1
|
Park K, Shin M, Natasha A, Kim J, Noh J, Kim SG, Kim B, Park J, Seo YR, Cho HK, Byun KS, Kim JH, Lee YS, Shim JO, Kim WK, Song JW. Novel human coronavirus in an infant patient with pneumonia, Republic of Korea. Emerg Microbes Infect 2025; 14:2466705. [PMID: 39945663 PMCID: PMC11849027 DOI: 10.1080/22221751.2025.2466705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/06/2025] [Accepted: 02/09/2025] [Indexed: 02/22/2025]
Abstract
Coronaviruses (CoVs) pose a significant threat to public health, causing a wide spectrum of clinical manifestations and outcomes. Beyond precipitating global outbreaks, Human CoVs (HCoVs) are frequently found among patients with respiratory infections. To date, limited attention has been directed towards alphacoronaviruses due to their low prevalence and fatality rates. Nasal swab and serum samples were collected from a paediatric patient, and an epidemiological survey was conducted. Retrospective surveillance investigated the molecular prevalence of CoV in 880 rodents collected in the Republic of Korea (ROK) from 2018 to 2022. Next-generation sequencing (NGS) and phylogenetic analyses characterized the novel HCoV and closely related CoVs harboured by Apodemus spp. On 15 December 2022, a 103-day-old infant was admitted with fever, cough, sputum production, and rhinorrhea, diagnosed with human parainfluenza virus 1 (HPIV-1) and rhinovirus co-infection. Elevated AST/ALT levels indicated transient liver dysfunction on the fourth day of hospitalization. Metagenomic NGS (mNGS) identified a novel HCoV in nasal swab and serum samples. Retrospective rodent surveillance and phylogenetic analyses showed the novel HCoV was closely related to alphacoronaviruses carried by Apodemus spp. in the ROK and China. This case highlights the potential of mNGS to identify emerging pathogens and raises awareness of possible extra-respiratory manifestations, such as transient liver dysfunction, associated with novel HCoVs. While the liver injury in this case may be attributable to the novel HCoV, further research is necessary to elucidate its clinical significance, epidemiological prevalence, and zoonotic origins.
Collapse
Affiliation(s)
- Kyungmin Park
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- Institute for Viral Diseases, Korea University College of Medicine, Seoul, Republic of Korea
| | - Minsoo Shin
- Department of Paediatrics, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Augustine Natasha
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jongwoo Kim
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Juyoung Noh
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seong-Gyu Kim
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Bohyeon Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jieun Park
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Ye-rin Seo
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hee-Kyung Cho
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kwan Soo Byun
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Korea University Medical Center, Seoul, Republic of Korea
| | - Ji Hoon Kim
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Korea University Medical Center, Seoul, Republic of Korea
| | - Young-Sun Lee
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Korea University Medical Center, Seoul, Republic of Korea
| | - Jung Ok Shim
- Department of Paediatrics, Korea University College of Medicine, Seoul, Republic of Korea
| | - Won-Keun Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
- Institute of Medical Research, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jin-Won Song
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Subramaniyan B, Falcon EC, Moore AR, Larabee JL, Nimmo SL, Berrios-Rivera JL, Reddig WJ, Blewett EL, Papin JF, Walters MS, Burgett AWG. Anti-SARS-CoV-2 Small Molecule Targeting of Oxysterol-Binding Protein (OSBP) Activates Cellular Antiviral Innate Immunity. ACS Infect Dis 2025; 11:1064-1077. [PMID: 40255103 PMCID: PMC12070403 DOI: 10.1021/acsinfecdis.4c00631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 03/11/2025] [Accepted: 04/04/2025] [Indexed: 04/22/2025]
Abstract
Human oxysterol-binding protein (OSBP) is a potentially druggable mediator in the replication of a broad spectrum of positive-sense (+) single-stranded RNA (ssRNA) viruses, including members of the Picornaviridae, Flaviviridae, and Coronaviridae. OSBP is a cytoplasmic lipid transporting protein capable of moving cholesterol and phosphoinositides between the endoplasmic reticulum (ER) and Golgi, and the ER and lysosome. Several structurally diverse antiviral compounds have been reported to function through targeting OSBP, including the natural product compound OSW-1. Our prior work shows that transient OSW-1 treatment induces a reduction in OSBP protein levels over multiple successive cell generations (i.e., multigenerational), with no apparent cellular toxicity, and the OSW-1-induced reduction of OSBP has antiviral activity against multiple (+)ssRNA viruses. This study extends these findings and establishes that OSW-1 has in vitro antiviral activity against multiple pathogenic (+)ssRNA viruses, including human rhinovirus (HRV1B), the feline coronavirus peritonitis virus (FIPV), human coronavirus 229E (HCoV-229E), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We also demonstrate that OSW-1 treatment in human airway epithelial cells alters the expression of multiple antiviral innate immune mediators, including the interferon (IFN) related genes IFNB1, IFNL3, CXCL10, ISG15, and MX1. Furthermore, OSW-1 enhances the induction of specific components of type I and III IFN antiviral responses triggered by the RNA viral mimetic polyinosinic-polycytidylic acid (Poly IC). In summary, this study further demonstrates the importance of OSBP in (+)ssRNA virus replication and presents OSBP as a potential regulator of cellular antiviral innate immune responses.
Collapse
Affiliation(s)
- Bharathiraja Subramaniyan
- Department
of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Emily C. Falcon
- Department
of Pharmaceutical Sciences, University of
Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Andrew R. Moore
- Department
of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Jason L. Larabee
- Department
of Microbiology and Immunology, University
of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Susan L. Nimmo
- Department
of Pharmaceutical Sciences, University of
Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Jorge L. Berrios-Rivera
- Department
of Pharmaceutical Sciences, University of
Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - William J. Reddig
- Department
of Biochemistry and Microbiology, Oklahoma
State University Center for Health Sciences, Tulsa, Oklahoma 74107, United States
| | - Earl L. Blewett
- Department
of Biochemistry and Microbiology, Oklahoma
State University Center for Health Sciences, Tulsa, Oklahoma 74107, United States
| | - James F. Papin
- Department
of Pathology, Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma 73104, United States
| | - Matthew S. Walters
- Department
of Medicine, Section of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73104, United States
| | - Anthony W. G. Burgett
- Department
of Pharmaceutical Sciences, University of
Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
- Stephenson
Cancer Center, University of Oklahoma Health
Sciences Center, Oklahoma
City, Oklahoma 73104, United States
| |
Collapse
|
3
|
Yang Y, Li D, Nie J, Wang J, Huang H, Hang X. A Nomogram for Predicting Survival in Patients with SARS-CoV-2 Omicron Variant Pneumonia Based on Admission Data. Infect Drug Resist 2025; 18:2093-2104. [PMID: 40303607 PMCID: PMC12039831 DOI: 10.2147/idr.s509178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/15/2025] [Indexed: 05/02/2025] Open
Abstract
Purpose Patients with severe SARS-CoV-2 omicron variant pneumonia pose a serious challenge. This study aimed to develop a nomogram for predicting survival using chest computed tomography (CT) imaging features and laboratory test results based on admission data. Patients and Methods A total of 436 patients with SARS-CoV-2 pneumonia (323 and 113 in the training and validation groups, respectively) were enrolled. Pneumonitis volume, assessed on chest CT scans at admission, was used to identify low- and high-risk groups. Risk analysis was performed using clinical symptoms, laboratory findings, and chest CT imaging features. A predictive algorithm was developed using Cox multivariate analysis. Results The high-risk group had a shorter survival duration than the low-risk group. Significant differences in mortality rate, neutrophil and lymphocyte counts, C-reactive protein (CRP) concentration, and urea nitrogen level were observed between the two groups. In the training group, age, pneumonia volume, total bilirubin, and blood urea nitrogen were independent prognostic factors. In the validation group, age, pneumonia volume, neutrophil count, and CRP were independent prognostic factors. A personalized prediction model for survival outcomes was developed using independent predictors. Conclusion A personalized prediction model was created to forecast the 5-, 10-, 15-, 20-, and 30-day survival rates of patients with COVID-19 omicron variant pneumonia based on admission data, and can be used to determine the survival rate and early treatment of severe patients.
Collapse
Affiliation(s)
- Yinghao Yang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
- Department of Infectious Diseases, the 988th Hospital of the Joint Logistic Support Force, Zhengzhou, People’s Republic of China
| | - Dong Li
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
- Department of Gastroenterology, The 971th Hospital of PLA Navy, Qingdao, People’s Republic of China
| | - Jinqiu Nie
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Junxue Wang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Huili Huang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Xiaofeng Hang
- Department of Infectious Diseases, Changzheng Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
4
|
Wideman SK, Wali L, Kovtunyk V, Chou S, Gusel V, Telimaa H, Najmi C, Stoeva D, Stöckl J, Gualdoni GA, Gorki AD, Radivojev S. Nebulized 2-deoxylated glucose analogues inhibit respiratory viral infection in advanced in vitro airway models. Sci Rep 2025; 15:9515. [PMID: 40108297 PMCID: PMC11923073 DOI: 10.1038/s41598-025-94476-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Respiratory viral infections, such as those caused by rhinoviruses (RVs) and human corona viruses (HCoV), result in a serious strain on healthcare systems and public health, underscoring an urgent need for inhaled broad-spectrum antiviral therapies. However, their development is challenging, as no standardized in vitro methodologies that can fully replicate the in vivo environment have been established. In this work, we aimed to investigate the antiviral and anti-inflammatory effect of three 2-deoxylated glucose analogues (2-DGA): 2-deoxy-D-glucose, 2-fluoro-2-deoxy-D-glucose and 2-fluoro-2-deoxy-D-mannose (2-FDM), by utilizing advanced in vitro air-liquid interface (ALI) airway models. We demonstrated that commonly used ALI models have variable susceptibility to RV, HCoV and influenza A virus (IAV) infection. Further, we showed that 2-DGA have an anti-inflammatory effect and suppress respiratory viral replication in models mimicking the upper and lower respiratory airways. Moreover, we confirmed that 2-DGA can be delivered via nebulization in vitro, highlighting their potential to be used as broad-spectrum inhaled antivirals. Finally, our results demonstrate the importance of incorporating complex in vitro methodologies, such as primary cell ALI cultures and aerosol exposure, at an early stage of drug development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Johannes Stöckl
- Institute of Immunology, Center of Pathophysiology, Immunology & Infectiology, Medical University of Vienna, Vienna, Austria
| | | | | | | |
Collapse
|
5
|
Naghibosadat M, Babuadze GG, Pei Y, Hurst J, Salvant E, Gaete K, Biondi M, Moloo B, Goldstein A, Avery S, Ma K, Pietraszek A, Wootton SK, Alhaboub A, Martin B, Mubareka S, Corredor J, Sultana A, Adeekoa A, Budylowski P, Ostrowski M, Chao J, Nagy E, Kozak R. Vaccination against SARS-CoV-2 provides low-level cross-protection against common cold coronaviruses in mouse and non-human primate animal models. J Virol 2025; 99:e0139024. [PMID: 39817773 PMCID: PMC11853048 DOI: 10.1128/jvi.01390-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/04/2024] [Indexed: 01/18/2025] Open
Abstract
The common cold coronaviruses are a source of ongoing morbidity and mortality particularly among elderly and immunocompromised individuals. While cross-reactive immune responses against multiple coronaviruses have been described following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and vaccination, it remains unclear if these confer any degree of cross-protection against the common cold coronaviruses. A recombinant fowl adenovirus vaccine expressing the SARS-CoV-2 spike protein (FAdV-9-S19) was generated, and protection from SARS-CoV-2 challenge was shown in K18-hACE2 mice. Vaccinated mice were also challenged with the common cold coronaviruses human coronavirus (HCoV)-OC43 and HCoV-NL63 by the intranasal route, and viral shedding and lung burden were reduced in these groups compared to unvaccinated animals. Histopathological analysis of lung tissues revealed significantly less inflammation and lower pathology scores in mice that received FAdV-9-S19 . Because no mouse model for the coronavirus HCoV-229E exists, we vaccinated and challenged cynomolgus macaques to evaluate cross-protection against HCoV-229E. Animals were monitored for clinical signs of disease and viral shedding. Infectious virus was detected in both groups throughout the course of infection; however, vaccinated animals showed reduced viral shedding at multiple time points after infection. Histopathological analysis of lung tissues following challenge also indicated a more moderate disease in the vaccinated animals. Therefore, vaccination with FAdV-9-S19 also provided a moderate cross-protection against HCoV-229E disease in the cynomolgus macaques infection model. Our study demonstrates that vaccination with a recombinant fowl adenovirus expressing SARS-CoV-2 spike protein can provide a low-level cross-protection against beta- and alphacoronaviruses. These findings are important for the design of future pan-coronavirus vaccines.IMPORTANCEThe common cold coronaviruses are a source of ongoing morbidity and mortality particularly among elderly and immunocompromised individuals, and no vaccine is currently available. Cross-reactive immune responses have been described following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination; however, it remains unclear what degree of cross-protection they confer against the common cold coronaviruses. We demonstrate that both humoral and cell-mediated immune responses provide a low-level cross-protection, resulting in reduced viral load and pathology for the common cold coronaviruses OC43 and NL63 in mouse models. Additionally, we present a novel non-human primate (NHP) model of infection with the common cold coronavirus 229E, demonstrating that it mimics the disease observed in humans and can serve as a model for future vaccine studies, as cross-protection was also observed. This is significant as it suggests that current vaccines could provide a low-level protection against other coronaviruses and could serve as part of vaccination strategy against future novel coronaviruses.
Collapse
MESH Headings
- Animals
- Mice
- Cross Protection/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Disease Models, Animal
- COVID-19 Vaccines/immunology
- SARS-CoV-2/immunology
- COVID-19/prevention & control
- COVID-19/immunology
- Vaccination
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Virus Shedding
- Lung/pathology
- Lung/virology
- Lung/immunology
- Coronavirus OC43, Human/immunology
- Female
- Humans
- Common Cold/prevention & control
- Common Cold/immunology
- Common Cold/virology
- Coronavirus NL63, Human/immunology
- Vaccines, Synthetic/immunology
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Cross Reactions
- Macaca fascicularis
Collapse
Affiliation(s)
- Maedeh Naghibosadat
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | | | - Yanlong Pei
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Jacklyn Hurst
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Elsa Salvant
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Kayla Gaete
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Mia Biondi
- School of Nursing, York University, Toronto, Ontario, Canada
| | - Badru Moloo
- University Health Network, Toronto, Ontario, Canada
| | | | - Stacey Avery
- University Health Network, Toronto, Ontario, Canada
| | - Kathleen Ma
- University Health Network, Toronto, Ontario, Canada
| | | | - Sarah K. Wootton
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Assad Alhaboub
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Benjamin Martin
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Samira Mubareka
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Juan Corredor
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Azmiri Sultana
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Adebayo Adeekoa
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Patrick Budylowski
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Mario Ostrowski
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- St. Michael's Hospital, Unity Health, Toronto, Ontario, Canada
| | - Jesse Chao
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Eva Nagy
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Robert Kozak
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Heffner AL, Rouault TA. A Comparison of Conserved Features in the Human Coronavirus Family Shows That Studies of Viruses Less Pathogenic than SARS-CoV-2, Such as HCoV-OC43, Are Good Model Systems for Elucidating Basic Mechanisms of Infection and Replication in Standard Laboratories. Viruses 2025; 17:256. [PMID: 40007010 PMCID: PMC11860170 DOI: 10.3390/v17020256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
In 2021, at the height of the COVID-19 pandemic, coronavirus research spiked, with over 83,000 original research articles related to the word "coronavirus" added to the online resource PubMed. Just 2 years later, in 2023, only 30,900 original research articles related to the word "coronavirus" were added. While, irrefutably, the funding of coronavirus research drastically decreased, a possible explanation for the decrease in interest in coronavirus research is that projects on SARS-CoV-2, the causative agent of COVID-19, halted due to the challenge of establishing a good cellular or animal model system. Most laboratories do not have the capabilities to culture SARS-CoV-2 'in house' as this requires a Biosafety Level (BSL) 3 laboratory. Until recently, BSL 2 laboratory research on endemic coronaviruses was arduous due to the low cytopathic effect in isolated cell culture infection models and the lack of means to quantify viral loads. The purpose of this review article is to compare the human coronaviruses and provide an assessment of the latest techniques that use the endemic coronaviruses-HCoV-229E, HCoV-OC43, HCoV-NL63, and HCoV-HKU1-as lower-biosafety-risk models for the more pathogenic coronaviruses-SARS-CoV-2, SARS-CoV, and MERS-CoV.
Collapse
Affiliation(s)
- Audrey L. Heffner
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Tracey A. Rouault
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Hnasko RM, Lin AV, McGarvey JA, Jackson ES. Immunoassay Detection of SARS-CoV-2 Using Monoclonal Antibody Binding to Viral Nucleocapsid Protein. Microb Biotechnol 2025; 18:e70117. [PMID: 39989430 PMCID: PMC11848557 DOI: 10.1111/1751-7915.70117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/25/2025] Open
Abstract
Immunoassays represent sensitive, easy-to-use, and cost-effective tests useful for the detection of the SARS-CoV-2 virus. In this manuscript, we report on the binding specificity of a pair of novel monoclonal antibodies (MAbs) generated against the SARS-CoV-2 nucleocapsid protein (NP) and their development into sensitive sandwich enzyme-linked immunosorbent assays (sELISA) and a lateral flow immunoassay (LFIA). Binding of these MAbs to hCoVs is limited to variants of SARS-CoV-2 and SARS-CoV NP. Chemiluminescent and absorbance spectroscopy sELISAs report a limit of detection (LOD) for the SARS-CoV-2 B.1.1.529 NP variant at 15 pg/mL, and the LFIA using a red-dyed 200 nm particle at 10 ng/mL. The sELISA exhibits broad SARS-CoV-2 viral variant detection with assay LOD for SARS-CoV-2 B.1.1.529 virus at 1.4 × 105 genome copies per mL (p ≤ 0.001). The availability of these MAbs should facilitate continued investment in the commercial development of immunoassays to increase global SARS-CoV-2 detection technologies.
Collapse
Affiliation(s)
- Robert M. Hnasko
- United States Department of Agriculture, Agricultural Research ServiceProduce Safety and Microbiology UnitAlbanyCaliforniaUSA
| | - Alice V. Lin
- United States Department of Agriculture, Agricultural Research ServiceProduce Safety and Microbiology UnitAlbanyCaliforniaUSA
| | - Jeffery A. McGarvey
- United States Department of Agriculture, Agricultural Research ServiceFoodborne Toxin Detection and Prevention UnitAlbanyCaliforniaUSA
| | - Eric S. Jackson
- United States Department of Agriculture, Agricultural Research ServiceFoodborne Toxin Detection and Prevention UnitAlbanyCaliforniaUSA
| |
Collapse
|
8
|
Greene KS, Choi A, Yang N, Chen M, Li R, Qiu Y, Ezzatpour S, Rojas KS, Shen J, Wilson KF, Katt WP, Aguilar HC, Lukey MJ, Whittaker GR, Cerione RA. Glutamine metabolism is essential for coronavirus replication in host cells and in mice. J Biol Chem 2025; 301:108063. [PMID: 39662828 PMCID: PMC11750454 DOI: 10.1016/j.jbc.2024.108063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 11/21/2024] [Accepted: 11/30/2024] [Indexed: 12/13/2024] Open
Abstract
Understanding the fundamental biochemical and metabolic requirements for the replication of coronaviruses within infected cells is of notable interest for the development of broad-based therapeutic strategies, given the likelihood of the emergence of new pandemic-potential virus species, as well as future variants of SARS-CoV-2. Here we demonstrate members of the glutaminase family of enzymes (GLS and GLS2), which catalyze the hydrolysis of glutamine to glutamate (i.e., the first step in glutamine metabolism), play key roles in coronavirus replication in host cells. Using a range of human seasonal and zoonotic coronaviruses, we show three examples where GLS expression increases during coronavirus infection of host cells, and another where GLS2 is upregulated. The viruses hijack the metabolic machinery responsible for glutamine metabolism to generate the building blocks for biosynthetic processes and satisfy the bioenergetic requirements demanded by the "glutamine addiction" of virus-infected cells. We demonstrate that genetic silencing of glutaminase enzymes reduces coronavirus infection and that newer members of two classes of allosteric inhibitors targeting these enzymes, designated as SU1, a pan-GLS/GLS2 inhibitor, and UP4, a specific GLS inhibitor, block viral replication in epithelial cells. Moreover, treatment of SARS-CoV-2 infected K18-human ACE2 transgenic mice with SU1 resulted in their complete survival compared to untreated control animals, which succumbed within 10 days post-infection. Overall, these findings highlight the importance of glutamine metabolism for coronavirus replication in human cells and mice and show that glutaminase inhibitors can block coronavirus infection and thereby may represent a novel class of broad-based anti-viral drug candidates.
Collapse
Affiliation(s)
- Kai Su Greene
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Annette Choi
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA
| | - Nianhui Yang
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Matthew Chen
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Ruizhi Li
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Yijian Qiu
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Shahrzad Ezzatpour
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA
| | - Katherine S Rojas
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Jonathan Shen
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Kristin F Wilson
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - William P Katt
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA
| | - Hector C Aguilar
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA
| | - Michael J Lukey
- Cancer Center, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Gary R Whittaker
- Department of Microbiology and Immunology, Cornell University, Ithaca, New York, USA; Public & Ecosystem Health, Cornell University, Ithaca, New York, USA
| | - Richard A Cerione
- Department of Molecular Medicine, Cornell University, Ithaca, New York, USA; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York, USA.
| |
Collapse
|
9
|
Li M, Wu Y, Li B, Lu C, Jian G, Shang X, Chen H, Huang J, He B. ACVPICPred: Inhibitory activity prediction of anti-coronavirus peptides based on artificial neural network. Comput Struct Biotechnol J 2024; 23:3625-3633. [PMID: 39469670 PMCID: PMC11513478 DOI: 10.1016/j.csbj.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/30/2024] Open
Abstract
Peptides, as small molecular compounds, exhibit prominent advantages in the inhibition of coronaviruses due to their safety, efficacy, and specificity, holding great promise as drugs against coronaviruses. The rapid and efficient determination of the activity of anti-coronavirus peptides (ACovPs) can greatly accelerate the development of drugs for treating coronavirus-related diseases. Hence, we present ACVPICPred, a computational model designed to predict the inhibitory activity of ACovPs based on their sequences and structural information. By leveraging bioinformatics tools AlphaFold3 for structural predictions and several feature extraction methods, the model integrates both sequence and structural features to enhance prediction accuracy. To address the limitations of existing datasets, we employed data augmentation techniques, including the introduction of noise and the SMOGN, to improve the model robustness. The model's performance was evaluated through five-fold cross-validation, achieving a Pearson correlation coefficient of 0.7668 (p < 0.05) and an R² of 0.5880 on the training dataset. Overall, in our study, compared to models that only use sequence features, models that combine structural features have achieved more robust results in various evaluation metrics. ACVPICPred is freely accessible at the following URL: http://i.uestc.edu.cn/acvpICPred/main/Main.php.
Collapse
Affiliation(s)
- Min Li
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Yifei Wu
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Bowen Li
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Chunying Lu
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Guifen Jian
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Xing Shang
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Heng Chen
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| | - Jian Huang
- School of Life Science and Technology, University of Electronic Science and Technology of China, No.2006, Xiyuan Ave, West Hi‑Tech Zone, Chengdu 6173001, Sichuan, China
| | - Bifang He
- Medical College, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
- State Key Laboratory of Public Big Data, Guizhou University, Huaxi District, Guiyang 550025, Guizhou, China
| |
Collapse
|
10
|
Georgiou EA, Paraskevas K, Koutra C, Persoons L, Schols D, De Jonghe S, Kostakis IK. Exploring 4,7-Disubstituted Pyrimido[4,5- d]pyrimidines as Antiviral and Anticancer Agents. Molecules 2024; 29:5549. [PMID: 39683709 DOI: 10.3390/molecules29235549] [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: 10/16/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Thirteen new 4,7-disubstituted pyrimido[4,5-d]pyrimidines were synthesized via a straightforward methodology starting from thiourea. The anti-proliferative activity of these compounds was evaluated across a diverse panel of eight cancer cell lines, with derivatives 7d and 7h showing efficacy against several hematological cancer types. Furthermore, all compounds were assessed for their antiviral potency against a panel of viruses. Compounds featuring a cyclopropylamino group and an aminoindane moiety exhibited remarkable efficacy against human coronavirus 229E (HCoV-229E). These findings highlight the pyrimidino[4,5-d]pyrimidine scaffold as an interesting framework for the design of novel antiviral agents against HCoVs, with compounds 7a, 7b, and 7f emerging as strong candidates for further investigation.
Collapse
Affiliation(s)
- Eleftheria A Georgiou
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece
| | - Konstantinos Paraskevas
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece
| | - Christina Koutra
- Department of Pharmacy, Division of Pharmacognosy and Natural Products Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece
| | - Leentje Persoons
- Molecular Genetics and Therapeutics in Virology and Oncology Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Herestraat 49, P.O. Box 1043, 3000 Leuven, Belgium
| | - Dominique Schols
- Molecular Structural and Translational Virology Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Herestraat 49, P.O. Box 1043, 3000 Leuven, Belgium
| | - Steven De Jonghe
- Molecular Structural and Translational Virology Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Herestraat 49, P.O. Box 1043, 3000 Leuven, Belgium
| | - Ioannis K Kostakis
- Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece
| |
Collapse
|
11
|
Gonzalez-Rubio J, Navarro-López JD, Jiménez-Díaz L, Najera A. Systematic review and meta analysis of cross immunity and the smokers paradox in COVID19. Sci Rep 2024; 14:24344. [PMID: 39420134 PMCID: PMC11487265 DOI: 10.1038/s41598-024-75632-6] [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: 02/24/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
COVID-19 pandemic, caused by the novel SARS-CoV-2 virus, has raised significant interest in understanding potential cross-immunity mechanisms. Recent evidence suggests that T-cells associated with common cold coronaviruses (229E, NL63, OC43, HKU1) may provide some level of cross-immunity against SARS-CoV-2. It is also known that the prevalence of smokers among patients admitted to hospital for COVID-19 is lower than expected according to the corresponding country's smoking prevalence, which is known as smoker's paradox in COVID-19. No clear consensus to explain it has yet been reached. This phenomenon suggests a complex interaction between smoking and immune response. Nonetheless, very few works have studied the prevalence of smokers in those infected by common cold coronaviruses, and its relation to COVID-19 has not been investigated. We performed a systematic review and meta-analysis to study the prevalence of smokers among patients infected by common cold coronaviruses, and to compare them to the corresponding country's smoking prevalence. L'Abbé plots were used to visually assess the consistency of the observed effects across the different studies included in the meta-analysis. Additionally, significant differences were found in smoking prevalence among the various types of ccCoV, indicating the need for further research into the biological mechanisms driving these disparities. The results show that smoking prevalence is higher among those patients infected by these coronaviruses than in the general population (OR = 1.37, 95% CI: 0.81-2.33). A study was separately done for the four coronavirus types, and the prevalence of smokers was higher in three of the four than that corresponding to country, gender and study year: OC43 (OR = 1.93, 95% CI: 0.64-5.82); HKU1 (OR = 3.62, 95% CI: 1.21-10.85); NL63 (OR = 1.93, 95% CI: 0.64-5.82); 229E (OR = 0.97, 95% CI: 0.50-1.90). The heterogeneity of the studies was assessed using the Cochrane Chi-squared test, I-squared (I2), and Tau-squared (τ2). This detailed statistical analysis enhances the robustness of our findings and highlights the variations in smoking prevalence among different ccCoVs. Our data suggest that COVID-19 might be less prevalent among smokers due to greater cross-immunity from a larger number or more recent infections by common cold coronaviruses than the non-smoking population, which would explain smoker's paradox in COVID-19. IMPLICATIONS. The low prevalence of current smokers among SARS-CoV-2 patients is a finding recurrently repeated, even leading to postulate the "smoker's paradox" in COVID-19. This fact compelled us to study the prevalence of smokers among patients infected by common cold coronaviruses, and to compare them to the corresponding country's smoking prevalence. Our data could explain smoker's paradox in COVID-19 by a greater cross immunity due to a larger number, or more recent infections by common cold coronaviruses than the non-smoking population. This manuscript allow understand potential unrevealed mechanism for low prevalence of current smokers among SARS-CoV-2 patients.
Collapse
Affiliation(s)
- Jesús Gonzalez-Rubio
- Department of Medical Sciences, Faculty of Medicine of Albacete, Neurophysiology & Behavior Lab, Institute of Biomedicine (IB), University of Castilla-La Mancha, Albacete, Spain.
| | - Juan D Navarro-López
- Department of Medical Sciences, Faculty of Medicine of Ciudad Real, Neurophysiology & Behavior Lab, Institute of Biomedicine (IB), University of Castilla-La Mancha, Ciudad Real, Spain.
| | - Lydia Jiménez-Díaz
- Department of Medical Sciences, Faculty of Medicine of Ciudad Real, Neurophysiology & Behavior Lab, Institute of Biomedicine (IB), University of Castilla-La Mancha, Ciudad Real, Spain.
| | - Alberto Najera
- Department of Medical Sciences, Faculty of Medicine of Albacete, Neurophysiology & Behavior Lab, Institute of Biomedicine (IB), University of Castilla-La Mancha, Albacete, Spain
| |
Collapse
|
12
|
Peng Q, Fu P, Zhou Y, Lang Y, Zhao S, Wen Y, Wang Y, Wu R, Zhao Q, Du S, Cao S, Huang X, Yan Q. Phylogenetic Analysis of Porcine Epidemic Diarrhea Virus (PEDV) during 2020-2022 and Isolation of a Variant Recombinant PEDV Strain. Int J Mol Sci 2024; 25:10878. [PMID: 39456662 PMCID: PMC11507624 DOI: 10.3390/ijms252010878] [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: 09/12/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Porcine epidemic diarrhea (PED) is an acute, highly contagious, and infectious disease caused by porcine epidemic diarrhea virus (PEDV). PEDV can affect pigs of all ages, with 50~100% mortality in neonatal piglets and substantial economic losses in the swine industry. In the present study, 347 fecal and intestinal samples were collected from seven regions in China during 2020-2022. A comprehensive molecular investigation of the spike (S) gene of PEDV strains was carried out, which included phylogenetic analysis of the obtained PEDV sequences. Epidemiological surveillance data indicate that the GIIc subgroup strains are widely distributed among pigs. A PEDV strain was successfully isolated from positive small intestine samples and identified through RT-PCR detection using specific N gene primers of PEDV, indirect immunofluorescence assay (IFA), TEM analysis, genome sequencing, and full-length S gene analysis, named PEDV/SC/2022. RDP and SimPlot analysis showed that the isolate originated from the recombination of PEDV/AH2012 and PEDV/AJ1102. In conclusion, our findings contribute to the current understanding of PEDV epidemiology and provide valuable information for the control of PED outbreaks in China.
Collapse
Affiliation(s)
- Qianling Peng
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Ping Fu
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yutong Zhou
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yifei Lang
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Shan Zhao
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yiping Wen
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yiping Wang
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Rui Wu
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Qin Zhao
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Senyan Du
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Sanjie Cao
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaobo Huang
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Qigui Yan
- Swine Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
13
|
Andersson K, Azatyan A, Ekenberg M, Güçlüler G, Sardon Puig L, Puumalainen M, Pramer T, Monteil VM, Mirazimi A. A CRISPR-Cas13b System Degrades SARS-CoV and SARS-CoV-2 RNA In Vitro. Viruses 2024; 16:1539. [PMID: 39459873 PMCID: PMC11512209 DOI: 10.3390/v16101539] [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: 09/05/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
In a time of climate change, population growth, and globalization, the risk of viral spread has significantly increased. The 21st century has already witnessed outbreaks of Severe Acute Respiratory Syndrome virus (SARS-CoV), Severe Acute Respiratory Syndrome virus 2 (SARS-CoV-2), Ebola virus and Influenza virus, among others. Viruses rapidly adapt and evade human immune systems, complicating the development of effective antiviral countermeasures. Consequently, the need for novel antivirals resilient to viral mutations is urgent. In this study, we developed a CRISPR-Cas13b system to target SARS-CoV-2. Interestingly, this system was also efficient against SARS-CoV, demonstrating broad-spectrum potential. Our findings highlight CRISPR-Cas13b as a promising tool for antiviral therapeutics, underscoring its potential in RNA-virus-associated pandemic responses.
Collapse
Affiliation(s)
- Klara Andersson
- Department of Laboratory Medicine, Unit of Clinical Microbiology, Karolinska Institutet, 17177 Stockholm, Sweden; (K.A.); (A.M.)
- Biomedrex Genetics, Alfred Nobels allé 8, 14152 Stockholm, Sweden; (A.A.); (M.E.); (G.G.); (L.S.P.); (M.P.); (T.P.)
| | - Ani Azatyan
- Biomedrex Genetics, Alfred Nobels allé 8, 14152 Stockholm, Sweden; (A.A.); (M.E.); (G.G.); (L.S.P.); (M.P.); (T.P.)
| | - Martin Ekenberg
- Biomedrex Genetics, Alfred Nobels allé 8, 14152 Stockholm, Sweden; (A.A.); (M.E.); (G.G.); (L.S.P.); (M.P.); (T.P.)
| | - Gözde Güçlüler
- Biomedrex Genetics, Alfred Nobels allé 8, 14152 Stockholm, Sweden; (A.A.); (M.E.); (G.G.); (L.S.P.); (M.P.); (T.P.)
| | - Laura Sardon Puig
- Biomedrex Genetics, Alfred Nobels allé 8, 14152 Stockholm, Sweden; (A.A.); (M.E.); (G.G.); (L.S.P.); (M.P.); (T.P.)
| | - Marjo Puumalainen
- Biomedrex Genetics, Alfred Nobels allé 8, 14152 Stockholm, Sweden; (A.A.); (M.E.); (G.G.); (L.S.P.); (M.P.); (T.P.)
| | - Theodor Pramer
- Biomedrex Genetics, Alfred Nobels allé 8, 14152 Stockholm, Sweden; (A.A.); (M.E.); (G.G.); (L.S.P.); (M.P.); (T.P.)
| | - Vanessa M. Monteil
- Department of Laboratory Medicine, Unit of Clinical Microbiology, Karolinska Institutet, 17177 Stockholm, Sweden; (K.A.); (A.M.)
- Public Health Agency of Sweden, 17182 Solna, Sweden
| | - Ali Mirazimi
- Department of Laboratory Medicine, Unit of Clinical Microbiology, Karolinska Institutet, 17177 Stockholm, Sweden; (K.A.); (A.M.)
- Public Health Agency of Sweden, 17182 Solna, Sweden
- National Veterinary Institute, 75189 Uppsala, Sweden
| |
Collapse
|
14
|
Jiang H, Li W, Zhou X, Zhang J, Li J. Crystal structures of coronaviral main proteases in complex with the non-covalent inhibitor X77. Int J Biol Macromol 2024; 276:133706. [PMID: 38981557 DOI: 10.1016/j.ijbiomac.2024.133706] [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: 05/17/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/11/2024]
Abstract
Main proteases (Mpros) are a class of conserved cysteine hydrolases among coronaviruses and play a crucial role in viral replication. Therefore, Mpros are ideal targets for the development of pan-coronavirus drugs. X77, previously developed against SARS-CoV Mpro, was repurposed as a non-covalent tight binder inhibitor against SARS-CoV-2 Mpro during COVID-19 pandemic. Many novel inhibitors with favorable efficacy have been discovered using X77 as a reference, suggesting that X77 could be a valuable scaffold for drug design. However, the broad-spectrum performance of X77 and underlying mechanism remain less understood. Here, we reported the crystal structures of Mpros from SARS-CoV-2, SARS-CoV, and MERS-CoV, and several Mpro mutants from SARS-CoV-2 variants bound to X77. A detailed analysis of these structures revealed key structural determinants essential for interaction and elucidated the binding modes of X77 with different coronaviral Mpros. The potencies of X77 against these investigated Mpros were further evaluated through molecular dynamic simulation and binding free energy calculation. These data provide molecular insights into broad-spectrum inhibition against coronaviral Mpros by X77 and the similarities and differences of X77 when bound to various Mpros, which will promote X77-based design of novel antivirals with broad-spectrum efficacy against different coronaviruses and SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Haihai Jiang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Wenwen Li
- College of Pharmaceutical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Xuelan Zhou
- College of Pharmaceutical Sciences, Gannan Medical University, Ganzhou 341000, China
| | - Jin Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Jian Li
- College of Pharmaceutical Sciences, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
15
|
Liang Z, Wang J, Zhang H, Gao L, Xu J, Li P, Yang J, Fu X, Duan H, Liu J, Liu T, Ma W, Wu K. Peptide S4 is an entry inhibitor of SARS-CoV-2 infection. Virology 2024; 597:110149. [PMID: 38917689 DOI: 10.1016/j.virol.2024.110149] [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: 03/11/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a significant socioeconomic burden, and combating COVID-19 is imperative. Blocking the SARS-CoV-2 RBD-ACE2 interaction is a promising therapeutic approach for viral infections, as SARS-CoV-2 binds to the ACE2 receptors of host cells via the RBD of spike proteins to infiltrate these cells. We used computer-aided drug design technology and cellular experiments to screen for peptide S4 with high affinity and specificity for the human ACE2 receptor through structural analysis of SARS-CoV-2 and ACE2 interactions. Cellular experiments revealed that peptide S4 effectively inhibited SARS-CoV-2 and HCoV-NL63 viruses from infecting host cells and was safe for cells at effective concentrations. Based on these findings, peptide S4 may be a potential pharmaceutical agent for clinical application in the treatment of the ongoing SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Zhiyu Liang
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China; Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jiamei Wang
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Huan Zhang
- Guangdong Center for Disease Control and Prevention, Guangdong, China
| | - Lixia Gao
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jun Xu
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Peiran Li
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jie Yang
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Xinting Fu
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Han Duan
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jiayan Liu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China; Institute of Antibody Engineering, School of Laboratory Medicine & Biotechnology, Southern Medical University, Guangzhou, China
| | - Tiancai Liu
- Institute of Antibody Engineering, School of Laboratory Medicine & Biotechnology, Southern Medical University, Guangzhou, China
| | - Weifeng Ma
- Department of Microbiology, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| | - Kun Wu
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
16
|
Guan Y, Cheng J, Lv Q, Wei X, Jiang B, Xiao P. Exploring new therapeutic potential of five commonly used Pteris medicinal plants through pharmaphylogenomics and network pharmacology. CHINESE HERBAL MEDICINES 2024. [DOI: 10.1016/j.chmed.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
17
|
Liu QR, Duan DF, Yan W, Zhang HY, Ding XL, Wang SB, Tang XR, Ao XL, Chen XL, Cao JY, Zhang RP, Hou B. LC/MS-guided isolation and GNPS procedures to identify flavonoid of HCoV-OC43 inhibitors. Fitoterapia 2024; 177:106077. [PMID: 38906387 DOI: 10.1016/j.fitote.2024.106077] [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: 03/18/2024] [Revised: 06/07/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
The screening of based target compounds supported by LC/MS, MS/MS and Global Natural Products Social (GNPS) used to identify the compounds 1-10 of Butea monsperma. They were evaluated in human malignant embryonic rhabdomyoma cells (RD cells) infected with Human coronavirus OC43 (HCoV-OC43) and showed significant inhibitory activity. Target inhibition tests showed that compounds 6 and 8 inhibited the proteolytic enzyme 3CLpro, which is widely present in coronavirus and plays an important role in the replication process, with an effective IC50 value. The study confirmed that dioxymethylene of compound 8 may be a key active fragment in inhibiting coronavirus (EC50 7.2 μM, SI > 139.1). The results have led to identifying natural bioactive compounds for possible inhibiting HCoV-OC43 and developing drug for Traditional Chinese Medicine (TCM).
Collapse
Affiliation(s)
- Qian-Ru Liu
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, PR China
| | - Ding-Fu Duan
- Hubei Jiangxia Laboratory, Wuhan 430200, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, PR China
| | - Wei Yan
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, PR China
| | - Hai-Ying Zhang
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, PR China
| | - Xiao-Lin Ding
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, PR China
| | - Shu-Bing Wang
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, PR China
| | - Xiao-Rui Tang
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, PR China
| | - Xuan-Li Ao
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, PR China
| | - Xing-Long Chen
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, PR China
| | - Jun-Yuan Cao
- Hubei Jiangxia Laboratory, Wuhan 430200, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei 430071, PR China.
| | - Rong-Ping Zhang
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, PR China.
| | - Bo Hou
- School of Chinese Materia Medica & Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming 650500, PR China.
| |
Collapse
|
18
|
Silawal S, Gögele C, Pelikan P, Werner C, Levidou G, Mahato R, Schulze-Tanzil G. A Histological Analysis and Detection of Complement Regulatory Protein CD55 in SARS-CoV-2 Infected Lungs. Life (Basel) 2024; 14:1058. [PMID: 39337843 PMCID: PMC11432792 DOI: 10.3390/life14091058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND A complement imbalance in lung alveolar tissue can play a deteriorating role in COVID-19, leading to acute respiratory distress syndrome (ARDS). CD55 is a transmembrane glycoprotein that inhibits the activation of the complement system at the intermediate cascade level, blocking the activity of the C3 convertase. OBJECTIVE In our study, lung specimens from COVID-19 and ARDS-positive COVID+/ARDS+ patients were compared with COVID-19 and ARDS-negative COVID-/ARDS- as well as COVID-/ARDS+ patients. METHODS Histochemical staining and immunolabeling of CD55 protein were performed. RESULTS The COVID-/ARDS- specimen showed higher expression and homogeneous distribution of glycosaminoglycans as well as compactly arranged elastic and collagen fibers of the alveolar walls in comparison to ARDS-affected lungs. In addition, COVID-/ARDS- lung tissues revealed stronger and homogenously distributed CD55 expression on the alveolar walls in comparison to the disrupted COVID-/ARDS+ lung tissues. CONCLUSIONS Even though the collapse of the alveolar linings and the accumulation of cellular components in the alveolar spaces were characteristic of COVID+/ARDS+ lung tissues, evaluating CD55 expression could be relevant to understand its relation to the disease. Furthermore, targeting CD55 upregulation as a potential therapy could be an option for post-infectious complications of COVID-19 and other inflammatory lung diseases in the future.
Collapse
Affiliation(s)
- Sandeep Silawal
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Clemens Gögele
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Petr Pelikan
- Institute for Pathology, Paracelsus Medical University, Nuremberg, General Hospital, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Christian Werner
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Georgia Levidou
- Institute for Pathology, Paracelsus Medical University, Nuremberg, General Hospital, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| | - Raman Mahato
- Department of Emergency and Intensive Care Medicine, Klinikum Ernst von Bergmann, Charlottenstraße 72, 14467 Potsdam, Germany
| | - Gundula Schulze-Tanzil
- Institute of Anatomy and Cell Biology, Paracelsus Medical University, Nuremberg and Salzburg, General Hospital Nuremberg, Prof. Ernst Nathan Str. 1, 90419 Nuremberg, Germany
| |
Collapse
|
19
|
Zhang Y, Wu F, Han Y, Wu Y, Huang L, Huang Y, Yan D, Jiang X, Ma J, Xu W. Unraveling the assembly mechanism of SADS-CoV virus nucleocapsid protein: insights from RNA binding, dimerization, and epitope diversity profiling. J Virol 2024; 98:e0092624. [PMID: 39082816 PMCID: PMC11334509 DOI: 10.1128/jvi.00926-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/02/2024] [Indexed: 08/21/2024] Open
Abstract
The swine acute diarrhea syndrome coronavirus (SADS-CoV) has caused significant disruptions in porcine breeding and raised concerns about potential human infection. The nucleocapsid (N) protein of SADS-CoV plays a vital role in viral assembly and replication, but its structure and functions remain poorly understood. This study utilized biochemistry, X-ray crystallography, and immunization techniques to investigate the N protein's structure and function in SADS-CoV. Our findings revealed distinct domains within the N protein, including an RNA-binding domain, two disordered domains, and a dimerization domain. Through biochemical assays, we confirmed that the N-terminal domain functions as an RNA-binding domain, and the C-terminal domain is involved in dimerization, with the crystal structure analysis providing visual evidence of dimer formation. Immunization experiments demonstrated that the disordered domain 2 elicited a significant antibody response. These identified domains and their interactions are crucial for viral assembly. This comprehensive understanding of the N protein in SADS-CoV enhances our knowledge of its assembly and replication mechanisms, enabling the development of targeted interventions and therapeutic strategies. IMPORTANCE SADS-CoV is a porcine coronavirus that originated from a bat HKU2-related coronavirus. It causes devastating swine diseases and poses a high risk of spillover to humans. The coronavirus N protein, as the most abundant viral protein in infected cells, likely plays a key role in viral assembly and replication. However, the structure and function of this protein remain unclear. Therefore, this study employed a combination of biochemistry and X-ray crystallography to uncover distinct structural domains in the N protein, including RNA-binding domains, two disordered domains, and dimerization domains. Additionally, we made the novel discovery that the disordered domain elicited a significant antibody response. These findings provide new insights into the structure and functions of the SADS-CoV N protein, which have important implications for future studies on SADS-CoV diagnosis, as well as the development of vaccines and anti-viral drugs.
Collapse
Affiliation(s)
- Ying Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Fang Wu
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Yongyue Han
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yuzhe Wu
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Liqiu Huang
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yuanwei Huang
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Di Yan
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiwen Jiang
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jingyun Ma
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Wei Xu
- Guangdong Provincial Key Laboratory of New Drug Screening & NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Key Laboratory of Infectious Diseases Research in South China, Ministry of Education, Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Raczkiewicz I, Rivière C, Bouquet P, Desmarets L, Tarricone A, Camuzet C, François N, Lefèvre G, Silva Angulo F, Robil C, Trottein F, Sahpaz S, Dubuisson J, Belouzard S, Goffard A, Séron K. Hyperforin, the major metabolite of St. John's wort, exhibits pan-coronavirus antiviral activity. Front Microbiol 2024; 15:1443183. [PMID: 39176276 PMCID: PMC11339956 DOI: 10.3389/fmicb.2024.1443183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024] Open
Abstract
Introduction The COVID-19 pandemic caused by the SARS-CoV-2 virus has underscored the urgent necessity for the development of antiviral compounds that can effectively target coronaviruses. In this study, we present the first evidence of the antiviral efficacy of hyperforin, a major metabolite of St. John's wort, for which safety and bioavailability in humans have already been established. Methods Antiviral assays were conducted in cell culture with four human coronaviruses: three of high virulence, SARS-CoV-2, SARS-CoV, and MERS-CoV, and one causing mild symptoms, HCoV-229E. The antiviral activity was also evaluated in human primary airway epithelial cells. To ascertain the viral step inhibited by hyperforin, time-of-addition assays were conducted. Subsequently, a combination assay of hyperforin with remdesivir was performed. Results The results demonstrated that hyperforin exhibited notable antiviral activity against the four tested human coronaviruses, with IC50 values spanning from 0.24 to 2.55 µM. Kinetic studies indicated that the observed activity occur at a post-entry step, potentially during replication. The antiviral efficacy of hyperforin was additionally corroborated in human primary airway epithelial cells. The results demonstrated a reduction in both intracellular and extracellular SARS-CoV-2 viral RNA, confirming that hyperforin targeted the replication step. Finally, an additive antiviral effect on SARS-CoV-2 was observed when hyperforin was combined with remdesivir. Discussion In conclusion, hyperforin has been identified as a novel pan-coronavirus inhibitor with activity in human primary airway epithelial cells, a preclinical model for coronaviruses. These findings collectively suggest that hyperforin has potential as a candidate antiviral agent against current and future human coronaviruses.
Collapse
Affiliation(s)
- Imelda Raczkiewicz
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Céline Rivière
- BioEcoAgro, Joint Research Unit 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV – Institut Charles Viollette, Lille, France
| | - Peggy Bouquet
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Lowiese Desmarets
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Audrey Tarricone
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Charline Camuzet
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Nathan François
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Gabriel Lefèvre
- BioEcoAgro, Joint Research Unit 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV – Institut Charles Viollette, Lille, France
| | - Fabiola Silva Angulo
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Cyril Robil
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - François Trottein
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Sevser Sahpaz
- BioEcoAgro, Joint Research Unit 1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte d’Opale, ICV – Institut Charles Viollette, Lille, France
| | - Jean Dubuisson
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Sandrine Belouzard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Anne Goffard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| | - Karin Séron
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 – UMR9017 – Center for Infection and Immunity of Lille (CIIL), Lille, France
| |
Collapse
|
21
|
Michaels TM, Essop MF, Joseph DE. Potential Effects of Hyperglycemia on SARS-CoV-2 Entry Mechanisms in Pancreatic Beta Cells. Viruses 2024; 16:1243. [PMID: 39205219 PMCID: PMC11358987 DOI: 10.3390/v16081243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The COVID-19 pandemic has revealed a bidirectional relationship between SARS-CoV-2 infection and diabetes mellitus. Existing evidence strongly suggests hyperglycemia as an independent risk factor for severe COVID-19, resulting in increased morbidity and mortality. Conversely, recent studies have reported new-onset diabetes following SARS-CoV-2 infection, hinting at a potential direct viral attack on pancreatic beta cells. In this review, we explore how hyperglycemia, a hallmark of diabetes, might influence SARS-CoV-2 entry and accessory proteins in pancreatic β-cells. We examine how the virus may enter and manipulate such cells, focusing on the role of the spike protein and its interaction with host receptors. Additionally, we analyze potential effects on endosomal processing and accessory proteins involved in viral infection. Our analysis suggests a complex interplay between hyperglycemia and SARS-CoV-2 in pancreatic β-cells. Understanding these mechanisms may help unlock urgent therapeutic strategies to mitigate the detrimental effects of COVID-19 in diabetic patients and unveil if the virus itself can trigger diabetes onset.
Collapse
Affiliation(s)
- Tara M. Michaels
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| | - M. Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa;
| | - Danzil E. Joseph
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch 7600, South Africa;
| |
Collapse
|
22
|
Li M, Yang Y, Wang P, Que W, Zhong L, Cai Z, Liu Y, Yang L, Liu Y. Transcriptome dynamics of the BHK21 cell line in response to human coronavirus OC43 infection. Microbiol Res 2024; 285:127750. [PMID: 38761489 DOI: 10.1016/j.micres.2024.127750] [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: 03/19/2024] [Revised: 04/16/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
The progress of viral infection involves numerous transcriptional regulatory events. The identification of the newly synthesized transcripts helps us to understand the replication mechanisms and pathogenesis of the virus. Here, we utilized a time-resolved technique called metabolic RNA labeling approach called thiol(SH)-linked alkylation for the metabolic sequencing of RNA (SLAM-seq) to differentially elucidate the levels of steady-state and newly synthesized RNAs of BHK21 cell line in response to human coronavirus OC43 (HCoV-OC43) infection. Our results showed that the Wnt/β-catenin signaling pathway was significantly enriched with the newly synthesized transcripts of BHK21 cell line in response to HCoV-OC43 infection. Moreover, inhibition of the Wnt pathway promoted viral replication in the early stage of infection, but inhibited it in the later stage of infection. Furthermore, remdesivir inhibits the upregulation of the Wnt/β-catenin signaling pathway induced by early infection with HCoV-OC43. Collectively, our study showed the diverse roles of Wnt/β-catenin pathway at different stages of HCoV-OC43 infection, suggesting a potential target for the antiviral treatment. In addition, although infection with HCoV-OC43 induces cytopathic effects in BHK21 cells, inhibiting apoptosis does not affect the intracellular replication of the virus. Monitoring newly synthesized RNA based on such time-resolved approach is a highly promising method for studying the mechanism of viral infections.
Collapse
Affiliation(s)
- Mianhuan Li
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, People's Republic of China; Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China
| | - Yang Yang
- Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China
| | - Pusen Wang
- Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China
| | - Weitao Que
- Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China
| | - Lin Zhong
- Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China
| | - Zhao Cai
- Shenzhen Mindray Bio-Medical Electronics Co.,Ltd, Shenzhen 518057, People's Republic of China
| | - Yang Liu
- Southern University of Science and Technology Hospital, Shenzhen 518055, People's Republic of China
| | - Liang Yang
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, People's Republic of China; Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China.
| | - Yingxia Liu
- Shenzhen Third People's Hospital, National Clinical Research Centre for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518112, People's Republic of China.
| |
Collapse
|
23
|
Wei Y, Song J, Zhang J, Chen S, Yu Z, He L, Chen J. Exploring TRIM proteins' role in antiviral defense against influenza A virus and respiratory coronaviruses. Front Cell Infect Microbiol 2024; 14:1420854. [PMID: 39077432 PMCID: PMC11284085 DOI: 10.3389/fcimb.2024.1420854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 06/19/2024] [Indexed: 07/31/2024] Open
Abstract
Numerous tripartite motif (TRIM) proteins, identified as E3 ubiquitin ligases, participate in various viral infections through ubiquitylation, ISGylation, and SUMOylation processes. Respiratory viruses, particularly influenza A virus (IAV) and respiratory coronaviruses (CoVs), have severely threatened public health with high morbidity and mortality, causing incalculable losses. Research on the regulation of TRIM proteins in respiratory virus infections is crucial for disease prevention and control. This review introduces TRIM proteins, summarizes recent discoveries regarding their roles and molecular mechanisms in IAV and CoVs infections, discusses current research gaps, and explores potential future trends in this rapidly developing field. It aims to enhance understanding of virus-host interactions and inform the development of new molecularly targeted therapies.
Collapse
Affiliation(s)
- Ying Wei
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang, China
| | - Junzhu Song
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang, China
| | - Jingyu Zhang
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang, China
| | - Songbiao Chen
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang, China
| | - Zuhua Yu
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang, China
| | - Lei He
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang, China
| | - Jian Chen
- Laboratory of Functional Microbiology and Animal Health, College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
- Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang, China
- The Key Lab of Animal Disease and Public Health, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
24
|
Xiao YQ, Long J, Zhang SS, Zhu YY, Gu SX. Non-peptidic inhibitors targeting SARS-CoV-2 main protease: A review. Bioorg Chem 2024; 147:107380. [PMID: 38636432 DOI: 10.1016/j.bioorg.2024.107380] [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: 02/20/2024] [Revised: 04/11/2024] [Accepted: 04/14/2024] [Indexed: 04/20/2024]
Abstract
The COVID-19 pandemic continues to pose a threat to global health, and sounds the alarm for research & development of effective anti-coronavirus drugs, which are crucial for the patients and urgently needed for the current epidemic and future crisis. The main protease (Mpro) stands as an essential enzyme in the maturation process of SARS-CoV-2, playing an irreplaceable role in regulating viral RNA replication and transcription. It has emerged as an ideal target for developing antiviral agents against SARS-CoV-2 due to its high conservation and the absence of homologous proteases in the human body. Among the SARS-CoV-2 Mpro inhibitors, non-peptidic compounds hold promising prospects owing to their excellent antiviral activity and improved metabolic stability. In this review, we offer an overview of research progress concerning non-peptidic SARS-CoV-2 Mpro inhibitors since 2020. The efforts delved into molecular structures, structure-activity relationships (SARs), biological activity, and binding modes of these inhibitors with Mpro. This review aims to provide valuable clues and insights for the development of anti-SARS-CoV-2 agents as well as broad-spectrum coronavirus Mpro inhibitors.
Collapse
Affiliation(s)
- Ya-Qi Xiao
- School of Chemical Engineering and Pharmacy, Pharmaceutical Research Institute, Wuhan Institute of Technology, Wuhan 430205, China
| | - Jiao Long
- School of Chemical Engineering and Pharmacy, Pharmaceutical Research Institute, Wuhan Institute of Technology, Wuhan 430205, China
| | - Shuang-Shuang Zhang
- School of Chemical Engineering and Pharmacy, Pharmaceutical Research Institute, Wuhan Institute of Technology, Wuhan 430205, China.
| | - Yuan-Yuan Zhu
- School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan 430205, China.
| | - Shuang-Xi Gu
- School of Chemical Engineering and Pharmacy, Pharmaceutical Research Institute, Wuhan Institute of Technology, Wuhan 430205, China.
| |
Collapse
|
25
|
Li S, Li H, Lian R, Xie J, Feng R. New perspective of small-molecule antiviral drugs development for RNA viruses. Virology 2024; 594:110042. [PMID: 38492519 DOI: 10.1016/j.virol.2024.110042] [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: 10/21/2023] [Revised: 02/20/2024] [Accepted: 03/01/2024] [Indexed: 03/18/2024]
Abstract
High variability and adaptability of RNA viruses allows them to spread between humans and animals, causing large-scale infectious diseases which seriously threat human and animal health and social development. At present, AIDS, viral hepatitis and other viral diseases with high incidence and low cure rate are still spreading around the world. The outbreaks of Ebola, Zika, dengue and in particular of the global pandemic of COVID-19 have presented serious challenges to the global public health system. The development of highly effective and broad-spectrum antiviral drugs is a substantial and urgent research subject to deal with the current RNA virus infection and the possible new viral infections in the future. In recent years, with the rapid development of modern disciplines such as artificial intelligence technology, bioinformatics, molecular biology, and structural biology, some new strategies and targets for antivirals development have emerged. Here we review the main strategies and new targets for developing small-molecule antiviral drugs against RNA viruses through the analysis of the new drug development progress against several highly pathogenic RNA viruses, to provide clues for development of future antivirals.
Collapse
Affiliation(s)
- Shasha Li
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, 730030, China; Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Huixia Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Ruiya Lian
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, 730030, China; Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Jingying Xie
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, 730030, China; Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Ruofei Feng
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Affairs Commission, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China.
| |
Collapse
|
26
|
Li Y, Tan X, Deng J, Liu X, Liu Q, Zhang Z, Huang X, Shen C, Xu K, Zhou L, Chen Y. An optimized high-throughput SARS-CoV-2 dual reporter trans-complementation system for antiviral screening in vitro and in vivo. Virol Sin 2024; 39:447-458. [PMID: 38548102 PMCID: PMC11280264 DOI: 10.1016/j.virs.2024.03.009] [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: 12/04/2023] [Accepted: 03/21/2024] [Indexed: 04/25/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still epidemic around the world. The manipulation of SARS-CoV-2 is restricted to biosafety level 3 laboratories (BSL-3). In this study, we developed a SARS-CoV-2 ΔN-GFP-HiBiT replicon delivery particles (RDPs) encoding a dual reporter gene, GFP-HiBiT, capable of producing both GFP signal and luciferase activities. Through optimal selection of the reporter gene, GFP-HiBiT demonstrated superior stability and convenience for antiviral evaluation. Additionally, we established a RDP infection mouse model by delivering the N gene into K18-hACE2 KI mouse through lentivirus. This mouse model supports RDP replication and can be utilized for in vivo antiviral evaluations. In summary, the RDP system serves as a valuable tool for efficient antiviral screening and studying the gene function of SARS-CoV-2. Importantly, this system can be manipulated in BSL-2 laboratories, decreasing the threshold of experimental requirements.
Collapse
Affiliation(s)
- Yingjian Li
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430072, China
| | - Xue Tan
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430072, China
| | - Jikai Deng
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430072, China
| | - Xuemei Liu
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430072, China
| | - Qianyun Liu
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430072, China
| | - Zhen Zhang
- Institute for Vaccine Research at Animal Bio-safety Level Ⅲ Laboratory, Wuhan University School of Medicine, Wuhan, 430071, China
| | - Xiaoya Huang
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430072, China
| | - Chao Shen
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430072, China
| | - Ke Xu
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430072, China
| | - Li Zhou
- Institute for Vaccine Research at Animal Bio-safety Level Ⅲ Laboratory, Wuhan University School of Medicine, Wuhan, 430071, China
| | - Yu Chen
- State Key Laboratory of Virology, RNA Institute, College of Life Sciences and Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
27
|
Keramidas P, Pitou M, Papachristou E, Choli-Papadopoulou T. Insights into the Activation of Unfolded Protein Response Mechanism during Coronavirus Infection. Curr Issues Mol Biol 2024; 46:4286-4308. [PMID: 38785529 PMCID: PMC11120126 DOI: 10.3390/cimb46050261] [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: 03/28/2024] [Revised: 04/24/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Coronaviruses represent a significant class of viruses that affect both animals and humans. Their replication cycle is strongly associated with the endoplasmic reticulum (ER), which, upon virus invasion, triggers ER stress responses. The activation of the unfolded protein response (UPR) within infected cells is performed from three transmembrane receptors, IRE1, PERK, and ATF6, and results in a reduction in protein production, a boost in the ER's ability to fold proteins properly, and the initiation of ER-associated degradation (ERAD) to remove misfolded or unfolded proteins. However, in cases of prolonged and severe ER stress, the UPR can also instigate apoptotic cell death and inflammation. Herein, we discuss the ER-triggered host responses after coronavirus infection, as well as the pharmaceutical targeting of the UPR as a potential antiviral strategy.
Collapse
Affiliation(s)
| | | | | | - Theodora Choli-Papadopoulou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (P.K.); (M.P.); (E.P.)
| |
Collapse
|
28
|
Mancusi A, Proroga YTR, Maiolino P, Marrone R, D’Emilio C, Girardi S, Egidio M, Boni A, Vicenza T, Suffredini E, Power K. Droplet Digital RT-PCR (dd RT-PCR) Detection of SARS-CoV-2 in Honey Bees and Honey Collected in Apiaries across the Campania Region. Viruses 2024; 16:729. [PMID: 38793611 PMCID: PMC11126096 DOI: 10.3390/v16050729] [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: 02/29/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Coronaviruses (CoVs), a subfamily of Orthocoronavirinae, are viruses that sometimes present a zoonotic character. Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is responsible for the recent outbreak of COVID-19, which, since its outbreak in 2019, has caused about 774,593,066 confirmed cases and 7,028,881 deaths. Aereosols are the main route of transmission among people; however, viral droplets can contaminate surfaces and fomites as well as particulate matter (PM) in suspensions of natural and human origin. Honey bees are well known bioindicators of the presence of pollutants and PMs in the environment as they can collect a great variety of substances during their foraging activities. The aim of this study was to evaluate the possible role of honey bees as bioindicators of the prevalence SARS-CoV-2. In this regard, 91 samples of honey bees and 6 of honey were collected from different apiaries of Campania region (Southern Italy) in four time periods from September 2020 to June 2022 and were analyzed with Droplet Digital RT-PCR for SARS-CoV-2 target genes Orf1b and N. The screening revealed the presence of SARS-CoV-2 in 12/91 in honey bee samples and in 2/6 honey samples. These results suggest that honey bees could also be used as indicators of outbreaks of airborne pathogens such as SARS-CoV-2.
Collapse
Affiliation(s)
- Andrea Mancusi
- Department of Food Security Coordination, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Via Salute No. 2, 80055 Portici, Italy; (A.M.); (Y.T.R.P.); (S.G.)
| | - Yolande Thérèse Rose Proroga
- Department of Food Security Coordination, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Via Salute No. 2, 80055 Portici, Italy; (A.M.); (Y.T.R.P.); (S.G.)
| | - Paola Maiolino
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (P.M.); (R.M.); (C.D.)
| | - Raffaele Marrone
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (P.M.); (R.M.); (C.D.)
| | - Claudia D’Emilio
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (P.M.); (R.M.); (C.D.)
| | - Santa Girardi
- Department of Food Security Coordination, Istituto Zooprofilattico Sperimentale del Mezzogiorno, Via Salute No. 2, 80055 Portici, Italy; (A.M.); (Y.T.R.P.); (S.G.)
| | - Marica Egidio
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Naples, Italy; (P.M.); (R.M.); (C.D.)
| | - Arianna Boni
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (T.V.); (E.S.)
| | - Teresa Vicenza
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (T.V.); (E.S.)
| | - Elisabetta Suffredini
- Department of Food Safety, Nutrition and Veterinary Public Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (A.B.); (T.V.); (E.S.)
| | - Karen Power
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy;
| |
Collapse
|
29
|
Sohrab SS, Alsaqaf F, Hassan AM, Tolah AM, Bajrai LH, Azhar EI. Genomic Diversity and Recombination Analysis of the Spike Protein Gene from Selected Human Coronaviruses. BIOLOGY 2024; 13:282. [PMID: 38666894 PMCID: PMC11048170 DOI: 10.3390/biology13040282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024]
Abstract
Human coronaviruses (HCoVs) are seriously associated with respiratory diseases in humans and animals. The first human pathogenic SARS-CoV emerged in 2002-2003. The second was MERS-CoV, reported from Jeddah, the Kingdom of Saudi Arabia, in 2012, and the third one was SARS-CoV-2, identified from Wuhan City, China, in late December 2019. The HCoV-Spike (S) gene has the highest mutation/insertion/deletion rate and has been the most utilized target for vaccine/antiviral development. In this manuscript, we discuss the genetic diversity, phylogenetic relationships, and recombination patterns of selected HCoVs with emphasis on the S protein gene of MERS-CoV and SARS-CoV-2 to elucidate the possible emergence of new variants/strains of coronavirus in the near future. The findings showed that MERS-CoV and SARS-CoV-2 have significant sequence identity with the selected HCoVs. The phylogenetic tree analysis formed a separate cluster for each HCoV. The recombination pattern analysis showed that the HCoV-NL63-Japan was a probable recombinant. The HCoV-NL63-USA was identified as a major parent while the HCoV-NL63-Netherland was identified as a minor parent. The recombination breakpoints start in the viral genome at the 142 nucleotide position and end at the 1082 nucleotide position with a 99% CI and Bonferroni-corrected p-value of 0.05. The findings of this study provide insightful information about HCoV-S gene diversity, recombination, and evolutionary patterns. Based on these data, it can be concluded that the possible emergence of new strains/variants of HCoV is imminent.
Collapse
Affiliation(s)
- Sayed Sartaj Sohrab
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia; (F.A.); (A.M.H.); (A.M.T.); (L.H.B.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
| | - Fatima Alsaqaf
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia; (F.A.); (A.M.H.); (A.M.T.); (L.H.B.)
| | - Ahmed Mohamed Hassan
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia; (F.A.); (A.M.H.); (A.M.T.); (L.H.B.)
| | - Ahmed Majdi Tolah
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia; (F.A.); (A.M.H.); (A.M.T.); (L.H.B.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, P.O. Box 21911, Rabigh 344, Saudi Arabia
| | - Leena Hussein Bajrai
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia; (F.A.); (A.M.H.); (A.M.T.); (L.H.B.)
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
| | - Esam Ibraheem Azhar
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia; (F.A.); (A.M.H.); (A.M.T.); (L.H.B.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
| |
Collapse
|
30
|
Gao F, Lin W, Wang X, Liao M, Zhang M, Qin N, Chen X, Xia L, Chen Q, Sha O. Identification of receptors and factors associated with human coronaviruses in the oral cavity using single-cell RNA sequencing. Heliyon 2024; 10:e28280. [PMID: 38560173 PMCID: PMC10981076 DOI: 10.1016/j.heliyon.2024.e28280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) ravaged the world, and Coronavirus Disease 2019 (COVID-19) exhibited highly prevalent oral symptoms that had significantly impacted the lives of affected patients. However, the involvement of four human coronavirus (HCoVs), namely SARS-CoV-2, SARS-CoV, MERS-CoV, and HCoV-229E, in oral cavity infections remained poorly understood. We integrated single-cell RNA sequencing (scRNA-seq) data of seven human oral tissues through consistent normalization procedure, including minor salivary gland (MSG), parotid gland (PG), tongue, gingiva, buccal, periodontium and pulp. The Seurat, scDblFinder, Harmony, SingleR, Ucell and scCancer packages were comprehensively used for analysis. We identified specific cell clusters and generated expression profiles of SARS-CoV-2 and coronavirus-associated receptors and factors (SCARFs) in seven oral regions, providing direction for predicting the tropism of four HCoVs for oral tissues, as well as for dental clinical treatment. Based on our analysis, it appears that various SCARFs, including ACE2, ASGR1, KREMEN1, DPP4, ANPEP, CD209, CLEC4G/M, TMPRSS family proteins (including TMPRSS2, TMPRSS4, and TMPRSS11A), and FURIN, are expressed at low levels in the oral cavity. Conversely, BSG, CTSB, and CTSL exhibit enrichment in oral tissues. Our study also demonstrates widespread expression of restriction factors, particularly IFITM1-3 and LY6E, in oral cells. Additionally, some replication, assembly, and trafficking factors appear to exhibit broad oral tissues expression patterns. Overall, the oral cavity could potentially serve as a high-risk site for SARS-CoV-2 infection, while displaying a comparatively lower degree of susceptibility towards other HCoVs (including SARS-CoV, MERS-CoV and HCoV-229E). Specifically, MSG, tongue, and gingiva represent potential sites of vulnerability for four HCoVs infection, with the MSG exhibiting a particularly high susceptibility. However, the expression patterns of SCARFs in other oral sites demonstrate relatively intricate and may only be specifically associated with SARS-CoV-2 infection. Our study sheds light on the mechanisms of HCoVs infection in the oral cavity as well as gains insight into the characteristics and distribution of possible HCoVs target cells in oral tissues, providing potential therapeutic targets for HCoVs infection in the oral cavity.
Collapse
Affiliation(s)
- Feng Gao
- School of Dentistry, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Institute of Dental Research, Shenzhen University, Shenzhen, China
| | - Weiming Lin
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xia Wang
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- The Chinese University of Hong Kong Shenzhen, School of Medicine, Shenzhen, China
| | - Mingfeng Liao
- The Third People's Hospital of Shenzhen, Shenzhen, China
| | - Mingxia Zhang
- The Third People's Hospital of Shenzhen, Shenzhen, China
| | - Nianhong Qin
- Department of Stomatology, Shenzhen People's Hospital, Shenzhen, China
| | - Xianxiong Chen
- School of Dentistry, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Lixin Xia
- Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Ou Sha
- School of Dentistry, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- Institute of Dental Research, Shenzhen University, Shenzhen, China
| |
Collapse
|
31
|
Zhang Y, Zhang J, Li D, Mao Q, Li X, Liang Z, He Q. A Cocktail of Lipid Nanoparticle-mRNA Vaccines Broaden Immune Responses against β-Coronaviruses in a Murine Model. Viruses 2024; 16:484. [PMID: 38543849 PMCID: PMC10976147 DOI: 10.3390/v16030484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 05/23/2024] Open
Abstract
Severe acute respiratory syndrome (SARS)-coronavirus (CoV), Middle Eastern respiratory syndrome (MERS)-CoV, and SARS-CoV-2 have seriously threatened human life in the 21st century. Emerging and re-emerging β-coronaviruses after the coronavirus disease 2019 (COVID-19) epidemic remain possible highly pathogenic agents that can endanger human health. Thus, pan-β-coronavirus vaccine strategies to combat the upcoming dangers are urgently needed. In this study, four LNP-mRNA vaccines, named O, D, S, and M, targeting the spike protein of SARS-CoV-2 Omicron, Delta, SARS-CoV, and MERS-CoV, respectively, were synthesized and characterized for purity and integrity. All four LNP-mRNAs induced effective cellular and humoral immune responses against the corresponding spike protein antigens in mice. Furthermore, LNP-mRNA S and D induced neutralizing antibodies against SARS-CoV and SARS-CoV-2, which failed to cross-react with MERS-CoV. Subsequent evaluation of sequential and cocktail immunizations with LNP-mRNA O, D, S, and M effectively elicited broad immunity against SARS-CoV-2 variants, SARS-CoV, and MERS-CoV. A direct comparison of the sequential with cocktail regimens indicated that the cocktail vaccination strategy induced more potent neutralizing antibodies and T-cell responses against heterotypic viruses as well as broader antibody activity against pan-β-coronaviruses. Overall, these results present a potential pan-β-coronavirus vaccine strategy for improved preparedness prior to future coronavirus threats.
Collapse
Affiliation(s)
- Yi Zhang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, State Key Laboratory of Drug Regulatory Science, Beijing 102629, China; (Y.Z.); (J.Z.)
- Shanghai Biological Products Research Institute Co., Ltd., State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, Shanghai 200052, China;
| | - Jialu Zhang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, State Key Laboratory of Drug Regulatory Science, Beijing 102629, China; (Y.Z.); (J.Z.)
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Dongmei Li
- Shanghai Biological Products Research Institute Co., Ltd., State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, Shanghai 200052, China;
| | - Qunying Mao
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, State Key Laboratory of Drug Regulatory Science, Beijing 102629, China; (Y.Z.); (J.Z.)
| | - Xiuling Li
- Shanghai Biological Products Research Institute Co., Ltd., State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, Shanghai 200052, China;
| | - Zhenglun Liang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, State Key Laboratory of Drug Regulatory Science, Beijing 102629, China; (Y.Z.); (J.Z.)
| | - Qian He
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, State Key Laboratory of Drug Regulatory Science, Beijing 102629, China; (Y.Z.); (J.Z.)
| |
Collapse
|
32
|
Bartels M, Sala Solé E, Sauerschnig LM, Rijkers GT. Back to the Future: Immune Protection or Enhancement of Future Coronaviruses. Microorganisms 2024; 12:617. [PMID: 38543668 PMCID: PMC10975256 DOI: 10.3390/microorganisms12030617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/14/2024] [Accepted: 03/16/2024] [Indexed: 11/12/2024] Open
Abstract
Before the emergence of SARS-CoV-1, MERS-CoV, and most recently, SARS-CoV-2, four other coronaviruses (the alpha coronaviruses NL63 and 229E and the beta coronaviruses OC43 and HKU1) had already been circulating in the human population. These circulating coronaviruses all cause mild respiratory illness during the winter seasons, and most people are already infected in early life. Could antibodies and/or T cells, especially against the beta coronaviruses, have offered some form of protection against (severe) COVID-19 caused by infection with SARS-CoV-2? Related is the question of whether survivors of SARS-CoV-1 or MERS-CoV would be relatively protected against SARS-CoV-2. More importantly, would humoral and cellular immunological memory generated during the SARS-CoV-2 pandemic, either by infection or vaccination, offer protection against future coronaviruses? Or rather than protection, could antibody-dependent enhancement have taken place, a mechanism by which circulating corona antibodies enhance the severity of COVID-19? Another related phenomenon, the original antigenic sin, would also predict that the effectiveness of the immune response to future coronaviruses would be impaired because of the reactivation of memory against irrelevant epitopes. The currently available evidence indicates that latter scenarios are highly unlikely and that especially cytotoxic memory T cells directed against conserved epitopes of human coronaviruses could at least offer partial protection against future coronaviruses.
Collapse
Affiliation(s)
| | | | | | - Ger T. Rijkers
- Science and Engineering Department, University College Roosevelt, 4331 CB Middelburg, The Netherlands; (M.B.); (E.S.S.); (L.M.S.)
| |
Collapse
|
33
|
Wang F, Yang G, Yan L. Crystal Structures of Fusion Cores from CCoV-HuPn-2018 and SADS-CoV. Viruses 2024; 16:272. [PMID: 38400047 PMCID: PMC10893436 DOI: 10.3390/v16020272] [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: 11/29/2023] [Revised: 12/28/2023] [Accepted: 12/28/2023] [Indexed: 02/25/2024] Open
Abstract
Cross-species spillover to humans of coronaviruses (CoVs) from wildlife animal reservoirs poses marked and global threats to human and animal health. Recently, sporadic infection of canine coronavirus-human pneumonia-2018 (CCoV-HuPn-2018) in hospitalized patients with pneumonia genetically related to canine and feline coronavirus were identified. In addition, swine acute diarrhea syndrome coronavirus (SADS-CoV) had the capability of broad tropism to cultured cells including from humans. Together, the transmission of Alphacoronaviruses that originated in wildlife to humans via intermediate hosts was responsible for the high-impact emerging zoonosis. Entry of CoV is mainly mediated by Spike and formation of a typical six helix bundle (6-HB) structure in the postfusion state of Spike is pivotal. Here, we present the complete fusion core structures of CCoV-HuPn-2018 and SADS-CoV from Alphacoronavirus at 2.10 and 2.59 Å, respectively. The overall structure of the CCoV-HuPn-2018 fusion core is similar to Alphacoronavirus like HCoV-229E, while SADS-CoV is analogous to Betacoronavirus like SARS-CoV-2. Collectively, we provide a structural basis for the development of pan-CoV small molecules and polypeptides based on the HR1-HR2 complex, concerning CCoV-HuPn-2018 and SADS-CoV.
Collapse
Affiliation(s)
- Fulian Wang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai 201210, China;
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai 201210, China
| | - Guang Yang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai 201210, China;
| | - Lei Yan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai 201210, China;
| |
Collapse
|
34
|
Zech F, Jung C, Jacob T, Kirchhoff F. Causes and Consequences of Coronavirus Spike Protein Variability. Viruses 2024; 16:177. [PMID: 38399953 PMCID: PMC10892391 DOI: 10.3390/v16020177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Coronaviruses are a large family of enveloped RNA viruses found in numerous animal species. They are well known for their ability to cross species barriers and have been transmitted from bats or intermediate hosts to humans on several occasions. Four of the seven human coronaviruses (hCoVs) are responsible for approximately 20% of common colds (hCoV-229E, -NL63, -OC43, -HKU1). Two others (SARS-CoV-1 and MERS-CoV) cause severe and frequently lethal respiratory syndromes but have only spread to very limited extents in the human population. In contrast the most recent human hCoV, SARS-CoV-2, while exhibiting intermediate pathogenicity, has a profound impact on public health due to its enormous spread. In this review, we discuss which initial features of the SARS-CoV-2 Spike protein and subsequent adaptations to the new human host may have helped this pathogen to cause the COVID-19 pandemic. Our focus is on host forces driving changes in the Spike protein and their consequences for virus infectivity, pathogenicity, immune evasion and resistance to preventive or therapeutic agents. In addition, we briefly address the significance and perspectives of broad-spectrum therapeutics and vaccines.
Collapse
Affiliation(s)
- Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Christoph Jung
- Institute of Electrochemistry, Ulm University, 89081 Ulm, Germany; (C.J.); (T.J.)
- Helmholtz-Institute Ulm (HIU) Electrochemical Energy Storage, 89081 Ulm, Germany
- Karlsruhe Institute of Technology (KIT), 76021 Karlsruhe, Germany
| | - Timo Jacob
- Institute of Electrochemistry, Ulm University, 89081 Ulm, Germany; (C.J.); (T.J.)
- Helmholtz-Institute Ulm (HIU) Electrochemical Energy Storage, 89081 Ulm, Germany
- Karlsruhe Institute of Technology (KIT), 76021 Karlsruhe, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
35
|
Frumenzio G, Chandramouli B, Besker N, Grottesi A, Talarico C, Frigerio F, Emerson A, Musiani F. Conformational response to ligand binding of TMPRSS2, a protease involved in SARS-CoV-2 infection: Insights through computational modeling. Proteins 2023; 91:1288-1297. [PMID: 37409524 DOI: 10.1002/prot.26548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2023]
Abstract
Thanks to the considerable research which has been undertaken in the last few years to improve our understanding of the biology and mechanism of action of SARS-CoV-2, we know how the virus uses its surface spike protein to infect host cells. The transmembrane prosthesis, serine 2 (TMPRSS2) protein, located on the surface of human cells, recognizes the cleavage site in the spike protein, leading to the release of the fusion peptide and entry of the virus into the host cells. Because of its role, TMPRSS2 has been proposed as a drug target to prevent infection by the virus. In this study, we aim to increase our understanding of TMPRSS2 using long scale microsecond atomistic molecular dynamics simulations, focusing on the conformational changes over time. The comparison between simulations conducted on the protein in the native (apo) and inhibited form (holo), has shown that in the holo form the inhibitor stabilizes the catalytic site and induces rearrangements in the extracellular domain of the protein. In turn, it leads to the formation of a new cavity in the vicinity of the ligand binding pocket that is stable in the microsecond time scale. Given the low specificity of known protease inhibitors, these findings suggest a new potential drug target site that can be used to improve TMPRSS2 specific recognition by newly designed inhibitors.
Collapse
Affiliation(s)
- Giorgia Frumenzio
- Super Computing Applications and Innovation, Department HPC, CINECA, Casalecchio di Reno, Italy
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | | | | | | | | | - Andrew Emerson
- Super Computing Applications and Innovation, Department HPC, CINECA, Casalecchio di Reno, Italy
| | - Francesco Musiani
- Laboratory of Bioinorganic Chemistry, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
36
|
Roggero PF, Calistri A, Palù G. The chaos law is a principal driver of natural selection: A proposition on the evolution of recently emerged coronaviruses. PLoS One 2023; 18:e0290453. [PMID: 37616261 PMCID: PMC10449193 DOI: 10.1371/journal.pone.0290453] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/08/2023] [Indexed: 08/26/2023] Open
Abstract
Here we propose that viruses emerging in the human population undergo an evolution that is conditioned by the rules of chaos. Our data support the notion that the initial growth rate "r" affects the chances of the virus to establish a long-lasting relationship with the new host. Indeed, an emerging virus is able to spread and adapt only when it displays an initial r falling in a range frankly associated with chaotic growth.
Collapse
Affiliation(s)
| | - Arianna Calistri
- Department of Molecular Medicine, University of Padua, via A. Gabelli, Padua, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padua, via A. Gabelli, Padua, Italy
- Italian Medicines Agency, Via del Tritone, Rome, Italy
| |
Collapse
|
37
|
Hulme J. COVID-19 and Diarylamidines: The Parasitic Connection. Int J Mol Sci 2023; 24:6583. [PMID: 37047556 PMCID: PMC10094973 DOI: 10.3390/ijms24076583] [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: 03/15/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
As emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants (Omicron) continue to outpace and negate combinatorial vaccines and monoclonal antibody therapies targeting the spike protein (S) receptor binding domain (RBD), the appetite for developing similar COVID-19 treatments has significantly diminished, with the attention of the scientific community switching to long COVID treatments. However, treatments that reduce the risk of "post-COVID-19 syndrome" and associated sequelae remain in their infancy, particularly as no established criteria for diagnosis currently exist. Thus, alternative therapies that reduce infection and prevent the broad range of symptoms associated with 'post-COVID-19 syndrome' require investigation. This review begins with an overview of the parasitic-diarylamidine connection, followed by the renin-angiotensin system (RAS) and associated angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSSR2) involved in SARS-CoV-2 infection. Subsequently, the ability of diarylamidines to inhibit S-protein binding and various membrane serine proteases associated with SARS-CoV-2 and parasitic infections are discussed. Finally, the roles of diarylamidines (primarily DIZE) in vaccine efficacy, epigenetics, and the potential amelioration of long COVID sequelae are highlighted.
Collapse
Affiliation(s)
- John Hulme
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si 461-701, Republic of Korea
| |
Collapse
|
38
|
Seroprevalence and Risk Factors for Bovine Coronavirus Infection among Dairy Cattle and Water Buffalo in Campania Region, Southern Italy. Animals (Basel) 2023; 13:ani13050772. [PMID: 36899629 PMCID: PMC10000194 DOI: 10.3390/ani13050772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/19/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Cattle and water buffalo are the main livestock species that are raised in the Campania region, southern Italy, and they contribute significantly to the regional rural economy. Currently there are limited data on the prevalence of relevant impact infections, such as bovine coronavirus (BCov), an RNA virus that causes acute enteric and respiratory disease. Although these diseases are described primarily in cattle, there have been reports of spillovers to other ruminants, including water buffalo. Here, we determined the seroprevalence of BCoV in cattle and water buffalo in the Campania region of southern Italy. An overall seroprevalence of 30.8% was determined after testing 720 sampled animals with a commercial enzyme-linked immunosorbent assay. A risk factor analysis revealed that the seropositivity rates in cattle (49.2%) were higher than in water buffalo (5.3%). In addition, higher seroprevalence rates were observed in older and purchased animals. In cattle, housing type and location were not associated with higher seroprevalence. The presence of BCoV antibodies in water buffalo was associated with the practice of co-inhabiting with cattle, demonstrating that this practice is incorrect and promotes the transmission of pathogens between different species. Our study found a considerable seroprevalence, which is consistent with previous research from other countries. Our results provide information on the widespread distribution of this pathogen as well as the risk factors that are involved in its transmission. This information could be useful in the control and surveillance of this infection.
Collapse
|
39
|
Dofuor AK, Quartey NKA, Osabutey AF, Boateng BO, Lutuf H, Osei JHN, Ayivi-Tosuh SM, Aiduenu AF, Ekloh W, Loh SK, Opoku MJ, Aidoo OF. The Global Impact of COVID-19: Historical Development, Molecular Characterization, Drug Discovery and Future Directions. CLINICAL PATHOLOGY (THOUSAND OAKS, VENTURA COUNTY, CALIF.) 2023; 16:2632010X231218075. [PMID: 38144436 PMCID: PMC10748929 DOI: 10.1177/2632010x231218075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/16/2023] [Indexed: 12/26/2023]
Abstract
In December 2019, an outbreak of a respiratory disease called the coronavirus disease 2019 (COVID-19) caused by a new coronavirus known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) began in Wuhan, China. The SARS-CoV-2, an encapsulated positive-stranded RNA virus, spread worldwide with disastrous consequences for people's health, economies, and quality of life. The disease has had far-reaching impacts on society, including economic disruption, school closures, and increased stress and anxiety. It has also highlighted disparities in healthcare access and outcomes, with marginalized communities disproportionately affected by the SARS-CoV-2. The symptoms of COVID-19 range from mild to severe. There is presently no effective cure. Nevertheless, significant progress has been made in developing COVID-19 vaccine for different therapeutic targets. For instance, scientists developed multifold vaccine candidates shortly after the COVID-19 outbreak after Pfizer and AstraZeneca discovered the initial COVID-19 vaccines. These vaccines reduce disease spread, severity, and mortality. The addition of rapid diagnostics to microscopy for COVID-19 diagnosis has proven crucial. Our review provides a thorough overview of the historical development of COVID-19 and molecular and biochemical characterization of the SARS-CoV-2. We highlight the potential contributions from insect and plant sources as anti-SARS-CoV-2 and present directions for future research.
Collapse
Affiliation(s)
- Aboagye Kwarteng Dofuor
- Department of Biological Sciences, School of Natural and Environmental Sciences, University of Environment and Sustainable Development, Somanya, Ghana
| | - Naa Kwarley-Aba Quartey
- Department of Food Science and Technology, Faculty of Biosciences, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | | | - Belinda Obenewa Boateng
- Coconut Research Program, Oil Palm Research Institute, Council for Scientific and Industrial Research, Sekondi-Takoradi, Ghana
| | - Hanif Lutuf
- Crop Protection Division, Oil Palm Research Institute, Council for Scientific and Industrial Research, Kade, Ghana
| | - Joseph Harold Nyarko Osei
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Selina Mawunyo Ayivi-Tosuh
- Department of Biochemistry, School of Life Sciences, Northeast Normal University, Changchun, Jilin Province, China
| | - Albert Fynn Aiduenu
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra, Ghana
| | - William Ekloh
- Department of Biochemistry, School of Biological Sciences, College of Agriculture and Natural Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Seyram Kofi Loh
- Department of Built Environment, School of Sustainable Development, University of Environment and Sustainable Development, Somanya, Ghana
| | - Maxwell Jnr Opoku
- Department of Biological Sciences, School of Natural and Environmental Sciences, University of Environment and Sustainable Development, Somanya, Ghana
| | - Owusu Fordjour Aidoo
- Department of Biological Sciences, School of Natural and Environmental Sciences, University of Environment and Sustainable Development, Somanya, Ghana
| |
Collapse
|
40
|
Antiviral Properties of Pennisetum purpureum Extract against Coronaviruses and Enteroviruses. Pathogens 2022; 11:pathogens11111371. [PMID: 36422622 PMCID: PMC9696772 DOI: 10.3390/pathogens11111371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 11/19/2022] Open
Abstract
Many severe epidemics are caused by enteroviruses (EVs) and coronaviruses (CoVs), including feline coronavirus (FCoV) in cats, epidemic diarrhea disease virus (PEDV) in pigs, infectious bronchitis virus (IBV) in chickens, and EV71 in human. Vaccines and antiviral drugs are used to prevent and treat the infection of EVs and CoVs, but the effectiveness is affected due to rapidly changing RNA viruses. Many plant extracts have been proven to have antiviral properties despite the continuous mutations of viruses. Napier grass (Pennisetum purpureum) has high phenolic content and has been used as healthy food materials, livestock feed, biofuels, and more. This study tested the antiviral properties of P. purpureum extract against FCoV, PEDV, IBV, and EV71 by in vitro cytotoxicity assay, TCID50 virus infection assay, and chicken embryo infection assay. The findings showed that P. purpureum extract has the potential of being disinfectant to limit the spread of CoVs and EVs because the extract can inhibit the infection of EV71, FCoV, and PEDV in cells, and significantly reduce the severity of symptoms caused by IBV in chicken embryos.
Collapse
|