
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xin Zhou
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - YiChun Wu
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - RuiRong Chen
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jie Li
- Global Health Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Public Health, Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Epidemiology, School of Public Health, Brown University, Providence, RI, USA
| | - Yong Li
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - HaiHua Shu
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
28
|
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
|
29
|
Lin HF, Jiang RD, Qin RX, Yao B, Zeng WT, Gao Y, Shi AM, Li JM, Liu MQ. Characterization of a SARS-CoV-2 Infection Model in Golden Hamsters with Diabetes Mellitus. Virol Sin 2025:S1995-820X(25)00059-8. [PMID: 40389095 DOI: 10.1016/j.virs.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 05/12/2025] [Indexed: 05/21/2025] Open
Abstract
Being widespread across the globe, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) keeps evolving and generating new variants and continuously poses threat to public health, especially to the population with chronic comorbidities. Diabetes mellitus is one of high-risk factors for severe outcome of coronavirus disease 2019 (COVID-19). Establishment of animal models that parallel the clinical and pathological features of COVID-19 complicated with diabetes is thus highly essential. Here, in this study, we constructed leptin receptor gene knockout hamsters with the phenotype of diabetes mellitus (db/db), and revealed that the diabetic hamsters were more susceptible to SARS-CoV-2 and its variants than wild-type hamsters. SARS-CoV-2 and its variants induced a stronger immune cytokine response in the lungs of diabetic hamsters than in wild-type hamsters. Comparative histopathology analyses also showed that infection of SARS-CoV-2 and the variants caused more severe lung tissue injury in diabetic hamsters, and may induce serious complications such as diabetic kidney disease and cardiac lesions. Our findings demonstrated that despite the decreased respiratory pathogenicity, the SARS-CoV-2 variants were still capable of impairing other organs such as kidney and heart in diabetic hamsters, suggesting that the risk of evolving SARS-CoV-2 variants to diabetic patients should never be neglected. This hamster model may help better understand the pathogenesis mechanism of severe COVID-19 in patients with diabetes. It will also aid in development and testing of effective therapeutics and prophylactic treatments against SARS-CoV-2 variants among these high-risk populations.
Collapse
Affiliation(s)
- Hao-Feng Lin
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Laboratory Clinical Base, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510120, China
| | - Ren-Di Jiang
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200433, China
| | - Rui-Xin Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Laboratory Animal Center, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China
| | - Bing Yao
- Jinling Hospital Department Reproductive Medical Center, Nanjing Medical University, Nanjing 211166, China
| | - Wen-Tao Zeng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Laboratory Animal Center, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China
| | - Yun Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Laboratory Animal Center, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China.
| | - Ai-Min Shi
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Laboratory Animal Center, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China.
| | - Jian-Min Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Laboratory Animal Center, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Department of Cell Biology, Animal Core facility, Key Laboratory of Model Animal, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China.
| | - Mei-Qin Liu
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Laboratory Clinical Base, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510120, China.
| |
Collapse
|
30
|
Yang JH, Luo CF, Xiang R, Min JM, Shao ZT, Zhao YL, Chen L, Huang L, Zhang Y, Liu SS, Li YQ, Pu EN, Shi WQ, Pan HF, Chen WJ, Du CH, Jiang JF. Host taxonomy and environment shapes insectivore viromes and viral spillover risks in Southwestern China. MICROBIOME 2025; 13:122. [PMID: 40380277 DOI: 10.1186/s40168-025-02115-9] [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] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 04/15/2025] [Indexed: 05/19/2025]
Abstract
BACKGROUND Zoonotic viruses originating from small mammals pose significant challenges to public health on a global scale. Insectivores, serving as natural reservoirs for a diverse array of zoonotic viruses, are known to carry a multitude of viral species. However, compared to the extensive research conducted on rodents (Rodentia) and bats (Chiroptera), the role of insectivores in harboring and transmitting unknown pathogens remains underexplored, which may lead to a severe underestimation of their contributions and impact to global public health. RESULTS This study employed a meta-transcriptomic approach to profile the viromes of 214 individual insectivores, encompassing 13 species from the families Soricidae, Erinaceidae, and Talpidae, collected across 12 counties in Yunnan Province, a recognized zoonotic hotspot. Based on virus reads, the analysis identified 42 viral families associated with vertebrates, highlighting significant virome diversity and host-specific viral tropisms among shrews, hedgehogs, and moles, along with notable geographic and environmental specificity of the viruses. Shrews exhibited greater viral richness and abundance compared to hedgehogs and moles, with variations influenced predominantly by host taxonomy, altitude, and geographic location. A total of 114 RNA-dependent RNA polymerase sequences were obtained, leading to the identification of 68 viruses, including 57 novel species. Instances of host jumping were observed in 11 viruses, with potential pathogenic viruses related to Mojiang paramyxovirus and members of the Hantaviridae family. Cross-species transmission was predominantly observed in viruses carried by shrews, while moles may play a pivotal role in facilitating viral transmission among insectivores. CONCLUSIONS This study enhances the understanding of the high diversity of mammalian viruses among insectivores in a relatively confined region and underscores the associations between virome composition and related zoonotic risks, providing a foundation for proactive measures to prevent and control the spillover of emerging zoonotic pathogens and potential future outbreaks. Video Abstract.
Collapse
Affiliation(s)
- Ji-Hu Yang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
- School of Public Health, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Chun-Feng Luo
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
- School of Public Health, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Rong Xiang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
| | - Jiu-Meng Min
- Huo-Yan Engineering Technology, BGI-Shenzhen, Shenzhen, 518083, People's Republic of China
| | - Zong-Ti Shao
- Yunnan Key Laboratory for Zoonosis Control and Prevention, Yunnan Institute for Endemic Diseases Control and Prevention, Dali, 671000, People's Republic of China
| | - Yi-Lin Zhao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
| | - Lu Chen
- Beijing Macro & Micro-Test Bio-Tech Co., Ltd, Beijing, 101300, People's Republic of China
| | - Lin Huang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
| | - Yun Zhang
- Yunnan Key Laboratory for Zoonosis Control and Prevention, Yunnan Institute for Endemic Diseases Control and Prevention, Dali, 671000, People's Republic of China
| | - Shun-Shuai Liu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
- Zibo Center for Disease Control and Prevention, Zibo, 255020, People's Republic of China
| | - Yu-Qiong Li
- Yunnan Key Laboratory for Zoonosis Control and Prevention, Yunnan Institute for Endemic Diseases Control and Prevention, Dali, 671000, People's Republic of China
| | - En-Nian Pu
- Yunnan Key Laboratory for Zoonosis Control and Prevention, Yunnan Institute for Endemic Diseases Control and Prevention, Dali, 671000, People's Republic of China
| | - Wen-Qiang Shi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China
| | - Hai-Feng Pan
- School of Public Health, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Wei-Jun Chen
- Huo-Yan Engineering Technology, BGI-Shenzhen, Shenzhen, 518083, People's Republic of China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| | - Chun-Hong Du
- Yunnan Key Laboratory for Zoonosis Control and Prevention, Yunnan Institute for Endemic Diseases Control and Prevention, Dali, 671000, People's Republic of China.
| | - Jia-Fu Jiang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, People's Republic of China.
- School of Public Health, Anhui Medical University, Hefei, 230032, People's Republic of China.
| |
Collapse
|
31
|
Pantoja C, Acosta FM, Granatir S, Anderson M, Wyr M, Tailor J, Fuori A, Dower W, Marr HB, Ramirez PW. Electromagnetic waves destabilize the SARS-CoV-2 Spike protein and reduce SARS-CoV-2 Virus-Like particle (SC2-VLP) infectivity. Sci Rep 2025; 15:16836. [PMID: 40374718 PMCID: PMC12081674 DOI: 10.1038/s41598-025-01896-1] [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: 09/11/2024] [Accepted: 05/09/2025] [Indexed: 05/17/2025] Open
Abstract
Infection and transmission of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) continues to pose a global public health concern. Using electromagnetic waves represents an alternative strategy to inactivate pathogenic viruses such as SARS-CoV-2. However, whether electromagnetic waves reduce SARS-CoV-2 infectivity is unclear. Here, we adapted a coplanar waveguide (CPW) to identify frequencies that could potentially neutralize SARS-CoV-2 virus-like particles (SC2-VLPs). Treatment of SC2-VLPs at frequencies between 2.5 and 3.5 GHz and an electric field of 413 V/m reduced infectivity. Exposure of SC2-VLPs to a frequency of 3.1 GHz -and to a lesser extent, 5.9 GHz- reduced their binding to antibodies targeting the SARS-CoV-2 Spike S1 receptor-binding domain (RBD) but did not alter the total levels of Spike, Nucleocapsid, Envelope, or Membrane proteins in virus particles. These results suggest that electromagnetic waves alter the conformation of Spike, thereby reducing viral attachment and entry. Overall, this data provides proof-of-concept in using electromagnetic waves for sanitation and prevention efforts to curb the transmission of SARS-CoV-2 and potentially other pathogenic enveloped viruses.
Collapse
Affiliation(s)
- Christina Pantoja
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Francisco M Acosta
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | | | - Michael Anderson
- 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
| | - Johann Tailor
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Angus Fuori
- 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
|
32
|
Joyce JD, Moore GA, Thompson CK, Bertke AS. Guinea Pigs Are Not a Suitable Model to Study Neurological Impacts of Ancestral SARS-CoV-2 Intranasal Infection. Viruses 2025; 17:706. [PMID: 40431717 PMCID: PMC12116045 DOI: 10.3390/v17050706] [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: 03/29/2025] [Revised: 05/06/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Neurological symptoms involving the central nervous system (CNS) and peripheral nervous system (PNS) are common complications of acute COVID-19 as well as post-COVID conditions. Most research into these neurological sequalae focuses on the CNS, disregarding the PNS. Guinea pigs were previously shown to be useful models of disease during the SARS-CoV-1 epidemic. However, their suitability for studying SARS-CoV-2 has not been experimentally demonstrated. To assess the suitability of guinea pigs as models for SARS-CoV-2 infection and the impact of SARS-CoV-2 infection on the PNS, and to determine routes of CNS invasion through the PNS, we intranasally infected wild-type Dunkin-Hartley guinea pigs with ancestral SARS-CoV-2 USA-WA1/2020. We assessed PNS sensory neurons (trigeminal ganglia, dorsal root ganglia), autonomic neurons (superior cervical ganglia), brain regions (olfactory bulb, brainstem, cerebellum, cortex, hippocampus), lungs, and blood for viral RNA (RT-qPCR), protein (immunostaining), and infectious virus (plaque assay) at three- and six-days post infection. We show that guinea pigs, which have previously been used as a model of SARS-CoV-1 pulmonary disease, are not susceptible to intranasal infection with ancestral SARS-CoV-2, and are not useful models in assessing neurological impacts of infection with SARS-CoV-2 isolates from the early pandemic.
Collapse
Affiliation(s)
- Jonathan D. Joyce
- Translational Biology, Medicine and Health, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA;
- Center for Emerging Zoonotic and Arthropod-Borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Greyson A. Moore
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Christopher K. Thompson
- School of Neuroscience, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Andrea S. Bertke
- Center for Emerging Zoonotic and Arthropod-Borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| |
Collapse
|
33
|
Huang Q, Kang L, Wei X, Gong C, Xie H, Li M, Wang Y, Dong M, Huang F. Epidemiology and genetic diversity of common human coronaviruses in Beijing, 2015-2023: A prospective multicenter study. Int J Infect Dis 2025:107926. [PMID: 40379085 DOI: 10.1016/j.ijid.2025.107926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/14/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
OBJECTIVES To investigate the epidemiological and genetic features of common human coronaviruses (HCoVs) in Beijing in the context of the COVID-19 pandemic. METHODS We collected clinical samples from patients with acute respiratory tract infections (ARTIs) in 35 sentinel hospitals from 2015 to 2023. HCoVs were detected via multiple real-time PCRs, and S gene sequencing and phylogenetic analysis were subsequently performed. RESULTS From 2015 to 2023, the combined detection rate of HCoVs was 1.55% (909/58,550). During the COVID-19 pandemic, a significant increase in HCoVs detection was observed (P < 0.001). Overall, the epidemic season of four HCoVs was from July to October, and each HCoV showed different epidemic seasons. Notably, HCoV-NL63 and HCoV-229E exhibited pronounced annual alternations in prevalence. The highest combined detection rates of HCoVs were in the ≥60 years age group (1.85%), followed by the 0-5 years age group (1.48%). HCoV-229E was more prevalent in patients with severe community-acquired pneumonia (sCAP) (P=0.001). Phylogenetic analyses revealed that the four HCoVs were subjected to negative selection pressure, and multiple high-frequency amino acid site mutations were observed. HCoV-229E formed an emerging lineage after 2021. CONCLUSIONS This nine-year multicenter study in Beijing systematically elucidated that the four HCoVs exhibit distinct epidemiological characteristics, susceptible populations, and common mutations in amino acid sites, especially in the context of COVID-19. Therefore, continuous epidemiological surveillance and genetic characterization studies are imperative for predictive warning and timely identification of emerging coronavirus.
Collapse
Affiliation(s)
- Qi Huang
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, China.
| | - Lu Kang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100013, China.
| | - Xiaofeng Wei
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100013, China.
| | - Cheng Gong
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100013, China.
| | - Hui Xie
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100013, China.
| | - Maozhong Li
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100013, China.
| | - Yiting Wang
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100013, China.
| | - Mei Dong
- Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100013, China.
| | - Fang Huang
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Center for Disease Prevention and Control, Beijing Academy for Preventive Medicine, Beijing Institute of Tuberculosis Control Research and Prevention, Beijing 100013, China; Beijing Research Center for Respiratory Infectious Diseases, Beijing 100013, China.
| |
Collapse
|
34
|
Nagai T, Mauliana A, Kobayashi K, Yamaguchi A, Miyazaki K, Yang Y, Takeshita J, Fujita T, Sunada K, Ishiguro H, Kondo T, Miyauchi M. Broad-spectrum antimicrobial effects of hydrogen boride nanosheets. J Mater Chem B 2025; 13:5723-5733. [PMID: 40266560 DOI: 10.1039/d4tb02854f] [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: 04/24/2025]
Abstract
Hydrogen boride (HB) nanosheets are novel 2D materials that have found application in various fields such as electronics, energy storage, and catalysis. The present study describes the novel antimicrobial effects of HB nanosheets. Transparent thin films of HB coated on a glass substrate inactivate pathogens, such as the omicron variant of SARS-CoV-2, influenza virus, feline calicivirus, and bacteriophages. The infectious titer of these microbes decreases to the detection limit within 10 min in the dark at room temperature. The antiviral function of the HB nanosheets is retained in the absence of moisture, mimicking the environment of dry surfaces. The HB nanosheets also inactivate bacteria and fungi such as Escherichia coli, Staphylococcus aureus, Aspergillus niger, and Penicillium pinophilum. We discussed the mechanism of the broad-spectrum antimicrobial function of HB nanosheets based on the physicochemical properties of HB nanosheets. Denaturation of microbial agents is derived from strong physicochemical interactions between the protein molecules in the pathogens and the surface of the HB films. The present study reports important new properties of HB nanosheets and demonstrates their utility in protecting against the spread of disease on a pandemic scale.
Collapse
Affiliation(s)
- Takeshi Nagai
- Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, 210-0821, Japan.
| | - Andi Mauliana
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Institute of Science Tokyo, Meguro, Tokyo, 152-8552, Japan.
| | - Keiichi Kobayashi
- Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, 210-0821, Japan.
| | - Akira Yamaguchi
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Institute of Science Tokyo, Meguro, Tokyo, 152-8552, Japan.
| | - Keisuke Miyazaki
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Institute of Science Tokyo, Meguro, Tokyo, 152-8552, Japan.
| | - Yue Yang
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Institute of Science Tokyo, Meguro, Tokyo, 152-8552, Japan.
| | - Jumpei Takeshita
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Institute of Science Tokyo, Meguro, Tokyo, 152-8552, Japan.
| | - Takeshi Fujita
- School of Engineering Science, Kochi University of Technology, Kochi 782-8502, Japan
| | - Kayano Sunada
- Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, 210-0821, Japan.
| | - Hitoshi Ishiguro
- Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, 210-0821, Japan.
| | - Takahiro Kondo
- Department of Materials Science, Institute of Pure and Applied Sciences, University of Tsukuba, Tsukuba 305-8573, Japan.
- The Advanced Institute for Materials Research, Tohoku University, Sendai, Miyagi 980-8577, Japan
- Hydrogen Boride Research Center and Tsukuba Research Center for Energy Materials Science, Institute of Pure and Applied Sciences and R&D Center for Zero CO2 Emission with Functional Materials, University of Tsukuba, Tsukuba 305-8573, Japan
| | - Masahiro Miyauchi
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Institute of Science Tokyo, Meguro, Tokyo, 152-8552, Japan.
| |
Collapse
|
35
|
Ito J, Strange A, Liu W, Joas G, Lytras S, Sato K. A protein language model for exploring viral fitness landscapes. Nat Commun 2025; 16:4236. [PMID: 40360496 PMCID: PMC12075601 DOI: 10.1038/s41467-025-59422-w] [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: 07/18/2024] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Successively emerging SARS-CoV-2 variants lead to repeated epidemic surges through escalated fitness (i.e., relative effective reproduction number between variants). Modeling the genotype-fitness relationship enables us to pinpoint the mutations boosting viral fitness and flag high-risk variants immediately after their detection. Here, we present CoVFit, a protein language model adapted from ESM-2, designed to predict variant fitness based solely on spike protein sequences. CoVFit was trained on genotype-fitness data derived from viral genome surveillance and functional mutation assays related to immune evasion. CoVFit successively ranked the fitness of unknown future variants harboring nearly 15 mutations with informative accuracy. CoVFit identified 959 fitness elevation events throughout SARS-CoV-2 evolution until late 2023. Furthermore, we show that CoVFit is applicable for predicting viral evolution through single amino acid mutations. Our study gives insight into the SARS-CoV-2 fitness landscape and provides a tool for efficiently identifying SARS-CoV-2 variants with higher epidemic risk.
Collapse
Affiliation(s)
- Jumpei Ito
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Adam Strange
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Wei Liu
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
- Swiss Institute of Bioinformatics, Geneva, Switzerland
| | - Gustav Joas
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Immunology and Respiratory Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Spyros Lytras
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Kei Sato
- Division of Systems Virology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK.
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Collaboration Unit for Infection, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
36
|
Gen R, Addetia A, Asarnow D, Park YJ, Quispe J, Chan MC, Brown JT, Lee J, Campbell MG, Lapointe CP, Veesler D. SARS-CoV-2 nsp1 mediates broad inhibition of translation in mammals. Cell Rep 2025; 44:115696. [PMID: 40359110 DOI: 10.1016/j.celrep.2025.115696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/13/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) non-structural protein 1 (nsp1) promotes innate immune evasion by inhibiting host translation in human cells. However, the role of nsp1 in other host species remains elusive, especially in bats-natural reservoirs of sarbecoviruses with a markedly different innate immune system than humans. We reveal that nsp1 potently inhibits translation in Rhinolophus lepidus bat cells, which belong to the same genus as known sarbecovirus reservoir hosts. We determined a cryoelectron microscopy structure of nsp1 bound to the R. lepidus 40S ribosomal subunit, showing that it blocks the mRNA entry channel by targeting a highly conserved site among mammals. Accordingly, we found that nsp1 blocked protein translation in mammalian cells from several species, underscoring its broadly inhibitory activity and conserved role in numerous SARS-CoV-2 hosts. Our findings illuminate the arms race between coronaviruses and mammalian host immunity, providing a foundation for understanding the determinants of viral maintenance in bat hosts and spillover.
Collapse
Affiliation(s)
- Risako Gen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Amin Addetia
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Daniel Asarnow
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Joel Quispe
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Matthew C Chan
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jack T Brown
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jimin Lee
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Melody G Campbell
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
37
|
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
|
38
|
Beissat K, Lattard V, Picard-Meyer E, Fafournoux A, Soro SD, Servat A, Vincent-Hubert F, Boué F, Chatron N, Monchâtre-Leroy E, Wasniewski M. Infectious potential and circulation of SARS-CoV-2 in wild rats. PLoS One 2025; 20:e0316882. [PMID: 40354427 PMCID: PMC12068656 DOI: 10.1371/journal.pone.0316882] [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: 01/06/2025] [Accepted: 04/08/2025] [Indexed: 05/14/2025] Open
Abstract
Since the beginning of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, a wide range of animal species (pets, mink…) have been naturally infected with this betacoronavirus. The emergence of new variants has increased the ability of SARS-CoV-2 to infect species that were not susceptible to the "original" SARS-CoV-2, such as mice and rats. This work attempted to evaluate the role of urban rats in the SARS-CoV-2 transmission by combining surveillance studies of rat populations in urban environments, in vivo experimental inoculation of SARS-CoV-2 and comparative viral-receptor interaction in silico analyses. We studied the circulation of SARS-CoV-2 in wild Rattus norvegicus (n = 401) captured in urban areas and sewage systems of several French cities. Except for 3 inconclusive samples (2/75 from Bordeaux and 1/261 from Lyon) none of the 353 sera tested showed anti-SARS-CoV-2 antibodies by microsphere immunoassay. However, the 3 inconclusive sera samples were negative by virus neutralisation assay. No SARS-CoV-2 viral RNA was detected in all lungs collected from the 401 captured urban brown rats. In complement, four rat groups (two wild-type colonies, Rattus norvegicus and Rattus rattus, and two laboratory strains, Sprague-Dawley and Wistar) were inoculated with the SARS-CoV-2 Omicron BA.5. At 4 days post-inoculation, no infectious viral particles were detected in the lungs and upper respiratory tract (URT) while viral RNA was detected at a low level only in the URT of all groups. In addition, seroconversion was observed 14 days after inoculation in the four groups. By molecular modelling, the Omicron BA.5 receptor binding domain (RBD) had lower affinities for Rattus norvegicus and Rattus rattus ACE2 than Homo sapiens ACE2. Based on these results the SARS-CoV-2 Omicron BA.5 was unable to infect laboratory and wild type rats. In addition, Rattus norvegicus collected for this study in different areas of France were not infected with SARS-CoV-2.
Collapse
Affiliation(s)
- Kevyn Beissat
- Nancy Laboratory for Rabies and Wildlife, ANSES, Malzéville, Nancy, France
- USC-1233 Rongeurs Sauvages Risques Sanitaires et Gestion des Populations (RS2GP), VetAgroSup, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Lyon University, Marcy-L’Etoile, France
| | - Virginie Lattard
- USC-1233 Rongeurs Sauvages Risques Sanitaires et Gestion des Populations (RS2GP), VetAgroSup, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Lyon University, Marcy-L’Etoile, France
| | | | - Ambre Fafournoux
- USC-1233 Rongeurs Sauvages Risques Sanitaires et Gestion des Populations (RS2GP), VetAgroSup, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Lyon University, Marcy-L’Etoile, France
| | - Sionfoungo Daouda Soro
- USC-1233 Rongeurs Sauvages Risques Sanitaires et Gestion des Populations (RS2GP), VetAgroSup, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Lyon University, Marcy-L’Etoile, France
| | - Alexandre Servat
- Nancy Laboratory for Rabies and Wildlife, ANSES, Malzéville, Nancy, France
| | - Françoise Vincent-Hubert
- IFREMER, Laboratoire Santé, Environnement et Microbiologie (LSEM) - MASAE, rue de l’île d’Yeu, , Nantes, France/ Groupement d’intérêt scientifique (GIS) Obépine, https://www.reseau-obepine.fr
| | - Franck Boué
- Nancy Laboratory for Rabies and Wildlife, ANSES, Malzéville, Nancy, France
| | - Nolan Chatron
- USC-1233 Rongeurs Sauvages Risques Sanitaires et Gestion des Populations (RS2GP), VetAgroSup, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Lyon University, Marcy-L’Etoile, France
| | | | - Marine Wasniewski
- Nancy Laboratory for Rabies and Wildlife, ANSES, Malzéville, Nancy, France
| |
Collapse
|
39
|
Kondo T, Suzuki R, Yajima H, Kawahara S, Yamaya K, Ichikawa T, Tsujino S, Suzuki S, Tamura T, Hashiguchi T, Fukuhara T. Determinants of susceptibility to SARS-CoV-2 infection in murine ACE2. J Virol 2025:e0054325. [PMID: 40353671 DOI: 10.1128/jvi.00543-25] [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: 03/28/2025] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) utilizes angiotensin-converting enzyme 2 (ACE2) as a receptor to enter host cells, and primary receptor recognition of the spike protein is a major determinant of the host range of SARS-CoV-2. Since the emergence of SARS-CoV-2, a considerable number of variants have emerged. However, the determinants of host tropism of SARS-CoV-2 remain elusive. We conducted infection assays with chimeric recombinant SARS-CoV-2 carrying the spike protein from 10 viral variants, assessing their entry efficiency using mammalian ACE2 orthologs from species that have close contact with humans. We found that only murine ACE2 exhibited different susceptibilities to infection with the SARS-CoV-2 variants. Moreover, we revealed that the mutation N501Y in the viral spike protein has a crucial role in determining the infectivity of cells expressing murine ACE2 and of mice in vivo. Next, we identified six amino acid substitutions at 24, 30, 31, 82, 83, and 353 in murine ACE2 that allowed for viral entry of the variants to which murine ACE2 was previously resistant. Furthermore, we showed that ACE2 from a species closely related to mice, Mus caroli, is capable of supporting entry of the viral variants that could not use murine ACE2. These results suggest that few ACE2 orthologs have different susceptibility to infection with SARS-CoV-2 variants as observed for murine ACE2. Collectively, our study reveals critical amino acids in ACE2 and the SARS-CoV-2 spike protein that are involved in the host tropism of SARS-CoV-2, shedding light on interspecies susceptibility to infection.IMPORTANCESARS-CoV-2 can infect many species besides humans, leading to the evolution of the virus and adaptation to other animal hosts, which could trigger a new COVID-19 wave. The SARS-CoV-2 spike protein utilizes ACE2 as a receptor for entry into host cells. The interaction of ACE2 with the spike protein determines the host range of SARS-CoV-2. In this study, using chimeric viruses carrying the spike protein of SARS-CoV-2 variants to infect cells expressing different ACE2 orthologs from species humans come in close contact with, we confirmed murine ACE2 alone showed different susceptibility to the variants. We identified residues in murine ACE2 and the viral spike that restrict viral entry. Furthermore, an ACE2 ortholog from a species genetically close to mice mediated entry of SARS-CoV-2 variants incapable of infecting mice. This research highlights the uniquely limited susceptibility of mice to different SARS-CoV-2 variants and provides invaluable insights into the host tropism of SARS-CoV-2.
Collapse
Affiliation(s)
- Takashi Kondo
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Rigel Suzuki
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development: IVReD, Hokkaido University, Sapporo, Japan
| | - Hisano Yajima
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Sachiho Kawahara
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Kodai Yamaya
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Takaya Ichikawa
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Shuhei Tsujino
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Saori Suzuki
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development: IVReD, Hokkaido University, Sapporo, Japan
| | - Tomokazu Tamura
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development: IVReD, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Takao Hashiguchi
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Kyoto University Immunomonitoring Center, Kyoto University, Kyoto, Japan
| | - Takasuke Fukuhara
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development: IVReD, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Department of Virology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- AMED-CREST, Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| |
Collapse
|
40
|
Wani MM, Cooper JM, Migliorini M, Strickland DK. The LDL receptor related protein 1 (LRP1) facilitates ACE2-mediated endocytosis of SARS-CoV2 spike protein-containing pseudovirions. J Biol Chem 2025:110227. [PMID: 40349772 DOI: 10.1016/j.jbc.2025.110227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 05/01/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, employs the viral spike (S) protein to associate with host cells. While angiotensin-converting enzyme 2 (ACE2) is a major receptor for the SARS-CoV-2 spike protein, evidence reveals that other cellular receptors may also contribute to viral entry. We interrogated the role of the low-density lipoprotein receptor-related protein 1 (LRP1) in the involvement of SARS-CoV-2 viral entry. Employing surface plasmon resonance studies, we demonstrated high affinity binding of the trimeric SARS-CoV-2 spike protein to purified LRP1. Further, we observed high affinity interaction of the SARS-CoV-2 spike protein with other low-density lipoprotein receptor (LDLR) family members as well, including LRP2 and the very low-density lipoprotein receptor (VLDLR). Binding of the SARS-CoV-2 spike protein to LRP1 was mediated by its receptor binding domain (RBD). Several LRP1 ligands require surface exposed lysine residues for their interaction with LRP1, and chemical modification of lysine residues on the RBD with sulfo-NHS-acetate ablated binding to LRP1. Using cellular model systems, we demonstrated that cells expressing LRP1, but not those lacking LRP1, rapidly internalized purified 125I-labeled S1 subunit of the SARS-CoV-2 spike protein. LRP1-mediated internalization of the 125I-labeled S1 subunit was enhanced in cells expressing ACE2. By employing pseudovirion particles containing a murine leukemia virus core and luciferase reporter that express the SARS-CoV-2 spike protein on their surface, we confirmed that LRP1 facilitates ACE2-mediated psuedovirion endocytosis. Together, these data implicate LRP1, and perhaps other LDLR family members as host factors for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Mashhood M Wani
- The Center for Vascular and Inflammatory Diseases, Departments of
| | - Joanna M Cooper
- The Center for Vascular and Inflammatory Diseases, Departments of; Physiology and
| | - Mary Migliorini
- The Center for Vascular and Inflammatory Diseases, Departments of
| | - Dudley K Strickland
- The Center for Vascular and Inflammatory Diseases, Departments of; Physiology and; Surgery, University of Maryland School of Medicine, Baltimore, MD 21201.
| |
Collapse
|
41
|
Pita Dos Santos LG, da Silva Coutinho G, Rodrigues Guimarães JM, Miranda da Silva M, Francisco da Silva A, Marcelino Neto PP, Coral Rodrigues BC, Aaron de Almeida W, Carlos Alves Dos Santos AJ, Napoleão TH, Pontual EV. Advances in COVID-19 Therapeutics: Exploring the role of lectins and protease inhibitors. Microb Pathog 2025; 205:107687. [PMID: 40349995 DOI: 10.1016/j.micpath.2025.107687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 05/01/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
The rapid global spread of SARS-CoV-2 has demanded innovative approaches to treatment and prevention. This article reviews the current landscape of COVID-19 therapeutics and vaccines, emphasizing the role of biotechnological products, particularly lectins and protease inhibitors. SARS-CoV-2, a single-stranded RNA virus, infects host cells via its spike (S) protein, which binds to the angiotensin-converting enzyme 2 (ACE2) receptor. This interaction is facilitated by host proteases like TMPRSS2, which are critical for viral entry. Treatments for COVID-19 primarily focus on antiviral drugs, anti-inflammatory agents, and monoclonal antibodies. Protease inhibitors that target viral enzymes like Mpro and PLpro have demonstrated potential. Additionally, vaccines, including mRNA-based, DNA-based, and those using viral vectors or inactivated viruses, are essential for preventing new infections. Lectins, proteins that bind specifically to carbohydrates, have emerged as potential antiviral agents. They can impede viral entry by binding to glycoproteins on the virus's surface or modulate immune responses. Studies indicate that lectins like cyanovirin-N and griffithsin exhibit significant antiviral activity against SARS-CoV-2. While most of the research on these biotechnological products is still in preclinical or early stages, their potential for treating and preventing COVID-19 is substantial. Further investigation and clinical trials are crucial to validate their efficacy and safety. This article underscores the need for continued exploration of novel therapeutic strategies to combat the evolving COVID-19 pandemic. However, the review is limited by the scarcity of clinical data on these products, highlighting the need for translational research.
Collapse
Affiliation(s)
| | - Gabriel da Silva Coutinho
- Departamento de Morfologia e Fisiologia Animal, Universidade Federal Rural de Pernambuco, Recife, Brazil
| | | | - Marcelo Miranda da Silva
- Departamento de Morfologia e Fisiologia Animal, Universidade Federal Rural de Pernambuco, Recife, Brazil
| | - Alex Francisco da Silva
- Departamento de Morfologia e Fisiologia Animal, Universidade Federal Rural de Pernambuco, Recife, Brazil
| | - Pedro Paulo Marcelino Neto
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Beto Cherles Coral Rodrigues
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Welton Aaron de Almeida
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Universidade de Pernambuco, Recife, Pernambuco, Brazil
| | | | - Thiago Henrique Napoleão
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Emmanuel Viana Pontual
- Departamento de Morfologia e Fisiologia Animal, Universidade Federal Rural de Pernambuco, Recife, Brazil.
| |
Collapse
|
42
|
Yin L, Zhang H, Shang Y, Wu S, Jin T. NLRP3 inflammasome: From drug target to drug discovery. Drug Discov Today 2025; 30:104375. [PMID: 40345614 DOI: 10.1016/j.drudis.2025.104375] [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: 12/02/2024] [Revised: 04/20/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
The immune system employs innate and adaptive immunity to combat pathogens and stress stimuli. Innate immunity rapidly detects pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) via pattern recognition receptors (PRRs), whereas adaptive immunity mediates antigen-specific T/B cell responses. The NLRP3 inflammasome, a key cytoplasmic PRR, consists of leucine-rich repeat, nucleotide-binding, and pyrin domains. Its activation requires priming (signal 1: Toll-like receptors/NOD-like receptors/cytokine receptors) and activation (signal 2: PAMPs/DAMPs/particulates). NLRP3 triggers cytokine storms and neuroinflammation, contributing to inflammatory diseases. Emerging therapies target NLRP3 via nuclear receptors (transcriptional regulation), adeno-associated virus (AAV) vectors (gene delivery), and microRNAs (post-transcriptional modulation). This review highlights NLRP3's signaling cascade, pathological roles, and combinatorial treatments leveraging nuclear receptors, AAVs, and microRNAs for immunomodulation.
Collapse
Affiliation(s)
- Ling Yin
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui 323000, China; College of Medicine, University of Florida, Gainesville, FL 32608, USA; Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Laboratory of Structural Immunology, Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027 China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| | - Hongliang Zhang
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui 323000, China
| | - Yuhua Shang
- Anhui Genebiol Biotech. Ltd., Hefei 230000, China
| | - Songquan Wu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui 323000, China.
| | - Tengchuan Jin
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui 323000, China; Anhui Genebiol Biotech. Ltd., Hefei 230000, China; Laboratory of Structural Immunology, Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027 China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science & Technology of China, Hefei 230027, China; Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
43
|
Hoque I, Singh N, Ghosh Dastidar U, Martin AK, Joshi A, Sardana Y, Singh Chawla R, Das N, Patra B, Devi R, Das S, Das D, Kumar S, Ringe RP, Bokara KK, Thakur KG, Talukdar A. Strategic Design and Optimization of Umifenovir Analogues: Balancing Antiviral Efficacy and hERG Toxicity against SARS-CoV-2. J Med Chem 2025; 68:9371-9406. [PMID: 40263709 DOI: 10.1021/acs.jmedchem.4c03093] [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: 04/24/2025]
Abstract
Arbidol (ARB, Umifenovir), a broad-spectrum antiviral from Russia, lacks Food and Drug Administration (FDA) approval due to insufficient clinical data and undocumented toxicity concerns. Its indole scaffold, with six unique substitutions, enables optimization for improved efficacy. This study optimized ARB's antiviral potency and safety by modifying the N1, C2, C3, and C4 positions. Antiviral efficacy was evaluated in SARS-CoV-2-infected VERO E6 cells, while optimization was guided by absorption, distribution, metabolism, and excretion (ADME), in vivo pharmacokinetic (PK) and hERG. Early modifications at N1 and C2 produced compounds 10 and 14 (IC50 = 1.5 μM), surpassing ARB (IC50 = 9.0 μM). Further refinements yielded compounds 42 (IC50 = 1.1 μM) and 56 (IC50 = 0.24 μM), resolving hERG toxicity (>30 μM). C3 modifications led to lead compounds 77, 79, and 81 (IC50 = 0.67-0.7 μM), achieving superior potency while eliminating hERG toxicity. Mechanism of entry inhibition and immunofluorescence confirmed compound 77 significantly reduced SARS-CoV-2 within Vero cells, supporting their preclinical potential.
Collapse
Affiliation(s)
- Israful Hoque
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, WB, India
| | - Nittu Singh
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
| | - Uddipta Ghosh Dastidar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, WB, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Alna Kuriyickal Martin
- CSIR-Center for Cellular and Molecular Biology, Medical Biotechnology Complex, Uppal Road, Hyderabad 500007, Telangana, India
| | - Akshay Joshi
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
| | - Yogesh Sardana
- CSIR-Center for Cellular and Molecular Biology, Medical Biotechnology Complex, Uppal Road, Hyderabad 500007, Telangana, India
| | - Ravneet Singh Chawla
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
| | - Nirmal Das
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, WB, India
| | - Binita Patra
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, WB, India
| | - Renuga Devi
- CSIR-Center for Cellular and Molecular Biology, Medical Biotechnology Complex, Uppal Road, Hyderabad 500007, Telangana, India
| | - Satyajeet Das
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
| | - Dipankar Das
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, WB, India
| | - Sahil Kumar
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
| | - Rajesh P Ringe
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
| | - Kiran Kumar Bokara
- CSIR-Center for Cellular and Molecular Biology, Medical Biotechnology Complex, Uppal Road, Hyderabad 500007, Telangana, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Krishan Gopal Thakur
- CSIR-Institute of Microbial Technology, Ministry of Science & Technology, Sector 39A Rd, 39A, Sector 39, 160036 Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Arindam Talukdar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, WB, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
44
|
Castellana E, Chiappetta MR. Agnostic Drugs: A New Paradigm in Pharmacological Therapy. Hosp Pharm 2025:00185787251340598. [PMID: 40352616 PMCID: PMC12061907 DOI: 10.1177/00185787251340598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Affiliation(s)
- Eleonora Castellana
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Piemonte, Italy
| | - Maria Rachele Chiappetta
- Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Torino, Piemonte, Italy
| |
Collapse
|
45
|
Chen B, Farzan M, Choe H. SARS-CoV-2 spike protein: structure, viral entry and variants. Nat Rev Microbiol 2025:10.1038/s41579-025-01185-8. [PMID: 40328900 DOI: 10.1038/s41579-025-01185-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 05/08/2025]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been a devastating global pandemic for 4 years and is now an endemic disease. With the emergence of new viral variants, COVID-19 is a continuing threat to public health despite the wide availability of vaccines. The virus-encoded trimeric spike protein (S protein) mediates SARS-CoV-2 entry into host cells and also induces strong immune responses, making it an important target for development of therapeutics and vaccines. In this Review, we summarize our latest understanding of the structure and function of the SARS-CoV-2 S protein, the molecular mechanism of viral entry and the emergence of new variants, and we discuss their implications for development of S protein-related intervention strategies.
Collapse
Affiliation(s)
- Bing Chen
- Division of Molecular Medicine, Boston Children's Hospital, and Department of Paediatrics, Harvard Medical School, Boston, MA, USA.
| | - Michael Farzan
- Division of Infectious Diseases, Boston Children's Hospital, and Department of Paediatrics, Harvard Medical School, Boston, MA, USA.
- Center for Integrated Solutions for Infectious Diseases (CISID), The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Hyeryun Choe
- Division of Infectious Diseases, Boston Children's Hospital, and Department of Paediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Resendes Barbosa I, Alves Amorim M, de Souza Oliveira VH, André E, Pereira Guedes G, Augusto Chaves O, Serpa C, Fintelman-Rodrigues N, Sacramento CQ, Moreno L Souza T, Sant'Anna CMR, Echevarria A. Novel Sulfonamide-Sydnone Hybrids: Complementary Insight into Anti-Inflammatory Action, Anti-SARS-CoV-2 Activity, Human Serum Albumin Interaction, and in silico Analysis. ChemMedChem 2025; 20:e202400697. [PMID: 39988470 DOI: 10.1002/cmdc.202400697] [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: 09/03/2024] [Revised: 02/10/2025] [Indexed: 02/25/2025]
Abstract
Acute lung injury (ALI) is a severe condition often seen in intensive care unit patients. Due to limited treatment options, ALI is linked to high rates of mortality and morbidity. Bacterial and viral infections are significant contributors to ALI. For instance, severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection can lead to a strong inflammatory response that may progress to ALI, a leading cause of death in COVID-19 cases. Prior research has demonstrated that sulfonamides and sydnones exhibit anti-inflammatory and antiviral properties, which has led us to develop compounds containing both scaffolds. Most of the new sulfonamide-sydnone hybrids are expected to be orally bioavailable based on in silico ADME predictions. They effectively suppressed the development of ALI in lipopolysaccharide (LPS)-challenged mice and inhibited viral replication in Calu-3 cells, with minimal cytotoxicity in non-infected Calu-3 and Vero E6 cells. Molecular docking investigations indicated some possible viral targets for the action of the sydnones, highlighting the possible interaction with non-structural proteins of SARS-CoV-2. Additionally, combined experimental and theoretical studies indicated that the new compounds can strongly interact with human serum albumin, suggesting a possible extended residence time in the human bloodstream.
Collapse
Affiliation(s)
- Igor Resendes Barbosa
- Institute of Chemistry, Federal Rural University of Rio de Janeiro, Seropédica, 23898-56, Rio de Janeiro, Brazil
| | - Mayara Alves Amorim
- Department of Pharmacology, Federal University of Paraná, Curitiba, 81531-980, PR, Brazil
| | | | - Eunice André
- Department of Pharmacology, Federal University of Paraná, Curitiba, 81531-980, PR, Brazil
| | - Guilherme Pereira Guedes
- Institute of Chemistry, Fluminense Federal University, Niterói, 24020-141, Rio de Janeiro, Brazil
| | - Otávio Augusto Chaves
- Department of Chemistry, Coimbra Chemistry Centre - Institute of Molecular Science (CQC-IMS), University of Coimbra, Rua Larga, 3004-535, Coimbra, Portugal
- Laboratory of Immunopharmacology, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias (CPIV), Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), 21040-361, Rio de Janeiro, RJ, Brazil
| | - Carlos Serpa
- Department of Chemistry, Coimbra Chemistry Centre - Institute of Molecular Science (CQC-IMS), University of Coimbra, Rua Larga, 3004-535, Coimbra, Portugal
| | - Natalia Fintelman-Rodrigues
- Laboratory of Immunopharmacology, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias (CPIV), Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), 21040-361, Rio de Janeiro, RJ, Brazil
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, 21040-900, Brazil
| | - Carolina Q Sacramento
- Laboratory of Immunopharmacology, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias (CPIV), Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), 21040-361, Rio de Janeiro, RJ, Brazil
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, 21040-900, Brazil
| | - Thiago Moreno L Souza
- Laboratory of Immunopharmacology, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias (CPIV), Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), 21040-361, Rio de Janeiro, RJ, Brazil
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, 21040-900, Brazil
| | - Carlos Mauricio R Sant'Anna
- Institute of Chemistry, Federal Rural University of Rio de Janeiro, Seropédica, 23898-56, Rio de Janeiro, Brazil
| | - Aurea Echevarria
- Institute of Chemistry, Federal Rural University of Rio de Janeiro, Seropédica, 23898-56, Rio de Janeiro, Brazil
| |
Collapse
|
47
|
Yang Q, Huang X, Zhang H, Sun J, Tang J, Chen Z, Liu L, Liu M, Sun Z, Tang Z, Wei D, Wang D, Wang Y, Yan M, Zhao L, Zhu A, Zhong Y, Yang H, Zhao Y, Dai J, Shi Y, Huang B, Zhang W, Zhao J, Chen X, Rao Z, Peng W. Expanding the utilization of binding pockets proves to be effective for noncovalent small molecule inhibitors against SARS-CoV-2 M pro. Eur J Med Chem 2025; 289:117497. [PMID: 40090296 DOI: 10.1016/j.ejmech.2025.117497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/27/2025] [Accepted: 03/08/2025] [Indexed: 03/18/2025]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in millions of deaths and continues to pose serious threats to global public health. The main protease (Mpro) of SARS-CoV-2 is crucial for viral replication and its conservation, making it an attractive drug target. Here, we employed a structure-based drug design strategy to develop and optimize novel inhibitors targeting SARS-CoV-2 Mpro. By fully exploring occupation of the S1, S2, and S3/S4 binding pockets, we identified eight promising inhibitors with half-maximal inhibitory concentration (IC50) values below 20 nM. The cocrystal structure of Mpro with compound 10 highlighted the crucial roles of the interactions within the S3/S4 pockets in inhibitor potency enhancement. These findings demonstrated that expanding the utilization of these binding pockets was an effective strategy for developing noncovalent small molecule inhibitors that target SARS-CoV-2 Mpro. Compound 4 demonstrated outstanding in vitro antiviral activity against wild-type SARS-CoV-2 with an EC50 of 9.4 nM. Moreover, oral treatment with compounds 1 and 9 exhibited excellent antiviral potency and substantially ameliorated virus-induced tissue damage in the lungs of Omicron BA.5-infected K18-human ACE2 (K18-hACE2) transgenic mice, indicating that these novel noncovalent inhibitors could be potential oral agents for the treatment of COVID-19.
Collapse
Affiliation(s)
- Qi Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Xupeng Huang
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Hongbo Zhang
- Beijing StoneWise Technology Co. Ltd., Beijing, 100080, China
| | - Jing Sun
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Jielin Tang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Zhao Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Lijie Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; Guangzhou Medical University, Guangzhou, 511436, China
| | - Man Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Zeyun Sun
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhenhao Tang
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Dandan Wei
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Dong Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yiliang Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Mengrong Yan
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Li Zhao
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; Guangzhou Medical University, Guangzhou, 511436, China
| | - Airu Zhu
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yihang Zhong
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Haitao Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yao Zhao
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, 518112, China
| | - Jun Dai
- Technology Centre, Guangzhou Customs, Guangzhou, 510623, China
| | - Yongxia Shi
- Technology Centre, Guangzhou Customs, Guangzhou, 510623, China
| | - Bo Huang
- Beijing StoneWise Technology Co. Ltd., Beijing, 100080, China.
| | - Wei Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xinwen Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China.
| | - Zihe Rao
- Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; Laboratory of Structural Biology, School of Life Sciences and School of Medicine, Tsinghua University, Beijing, 100084, China; Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Wei Peng
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China; Guangzhou National Laboratory, Guangzhou, 510005, China; Innovative Center for Pathogen Research, Guangzhou National Laboratory, Guangzhou, 510005, China; The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China; University of South China, Hengyang, 421001, China.
| |
Collapse
|
48
|
Li M, Gu X, Yang J, Zhang C, Zhou Y, Huang P, Wang X, Zhang L, Jiang L, Zhai L, Yu M, Cheng G, Yang L. Luteolin: A potential therapeutic agent for respiratory diseases. Eur J Pharmacol 2025; 999:177699. [PMID: 40324574 DOI: 10.1016/j.ejphar.2025.177699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/10/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Acute lung injury, COVID-19, lung cancer, and asthma are a few of the respiratory conditions that are the main causes of morbidity and mortality worldwide. The increasing incidence and mortality rates have attracted significant attention to the prevention and treatment of these conditions. In recent years, there has been a renewed interest in utilizing naturally derived compounds as therapeutic agents for respiratory diseases. Luteolin (Lut), a flavonoid compound, possesses an extensive range of pharmacological characteristics, encompassing anti-inflammatory, antioxidative, antineoplastic, and antimicrobial activities. However, a comprehensive summary of Lut's therapeutic effects and mechanisms in respiratory diseases remains lacking. This review examines the physicochemical properties, toxicity, and avenues of Lut's action in respiratory ailments. Lut exerts therapeutic effects through pathways such as nuclear factor kappa-B (NF-κB), nuclear factor erythroid 2-related factor 2 (Nrf2), mitogen-activated protein kinase (MAPK), janus kinase 1 (JAK1)/signal transducer and activator of transcription 3 (STAT3), phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT), and pyroptosis, modulating key processes such as the suppression of inflammatory mediators, attenuation of oxidative assault, and induction of apoptosis in lung cancer cells. This review strives to provide critical realizations into respiratory disease therapeutics and contribute to the foundation for drug development.
Collapse
Affiliation(s)
- Meng Li
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xinru Gu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jiaming Yang
- Department of Anatomy, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Ce Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yi Zhou
- Department of Anatomy, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Peifeng Huang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xuezhen Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lulu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Liping Jiang
- Department of Parasitology, Xiangya School of Basic Medical Sciences, Central South University, Changsha, Hunan, 410013, China
| | - Lidong Zhai
- Department of Anatomy, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Mingyu Yu
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, China; Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, 518132, China; Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen, 518000, China; Southwest United Graduate School, Kunming, 650504, China.
| | - Long Yang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; School of Public Health, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
49
|
Vanderkamp SG, Niazy M, Stegelmeier AA, Stinson KJ, Ricker N, Bridle BW. Cytokine, chemokine, and acute-phase protein profiles in plasma as correlative biomarkers of clinical outcomes for patients with COVID-19. Sci Rep 2025; 15:15397. [PMID: 40316702 PMCID: PMC12048561 DOI: 10.1038/s41598-025-99248-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 04/17/2025] [Indexed: 05/04/2025] Open
Abstract
Coronavirus disease identified in 2019 (COVID-19), caused by severe acute respiratory syndrome-coronavirus-2, had a global impact on human health and the economy. The aim of this study was to quantify cytokines, chemokines, and acute phase proteins in the plasma of patients with COVID-19 to elucidate potential biomarkers to inform prognostic and treatment decisions. Clustering analysis using the K-prototypes method identified underlying biological patterns in patients with COVID-19. The penalized multinomial logistic regression analysis identified two comorbidities (hypertension, congestive heart failure) and thirteen analytes as potential risk factors for COVID-19 progression with 88.2% accuracy. Based on a patient's age, high concentrations of interleukin (IL)-6, monocyte chemoattractant protein-1, and pentraxin 3 were important biomarkers for lethal COVID-19. Decreased concentrations of interferon gamma-induced protein-10, IL-10, and soluble tumor necrosis factor receptor I were found to be associated with mild COVID-19, while increasing concentrations of these analytes could be used to predict COVID-19 severity.
Collapse
Affiliation(s)
- Sierra G Vanderkamp
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Maysa Niazy
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Ashley A Stegelmeier
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | | | - Nicole Ricker
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
50
|
Imtiaz A, Akkam AY, AlThumali LH, AlHarthi AM, Porte HG, AlOsaimi WS, AlAsmari MMA, Salih RBG, Mitra B. Diagnostic accuracy of visual triage checklist in early recognition of COVID-19 cases in the pediatric population: A retrospective cohort study. BMC Pediatr 2025; 25:352. [PMID: 40317001 PMCID: PMC12046900 DOI: 10.1186/s12887-025-05693-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/16/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND During the coronavirus disease 2019 (COVID-19) pandemic, healthcare facilities developed surveillance systems to identify patients suspected of having COVID-19 to segregate them during their hospital stay. As a part of this infection control strategy, the Ministry of Health in the Kingdom of Saudi Arabia developed a visual triage (VT) checklist for early screening and isolation of patients in the hospital. The aim of this study was to evaluate the diagnostic accuracy of this visual triage checklist in identifying children with COVID-19. METHODS This was a retrospective, single center study that included all children who were tested for COVID-19 and were admitted to the hospital through the pediatric emergency department. The diagnostic accuracy of the visual triage checklist was assessed using COVID-19 PCR as the gold standard. RESULTS A total of 1333 patients were included. The visual triage checklist had a sensitivity of 94.3% (95% CI: 87.2-98.1) and a specificity of 16.0% (95% CI: 14-18) with an area under the receiver operating characteristic curve of 0.55 (0.53-0.58). The positive predictive value of the checklist was low at 7.35% (95% CI: 5.9-9.0). CONCLUSION The VT checklist has high sensitivity, and is therefore potentially useful as an initial screening tool. However, the diagnosis of COVID-19 requires early secondary confirmation to avoid the large number of false positive cases associated with this tool.
Collapse
Affiliation(s)
- Ayesha Imtiaz
- Pediatric Emergency Department, King Saud Medical City, Riyadh, Kingdom of Saudi Arabia.
| | - Abdullah Y Akkam
- Pediatric Emergency Department, King Saud Medical City, Riyadh, Kingdom of Saudi Arabia
| | | | | | - Honey G Porte
- Pediatric Emergency Department, King Saud Medical City, Riyadh, Kingdom of Saudi Arabia
| | | | - Maha M A AlAsmari
- Prince Sultan Military Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Rawda B G Salih
- Pediatric Emergency Department, King Saud Medical City, Riyadh, Kingdom of Saudi Arabia
| | - Biswadev Mitra
- School of Public Health & Preventive Medicine, Monash University, 553 St Kilda Rd, VIC, Melbourne, Australia
- Emergency & Trauma Centre, The Alfred Hospital, 55 Commercial Road, Melbourne, Australia
| |
Collapse
|