1
|
Iyaniwura SA, Ribeiro RM, Zitzmann C, Phan T, Ke R, Perelson AS. The kinetics of SARS-CoV-2 infection based on a human challenge study. Proc Natl Acad Sci U S A 2024; 121:e2406303121. [PMID: 39508770 DOI: 10.1073/pnas.2406303121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024] Open
Abstract
Studying the early events that occur after viral infection in humans is difficult unless one intentionally infects volunteers in a human challenge study. Here, we use data about severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in such a study in combination with mathematical modeling to gain insights into the relationship between the amount of virus in the upper respiratory tract and the immune response it generates. We propose a set of dynamic models of increasing complexity to dissect the roles of target cell limitation, innate immunity, and adaptive immunity in determining the observed viral kinetics. We introduce an approach for modeling the effect of humoral immunity that describes a decline in infectious virus after immune activation. We fit our models to viral load and infectious titer data from all the untreated infected participants in the study simultaneously. We found that a power-law with a power h < 1 describes the relationship between infectious virus and viral load. Viral replication at the early stage of infection is rapid, with a doubling time of ~2 h for viral RNA and ~3 h for infectious virus. We estimate that adaptive immunity is initiated ~7 to 10 d postinfection and appears to contribute to a multiphasic viral decline experienced by some participants; the viral rebound experienced by other participants is consistent with a decline in the interferon response. Altogether, we quantified the kinetics of SARS-CoV-2 infection, shedding light on the early dynamics of the virus and the potential role of innate and adaptive immunity in promoting viral decline during infection.
Collapse
Affiliation(s)
- Sarafa A Iyaniwura
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Ruy M Ribeiro
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Carolin Zitzmann
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Tin Phan
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Ruian Ke
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| | - Alan S Perelson
- Theoretical Division, Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545
| |
Collapse
|
2
|
Majolo JH, Gonçalves JIB, Souza RP, González LC, Sperotto N, Silveira MD, Oliveira SD, Bizarro CV, Machado P, Basso LA, Souza APD, Oliveira JR, Ferreira CAS. Losartan and enalapril maleate differently influence SARS-CoV-2-infected vero cells. Sci Rep 2024; 14:24801. [PMID: 39433817 PMCID: PMC11493994 DOI: 10.1038/s41598-024-76657-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic has posed significant challenges to global healthcare systems, particularly impacting individuals with pre-existing conditions like hypertension. This study sought to assess the impact of the antihypertensive medications, losartan and enalapril maleate on SARS-CoV-2 infected cells. Vero E6 cells were infected and treated in vitro, evaluating cell viability via the MTT colorimetric assay. Additionally, the study measured relative levels of viral RNA and selected gene messenger RNAs using reverse transcriptase followed by quantitative real-time polymerase chain reaction. RESULTS The findings revealed that losartan substantially reduced nucleocapsid RNA levels of SARS-CoV-2 to nearly undetectable levels, while enalapril maleate did not demonstrate a significant effect. In response to viral infection, the expression of il-18, p53, p21, and p62 increased compared to uninfected-untreated cells. Notably, il-6 expression was upregulated by both infection and treatments. A comparison between infected cells treated with losartan or enalapril maleate highlighted the presence of distinct profiles in the expression of il-6, p53, p21, and p62. CONCLUSIONS The data from our study suggest that these medications could interfere with certain effects triggered by SARS-CoV-2 infection in Vero E6 cells. However, their influence appears to vary both quantitatively and qualitatively in the modulation of metabolic and signal transduction pathways.
Collapse
Affiliation(s)
- Julia H Majolo
- Laboratory of Immunology and Microbiology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - João I B Gonçalves
- Laboratory of Clinical and Experimental Immunology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Renata P Souza
- Laboratory of Immunology and Microbiology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Laura C González
- Research Center on Molecular and Functional Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Nathalia Sperotto
- Research Center on Molecular and Functional Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Maiele D Silveira
- Research Center on Molecular and Functional Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Sílvia D Oliveira
- Laboratory of Immunology and Microbiology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Cristiano V Bizarro
- Research Center on Molecular and Functional Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Pablo Machado
- Research Center on Molecular and Functional Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Luiz A Basso
- Research Center on Molecular and Functional Biology (CPBMF), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Ana P D Souza
- Laboratory of Clinical and Experimental Immunology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil
| | - Jarbas R Oliveira
- Laboratory of Cellular Biophysics and Inflammation, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil.
| | - Carlos A S Ferreira
- Laboratory of Immunology and Microbiology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, Porto Alegre, 90619-900, Rio Grande do Sul, Brazil.
| |
Collapse
|
3
|
Cabel CR, Guzman BA, Alizadeh E, Li S, Holberg C, Wichaidit C, Cusanovich DA, Paek AL, Thatcher GRJ, Doorslaer KV, Nargi RS, Sutton RE, Suryadevara N, Crowe JE, Carnahan RH, Campos SK, Thorne CA. Cell-based high-content approach for SARS-CoV-2 neutralization identifies unique monoclonal antibodies and PI3K pathway inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.04.616743. [PMID: 39416139 PMCID: PMC11483034 DOI: 10.1101/2024.10.04.616743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The sudden rise of the SARS-CoV-2 virus and the delay in the development of effective therapeutics to mitigate it made evident a need for ways to screen for compounds that can block infection and prevent further pathogenesis and spread. Yet, identifying effective drugs efficacious against viral infection and replication with minimal toxicity for the patient can be difficult. Monoclonal antibodies were shown to be effective, yet as the SARS-CoV-2 mutated, these antibodies became ineffective. Small molecule antivirals were identified using pseudovirus constructs to recapitulate infection in non-human cells, such as Vero E6 cells. However, the impact was limited due to poor translation of these compounds in the clinical setting. This is partly due to the lack of similarity of screening platforms to the in vivo physiology of the patient and partly because drugs effective in vitro showed dose-limiting toxicities. In this study, we performed two high-throughput screens in human lung adenocarcinoma cells with authentic SARS-CoV-2 virus to identify both monoclonal antibodies that neutralize the virus and clinically useful kinase inhibitors to block the virus and prioritize minimal host toxicity. Using high-content imaging combined with single-cell and multidimensional analysis, we identified antibodies and kinase inhibitors that reduce virus infection without affecting the host. Our screening technique uncovered novel antibodies and overlooked kinase inhibitors (i.e. PIK3i, mTORi, multiple RTKi) that could be effective against SARS-CoV-2 virus. Further characterization of these molecules will streamline the repurposing of compounds for the treatment of future pandemics and uncover novel mechanisms viruses use to hijack and infect host cells.
Collapse
|
4
|
Nazir F, John Kombe Kombe A, Khalid Z, Bibi S, Zhang H, Wu S, Jin T. SARS-CoV-2 replication and drug discovery. Mol Cell Probes 2024; 77:101973. [PMID: 39025272 DOI: 10.1016/j.mcp.2024.101973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
The coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has killed millions of people and continues to wreak havoc across the globe. This sudden and deadly pandemic emphasizes the necessity for anti-viral drug development that can be rapidly administered to reduce morbidity, mortality, and virus propagation. Thus, lacking efficient anti-COVID-19 treatment, and especially given the lengthy drug development process as well as the critical death tool that has been associated with SARS-CoV-2 since its outbreak, drug repurposing (or repositioning) constitutes so far, the ideal and ready-to-go best approach in mitigating viral spread, containing the infection, and reducing the COVID-19-associated death rate. Indeed, based on the molecular similarity approach of SARS-CoV-2 with previous coronaviruses (CoVs), repurposed drugs have been reported to hamper SARS-CoV-2 replication. Therefore, understanding the inhibition mechanisms of viral replication by repurposed anti-viral drugs and chemicals known to block CoV and SARS-CoV-2 multiplication is crucial, and it opens the way for particular treatment options and COVID-19 therapeutics. In this review, we highlighted molecular basics underlying drug-repurposing strategies against SARS-CoV-2. Notably, we discussed inhibition mechanisms of viral replication, involving and including inhibition of SARS-CoV-2 proteases (3C-like protease, 3CLpro or Papain-like protease, PLpro) by protease inhibitors such as Carmofur, Ebselen, and GRL017, polymerases (RNA-dependent RNA-polymerase, RdRp) by drugs like Suramin, Remdesivir, or Favipiravir, and proteins/peptides inhibiting virus-cell fusion and host cell replication pathways, such as Disulfiram, GC376, and Molnupiravir. When applicable, comparisons with SARS-CoV inhibitors approved for clinical use were made to provide further insights to understand molecular basics in inhibiting SARS-CoV-2 replication and draw conclusions for future drug discovery research.
Collapse
Affiliation(s)
- Farah Nazir
- Center of Disease Immunity and Investigation, College of Medicine, Lishui University, Lishui, 323000, China
| | - Arnaud John Kombe Kombe
- Laboratory of Structural Immunology, Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Zunera Khalid
- Laboratory of Structural Immunology, Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Shaheen Bibi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Anhui, China
| | - Hongliang Zhang
- Center of Disease Immunity and Investigation, College of Medicine, Lishui University, Lishui, 323000, China
| | - Songquan Wu
- Center of Disease Immunity and Investigation, College of Medicine, Lishui University, Lishui, 323000, China.
| | - Tengchuan Jin
- Center of Disease Immunity and Investigation, College of Medicine, Lishui University, Lishui, 323000, China; Laboratory of Structural Immunology, Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Anhui, China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science & Technology of China, Hefei, 230027, China; Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
5
|
Baran J, Kuryk Ł, Garofalo M, Pancer K, Wieczorek M, Kazek M, Staniszewska M. Induction of an immune response by a nonreplicating adenoviruses-based formulation versus a commercial pseudo-SARS-CoV-2 vaccine. BIOTECHNOLOGIA 2024; 105:263-272. [PMID: 39439719 PMCID: PMC11492889 DOI: 10.5114/bta.2024.141805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/17/2024] [Accepted: 06/04/2024] [Indexed: 10/25/2024] Open
Abstract
Screening for effective vaccines requires broad studies on their immunogenicity in vitro and ex vivo . We used a PBMC-based system to assess changes in CD4+ T cells, CD8+ T cells, and CD19+ B cells upon stimulation with different combinations of antigens and adjuvants. We studied the activation mechanism using flow cytometry and two different adenoviral adjuvants characterized by the presence or absence of costimulatory ligands for the ICOS and CD40 receptors. Our studies identified the cellular targets and molecular mechanisms driving ongoing switched-antibody diversification. Class-switched memory B cells were the main precursor cells (95.03% ± 0.38 vs. mock 82.33% ± 0.45, P < 0.05) after treatment with the immunogenic formula: adenovirus armed (MIX1) or not (MIX2) with the ICOS and CD40 ligand, the recombinant receptor binding domain (rRBD), and Lentifect™ SARS-CoV-2 spike-pseudotyped lentivirus (GeneCopoeia, USA). Bcell class-switching towards the IgG+IgM+- positive phenotypes was noted (~50-fold increase vs. mock, P < 0.05). A significant increase was observed in the CD8+TEM population of the MIX1 (~2-fold, P < 0.05) and MIX2 (~4.7-fold, P < 0.05) treated samples. CD8+TEMRA increased when PBMCs were treated with MIX2 (9.63% ± 0.90, P < 0.05) vs. mock (2.63% ± 1.96). Class-switched memory B cells were the dominant antigen-specific cells in primary reactions. We indicated a correlation between the protection offered by vaccine regimens and their ability to induce high frequencies of multifunctional T cells.
Collapse
Affiliation(s)
- Joanna Baran
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warszawa, Poland
| | - Łukasz Kuryk
- National Institute of Public Health National Institute of Hygiene (NIH) – National Institute of Research, Warszawa, Poland
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Katarzyna Pancer
- National Institute of Public Health National Institute of Hygiene (NIH) – National Institute of Research, Warszawa, Poland
| | - Magdalena Wieczorek
- National Institute of Public Health National Institute of Hygiene (NIH) – National Institute of Research, Warszawa, Poland
| | - Michalina Kazek
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warszawa, Poland
| | - Monika Staniszewska
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warszawa, Poland
| |
Collapse
|
6
|
O’Reilly S, Byrne J, Feeney ER, Mallon PWG, Gautier V. Navigating the Landscape of B Cell Mediated Immunity and Antibody Monitoring in SARS-CoV-2 Vaccine Efficacy: Tools, Strategies and Clinical Trial Insights. Vaccines (Basel) 2024; 12:1089. [PMID: 39460256 PMCID: PMC11511438 DOI: 10.3390/vaccines12101089] [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: 07/31/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 10/28/2024] Open
Abstract
Correlates of Protection (CoP) are biomarkers above a defined threshold that can replace clinical outcomes as primary endpoints, predicting vaccine effectiveness to support the approval of new vaccines or follow up studies. In the context of COVID-19 vaccination, CoPs can help address challenges such as demonstrating vaccine effectiveness in special populations, against emerging SARS-CoV-2 variants or determining the durability of vaccine-elicited immunity. While anti-spike IgG titres and viral neutralising capacity have been characterised as CoPs for COVID-19 vaccination, the contribution of other components of the humoral immune response to immediate and long-term protective immunity is less well characterised. This review examines the evidence supporting the use of CoPs in COVID-19 clinical vaccine trials, and how they can be used to define a protective threshold of immunity. It also highlights alternative humoral immune biomarkers, including Fc effector function, mucosal immunity, and the generation of long-lived plasma and memory B cells and discuss how these can be applied to clinical studies and the tools available to study them.
Collapse
Affiliation(s)
- Sophie O’Reilly
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Joanne Byrne
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eoin R. Feeney
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Elm Park, Dublin 4, Ireland
| | - Patrick W. G. Mallon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Department of Infectious Diseases, St Vincent’s University Hospital, Elm Park, Dublin 4, Ireland
| | - Virginie Gautier
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Belfield, Dublin 4, Ireland
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
7
|
Hsu JC, Saenkham-Huntsinger P, Huang P, Octaviani CP, Drelich AK, Peng BH, Tseng CTK. Characterizing neuroinvasion and neuropathology of SARS-CoV-2 by using AC70 human ACE2 transgenic mice. Front Microbiol 2024; 15:1455462. [PMID: 39380676 PMCID: PMC11458418 DOI: 10.3389/fmicb.2024.1455462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024] Open
Abstract
COVID-19 presents with a plethora of neurological signs and symptoms despite being characterized as a respiratory disease, including seizures, anxiety, depression, amnesia, attention deficits, and alterations in consciousness. The olfactory nerve is widely accepted as the neuroinvasive route by which the etiological agent SARS-CoV-2 enters the brain, but the trigeminal nerve is an often-overlooked additional route. Based on this consensus, we initially conducted a pilot experiment investigating the olfactory nerve route of SARS-CoV-2 neuroinvasion via intranasal inoculation in AC70 human ACE2 transgenic mice. Notably, we found that the trigeminal ganglion is an early and highly efficient site of viral replication, which then rapidly spread widely throughout the brain where neurons were primarily targeted. Despite the extensive viral infection across the brain, obvious evidence of tissue pathology including inflammatory infiltration, glial activation, and apoptotic cell deaths were not consistently observed, albeit inflammatory cytokines were significantly induced. However, the expression levels of different genes related to neuronal function, including the neurotransmitter dopamine pathway as well as synaptic function, and markers of neuronal damage were altered as compared to mock-infected mice. Our findings suggest that the trigeminal nerve may serve as a neuroinvasive route complementary to the olfactory nerve and that the ensuing neuroinvasion presented a unique neuropathological profile. This study provides insights into potential neuropathogenic mechanisms utilized by coronaviruses.
Collapse
Affiliation(s)
- Jason C. Hsu
- Department of Biochemistry, Cell & Molecular Biology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Panatda Saenkham-Huntsinger
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Pinghan Huang
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Cassio Pontes Octaviani
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Aleksandra K. Drelich
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Bi-Hung Peng
- Department of Neuroscience, Cell Biology, & Anatomy, The University of Texas Medical Branch, Galveston, TX, United States
| | - Chien-Te K. Tseng
- Department of Biochemistry, Cell & Molecular Biology, The University of Texas Medical Branch, Galveston, TX, United States
- Department of Microbiology & Immunology, The University of Texas Medical Branch, Galveston, TX, United States
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
8
|
Sugimoto S, Kawase M, Suwa R, Kume Y, Chishiki M, Ono T, Okabe H, Norito S, Hanaki KI, Hosoya M, Hashimoto K, Shirato K. Comparison of mutations in human parainfluenza viruses during passage in primary human bronchial/tracheal epithelial air-liquid interface cultures and cell lines. Microbiol Spectr 2024; 12:e0116424. [PMID: 39078148 PMCID: PMC11370246 DOI: 10.1128/spectrum.01164-24] [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: 05/09/2024] [Accepted: 05/31/2024] [Indexed: 07/31/2024] Open
Abstract
Human parainfluenza virus (HPIV) causes respiratory infections, which are exacerbated in children and older people. Correct evaluation of viral characteristics is essential for the study of countermeasures. However, adaptation of viruses to cultured cells during isolation or propagation might select laboratory passage-associated mutations that modify the characteristics of the virus. It was previously reported that adaptation of HPIV3, but not other HPIVs, was avoided in human airway epithelia. To examine the influence of laboratory passage on the genomes of HPIV1-HPIV4, we evaluated the occurrence of mutations after passage in primary human bronchial/tracheal epithelial cell air-liquid interface (HBTEC-ALI) culture and conventional cultured cells (Vero cells expressing the transmembrane protease, serine 2, and normal Vero cells). The occurrence of mutations was significantly lower in HBTEC-ALI than in conventional culture. In HBTEC-ALI culture, most of the mutations were silent or remained at low variant frequency, resulting in less impact on the viral consensus sequence. In contrast, passage in conventional culture induced or selected genetic mutations at high frequency with passage-associated unique substitutions. High mutagenesis of hemagglutinin-neuraminidase was commonly observed in all four HPIVs, and mutations even occurred in a single passage. In addition, in HPIV1 and HPIV2, mutations in the large protein were more frequent. These results indicate that passage in HBTEC-ALI culture is more suitable than conventional culture for maintaining the original characteristics of clinical isolates in all four HPIVs, which can help with the understanding of viral pathogenesis. IMPORTANCE Adaptation of viruses to cultured cells can increase the risk of misinterpretation in virological characterization of clinical isolates. In human parainfluenza virus (HPIV) 3, it has been reported that the human airway epithelial and lung organoid models are preferable for the study of viral characteristics of clinical strains without mutations. Therefore, we analyzed clinical isolates of all four HPIVs for the occurrence of mutations after five laboratory passages in human bronchial/tracheal epithelial cell air-liquid interface (HBTEC-ALI) or conventional culture. We found a high risk of hemagglutinin-neuraminidase mutagenesis in all four HPIVs in conventional cultured cells. In addition, in HPIV1 and HPIV2, mutations of the large protein were also more frequent in conventional cultured cells than in HBTEC-ALI culture. HBTEC-ALI culture was useful for maintaining the original sequence and characteristics of clinical isolates in all four HPIVs. The present study contributes to the understanding of HPIV pathogenesis and antiviral strategies.
Collapse
Affiliation(s)
- Satoko Sugimoto
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
- Research Center for Biosafety, Laboratory Animal, and Pathogen Bank, National Institute of Infectious Diseases, Tokyo, Japan
| | - Miyuki Kawase
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| | - Reiko Suwa
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yohei Kume
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Mina Chishiki
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Takashi Ono
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Hisao Okabe
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Sakurako Norito
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Ken-Ichi Hanaki
- Research Center for Biosafety, Laboratory Animal, and Pathogen Bank, National Institute of Infectious Diseases, Tokyo, Japan
| | - Mitsuaki Hosoya
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Koichi Hashimoto
- Department of Pediatrics, School of Medicine, Fukushima Medical University, Fukushima, Japan
| | - Kazuya Shirato
- Department of Virology III, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
9
|
Atemin A, Ivanova A, Peppel W, Stamatov R, Gallegos R, Durden H, Uzunova S, Vershinin MD, Saffarian S, Stoynov SS. Kinetic Landscape of Single Virus-like Particles Highlights the Efficacy of SARS-CoV-2 Internalization. Viruses 2024; 16:1341. [PMID: 39205315 PMCID: PMC11359012 DOI: 10.3390/v16081341] [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: 06/26/2024] [Revised: 07/22/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
The efficiency of virus internalization into target cells is a major determinant of infectivity. SARS-CoV-2 internalization occurs via S-protein-mediated cell binding followed either by direct fusion with the plasma membrane or endocytosis and subsequent fusion with the endosomal membrane. Despite the crucial role of virus internalization, the precise kinetics of the processes involved remains elusive. We developed a pipeline, which combines live-cell microscopy and advanced image analysis, for measuring the rates of multiple internalization-associated molecular events of single SARS-CoV-2-virus-like particles (VLPs), including endosome ingression and pH change. Our live-cell imaging experiments demonstrate that only a few minutes after binding to the plasma membrane, VLPs ingress into RAP5-negative endosomes via dynamin-dependent scission. Less than two minutes later, VLP speed increases in parallel with a pH drop below 5, yet these two events are not interrelated. By co-imaging fluorescently labeled nucleocapsid proteins, we show that nucleocapsid release occurs with similar kinetics to VLP acidification. Neither Omicron mutations nor abrogation of the S protein polybasic cleavage site affected the rate of VLP internalization, indicating that they do not confer any significant advantages or disadvantages during this process. Finally, we observe that VLP internalization occurs two to three times faster in VeroE6 than in A549 cells, which may contribute to the greater susceptibility of the former cell line to SARS-CoV-2 infection. Taken together, our precise measurements of the kinetics of VLP internalization-associated processes shed light on their contribution to the effectiveness of SARS-CoV-2 propagation in cells.
Collapse
Affiliation(s)
- Aleksandar Atemin
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| | - Aneliya Ivanova
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| | - Wiley Peppel
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| | - Rumen Stamatov
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| | - Rodrigo Gallegos
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| | - Haley Durden
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
| | - Sonya Uzunova
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| | - Michael D. Vershinin
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Saveez Saffarian
- Department of Physics and Astronomy, University of Utah, Salt Lake City, UT 84112, USA; (W.P.); (R.G.); (H.D.)
- Center for Cell and Genome Science, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Stoyno S. Stoynov
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str., 1113 Sofia, Bulgaria; (A.A.); (A.I.); (R.S.); (S.U.)
| |
Collapse
|
10
|
Kocas M, Comoglu T, Ozkul A. Development and in vitro antiviral activity of ivermectin liposomes as a potential drug carrier system. Arch Pharm (Weinheim) 2024; 357:e2300708. [PMID: 38702288 DOI: 10.1002/ardp.202300708] [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: 12/04/2023] [Revised: 03/12/2024] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
This study aimed to assess and compare diverse formulations of ivermectin-loaded liposomes, employing lipid film hydration and ethanol injection methods. Three lipids (DOPC, SPC, and DSPC) were used in predetermined molar ratios. A total of 18 formulations were created, and a factorial design determined the optimal formulation based on particle size, polydispersity index (PDI), zeta potential, and encapsulation efficiency. The average mean particle size, PDI and zeta potential of the selected formulations (F1, F2, F7, F9, and F11) was, respectively, 196.40 ± 44.60 nm, 0.39 ± 0.09, and -40.24 ± 9.17. The encapsulation efficiency exceeded 80%, with a mean loading capacity of 4.00 ± 1.70%. In vitro studies included transmission electron microscopy, Fourier transform infrared spectroscopy, drug release, and antiviral activity assessments against SARS-CoV-2. The liposomal formulations demonstrated superior antiviral activity compared to free ivermectin, as indicated by lower IC50 values. The results of this study emphasize the effectiveness of ivermectin-loaded liposomes in inhibiting viral activity, highlighting their potential as promising candidates for antiviral therapy. The findings suggest that the strategic use of liposomes as drug carriers can significantly modulate and improve the antiviral properties of ivermectin, offering a novel approach to harnessing its full therapeutic potential. Collectively, these results provide a robust foundation for further exploration of ivermectin as a viral protection tool and optimization of its delivery mechanisms.
Collapse
Affiliation(s)
- Meryem Kocas
- Department of Pharmaceutical Technology, Selcuk University Faculty of Pharmacy, Konya, Turkey
- Graduate School of Health Sciences, Ankara University, Ankara, Turkey
- Department of Pharmaceutical Technology, Ankara University Faculty of Pharmacy, Ankara, Turkey
| | - Tansel Comoglu
- Department of Pharmaceutical Technology, Ankara University Faculty of Pharmacy, Ankara, Turkey
| | - Aykut Ozkul
- Department of Virology, Ankara University Faculty of Veterinary Medicine, Ankara, Turkey
| |
Collapse
|
11
|
Mittal RK, Purohit P, Sankaranarayanan M, Muzaffar-Ur-Rehman M, Taramelli D, Signorini L, Dolci M, Basilico N. In-vitro antiviral activity and in-silico targeted study of quinoline-3-carboxylate derivatives against SARS-Cov-2 isolate. Mol Divers 2024; 28:2651-2665. [PMID: 37480422 DOI: 10.1007/s11030-023-10703-w] [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: 06/07/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
In recent years, the viral outbreak named COVID-19 showed that infectious diseases have a huge impact on both global health and the financial and economic sectors. The lack of efficacious antiviral drugs worsened the health problem. Based on our previous experience, we investigated in vitro and in silico a series of quinoline-3-carboxylate derivatives against a SARS-CoV-2 isolate. In the present study, the in-vitro antiviral activity of a series of quinoline-3-carboxylate compounds and the in silico target-based molecular dynamics (MD) and metabolic studies are reported. The compounds' activity against SARS-CoV-2 was evaluated using plaque assay and RT-qPCR. Moreover, from the docking scores, it appears that the most active compounds (1j and 1o) exhibit stronger binding affinity to the primary viral protease (NSP5) and the exoribonuclease domain of non structural protein 14 (NSP14). Additionally, the in-silico metabolic analysis of 1j and 1o defines CYP2C9 and CYP3A4 as the major P450 enzymes involved in their metabolism.
Collapse
Affiliation(s)
- Ravi Kumar Mittal
- National Institute of Pharmaceutical Education and Research, S A S Nagar Mohali, Punjab, 160062, India
- Galgotias College of Pharmacy, Greater Noida, UttarPradesh, India
| | - Priyank Purohit
- School of Pharmacy, Graphic Era Hill University, Dehradun, Uttarakhand, 248002, India.
| | - Murugesan Sankaranarayanan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, BITS Pilani, Pilani Campus, Pilani, Rajasthan, 333031, India
| | - Mohammed Muzaffar-Ur-Rehman
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, BITS Pilani, Pilani Campus, Pilani, Rajasthan, 333031, India
| | - Donatella Taramelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Pascal Street 36, 20133, Milan, Italy
| | - Lucia Signorini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Pascal Street 36, 20133, Milan, Italy
| | - Maria Dolci
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Pascal Street 36, 20133, Milan, Italy
| | - Nicoletta Basilico
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Pascal Street 36, 20133, Milan, Italy
| |
Collapse
|
12
|
Eren ZB, Vatansever C, Kabadayı B, Haykar B, Kuloğlu ZE, Ay S, Nurlybayeva K, Eyikudamacı G, Barlas T, Palaoğlu E, Beşli Y, Kuşkucu MA, Ergönül Ö, Can F. Surveillance of respiratory viruses by aerosol screening in indoor air as an early warning system for epidemics. ENVIRONMENTAL MICROBIOLOGY REPORTS 2024; 16:e13303. [PMID: 38982659 PMCID: PMC11233404 DOI: 10.1111/1758-2229.13303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 05/15/2024] [Indexed: 07/11/2024]
Abstract
The development of effective methods for the surveillance of seasonal respiratory viruses is required for the timely management of outbreaks. We aimed to survey Influenza-A, Influenza-B, RSV-A, Rhinovirus and SARS-CoV-2 surveillance in a tertiary hospital and a campus over 5 months. The effectiveness of air screening as an early warning system for respiratory viruses was evaluated in correlation with respiratory tract panel test results. The overall viral positivity was higher on the campus than in the hospital (55.0% vs. 38.0%). Influenza A was the most prevalent pathogen in both locations. There were two influenza peaks (42nd and 49th weeks) in the hospital air, and a delayed peak was detected on campus in the 1st-week of January. Panel tests indicated a high rate of Influenza A in late December. RSV-A-positivity was higher on the campus than the hospital (21.6% vs. 7.4%). Moreover, we detected two RSV-A peaks in the campus air (48th and 51st weeks) but only one peak in the hospital and panel tests (week 49). Although rhinovirus was the most common pathogen in panel tests, rhinovirus positivity was low in air samples. The air screening for Influenza-B and SARS-Cov-2 revealed comparable positivity rates with panel tests. Air screening can be integrated into surveillance programs to support infection control programs for potential epidemics of respiratory virus infections except for rhinoviruses.
Collapse
Affiliation(s)
| | - Cansel Vatansever
- Koç University İşBank Center for Infectious Diseases (KUISCID)IstanbulTurkey
| | | | | | - Zeynep Ece Kuloğlu
- Koç University İşBank Center for Infectious Diseases (KUISCID)IstanbulTurkey
- Koç UniversityGraduate School of Health SciencesIstanbulTurkey
| | - Sedat Ay
- Koç University School of MedicineIstanbulTurkey
| | | | - Gül Eyikudamacı
- Koç University İşBank Center for Infectious Diseases (KUISCID)IstanbulTurkey
- Koç UniversityGraduate School of Health SciencesIstanbulTurkey
| | - Tayfun Barlas
- Koç University İşBank Center for Infectious Diseases (KUISCID)IstanbulTurkey
| | - Erhan Palaoğlu
- Department of Clinical LaboratoryAmerican HospitalIstanbulTurkey
| | - Yeşim Beşli
- Department of Clinical LaboratoryAmerican HospitalIstanbulTurkey
| | - Mert Ahmet Kuşkucu
- Koç University İşBank Center for Infectious Diseases (KUISCID)IstanbulTurkey
- Department of Medical MicrobiologyKoç University School of MedicineIstanbulTurkey
| | - Önder Ergönül
- Koç University İşBank Center for Infectious Diseases (KUISCID)IstanbulTurkey
- Department of Infectious Disease and Clinical MicrobiologyKoç University School of MedicineIstanbulTurkey
| | - Fusun Can
- Koç University İşBank Center for Infectious Diseases (KUISCID)IstanbulTurkey
- Department of Medical MicrobiologyKoç University School of MedicineIstanbulTurkey
| |
Collapse
|
13
|
Chintapula U, Karim SU, Iyer PR, Asokan-Sheeja H, Neupane B, Nazneen F, Dong H, Bai F, Nguyen KT. A novel nanocomposite drug delivery system for SARS-CoV-2 infections. NANOSCALE ADVANCES 2024; 6:3747-3758. [PMID: 39050946 PMCID: PMC11265598 DOI: 10.1039/d4na00361f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 07/27/2024]
Abstract
To develop an inhalable drug delivery system, we synthesized poly (lactic-co-glycolic acid) nanoparticles with Remdesivir (RDV NPs) as an antiviral agent against SARS-CoV-2 replication and formulated Remdesivir-loaded nanocomposites (RDV NCs) via coating of RDV NPs with novel supramolecular cell-penetrating peptide nanofibers (NFs) to enhance cellular uptake and intracellular drug delivery. RDV NPs and RDV NCs were characterized using variou techniques, including Transmission Electron Microscopy (TEM), Dynamic Light Scattering (DLS), and fluorescent microscopy. The cytotoxicity of RDV NCs was assessed in Vero E6 cells and primary human lung epithelial cells, with no significant cytotoxicity observed up to 1000 μg mL-1 and 48 h. RDV NCs were spherically shaped with a size range of 200-300 nm and a zeta potential of ∼+31 mV as well as indicating the presence of coated nanofibers. Reverse Transcription-quantitative Polymerase Chain Reaction (RT-qPCR), immunofluorescence and plaque assays of SARS-CoV-2 infected Vero E6 treated with RDV NCs showed significantly higher antiviral activities compared to those of free drug and uncoated RDV NPs. RDV NCs exhibited high antiviral activity against SARS-CoV-2, and the nanocomposite platform has the potential to be developed into an inhalable drug delivery system for other viral infections in the lungs.
Collapse
Affiliation(s)
- Uday Chintapula
- Department of Bioengineering, University of Texas at Arlington Arlington TX 76010 USA
| | - Shazeed-Ul Karim
- Department of Cell and Molecular Biology, University of Southern Mississippi Hattiesburg MS 39406 USA
| | | | - Haritha Asokan-Sheeja
- Department of Chemistry and Biochemistry, University of Texas at Arlington Arlington TX 76010 USA
| | - Biswas Neupane
- Department of Cell and Molecular Biology, University of Southern Mississippi Hattiesburg MS 39406 USA
| | - Farzana Nazneen
- Department of Cell and Molecular Biology, University of Southern Mississippi Hattiesburg MS 39406 USA
| | - He Dong
- Department of Chemistry and Biochemistry, University of Texas at Arlington Arlington TX 76010 USA
| | - Fengwei Bai
- Department of Cell and Molecular Biology, University of Southern Mississippi Hattiesburg MS 39406 USA
| | - Kytai T Nguyen
- Department of Bioengineering, University of Texas at Arlington Arlington TX 76010 USA
| |
Collapse
|
14
|
Phadke KS, Higdon NBA, Bellaire BH. In vitro comparison of viral replication and cytopathology induced by SARS-CoV-2 variants. Access Microbiol 2024; 6:000716.v3. [PMID: 39130731 PMCID: PMC11316578 DOI: 10.1099/acmi.0.000716.v3] [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: 10/04/2023] [Accepted: 05/28/2024] [Indexed: 08/13/2024] Open
Abstract
A myriad of coronaviruses cause diseases from a common cold to severe lung infections and pneumonia. SARS-CoV-2 was discovered to be the etiologic agent of the Coronavirus pandemic and many laboratory techniques were examined for virus culture and basic and applied research. Understanding the replication kinetics and characterizing the effect the virus has on different cell lines is crucial for developing in vitro studies. With the emergence of multiple variants of SARS-CoV-2, a comparison between their infectivity and replication in common cell lines will help give us a clear understanding of their characteristic differences in pathogenicity. In this study we compared the cytopathic effect and replication of Wild-Type (USA/WA1), Omicron (B.1.1.529), and Delta (B.1.617.2) variants on five different cell lines; VeroE6, VeroE6 cells expressing high endogenous ACE2, VeroE6 cells expressing human ACE2 and TMPRSS2, Calu3 cells highly expressing human ACE2 and A549 cells. This data will aid researchers with experimental planning and viral pathogenicity analysis and provide a baseline for testing any future variants.
Collapse
Affiliation(s)
- Kruttika S. Phadke
- Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames 50011, USA
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames 50011, USA
| | - Nathaniel B. A. Higdon
- Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames 50011, USA
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames 50011, USA
| | - Bryan H. Bellaire
- Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames 50011, USA
- Interdepartmental Microbiology Graduate Program, Iowa State University, Ames 50011, USA
| |
Collapse
|
15
|
Yang JH, Oh Y, Moon SH, Lee GH, Kim JY, Shin YK, Tark D, Cho HS. Suspected Human-to-Cat Spillover of SARS-CoV-2 Omicron Variant in South Korea. Viruses 2024; 16:1113. [PMID: 39066274 PMCID: PMC11281702 DOI: 10.3390/v16071113] [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: 06/25/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
This retrospective study reports the isolation and characterization of Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) from a household cat in South Korea. The cat, which was presented with respiratory symptoms, was identified during a retrospective analysis of samples collected between April 2021 and March 2022. Genomic sequencing revealed that the isolated virus belonged to the Omicron variant (BA.1), coinciding with its global emergence in early 2022. This case study provides evidence for the potential of direct human-to-cat transmission of the Omicron variant in South Korea during its period of widespread circulation. Our findings underscore the importance of continuous monitoring of SARS-CoV-2 in both human and animal populations to track viral evolution and potential spillover events.
Collapse
Affiliation(s)
- Ju-Hee Yang
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 545431, Republic of Korea; (J.-H.Y.); (G.-H.L.)
| | - Yeonsu Oh
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea;
| | - Sung-Hyun Moon
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea;
| | - Gun-Hee Lee
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 545431, Republic of Korea; (J.-H.Y.); (G.-H.L.)
| | - Jae-Young Kim
- Tae Neung Animal Hospital, Seoul 02033, Republic of Korea;
| | - Yeon-Kyung Shin
- Foreign Animal Disease Division, Animal and Plant Quarantine Agency, Gimcheon 39660, Republic of Korea;
| | - Dongseob Tark
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan 545431, Republic of Korea; (J.-H.Y.); (G.-H.L.)
| | - Ho-Seong Cho
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea;
| |
Collapse
|
16
|
Wei L, Song L, Dunker AK, Foster JA, Uversky VN, Goh GKM. A Comparative Experimental and Computational Study on the Nature of the Pangolin-CoV and COVID-19 Omicron. Int J Mol Sci 2024; 25:7537. [PMID: 39062780 PMCID: PMC11277539 DOI: 10.3390/ijms25147537] [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: 05/17/2024] [Revised: 06/28/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
The relationship between pangolin-CoV and SARS-CoV-2 has been a subject of debate. Further evidence of a special relationship between the two viruses can be found by the fact that all known COVID-19 viruses have an abnormally hard outer shell (low M disorder, i.e., low content of intrinsically disordered residues in the membrane (M) protein) that so far has been found in CoVs associated with burrowing animals, such as rabbits and pangolins, in which transmission involves virus remaining in buried feces for a long time. While a hard outer shell is necessary for viral survival, a harder inner shell could also help. For this reason, the N disorder range of pangolin-CoVs, not bat-CoVs, more closely matches that of SARS-CoV-2, especially when Omicron is included. The low N disorder (i.e., low content of intrinsically disordered residues in the nucleocapsid (N) protein), first observed in pangolin-CoV-2017 and later in Omicron, is associated with attenuation according to the Shell-Disorder Model. Our experimental study revealed that pangolin-CoV-2017 and SARS-CoV-2 Omicron (XBB.1.16 subvariant) show similar attenuations with respect to viral growth and plaque formation. Subtle differences have been observed that are consistent with disorder-centric computational analysis.
Collapse
Affiliation(s)
- Lai Wei
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100089, China;
| | - Lihua Song
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100089, China;
| | - A. Keith Dunker
- Center for Computational Biology and Bioinformatics, Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - James A. Foster
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA;
- Institute for Bioinformatics and Evolutionary Studies, University of Idaho, Moscow, ID 83844, USA
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | | |
Collapse
|
17
|
Chao Z, Selivanovitch E, Kallitsis K, Lu Z, Pachaury A, Owens R, Daniel S. Recreating the biological steps of viral infection on a cell-free bioelectronic platform to profile viral variants of concern. Nat Commun 2024; 15:5606. [PMID: 38961055 PMCID: PMC11222515 DOI: 10.1038/s41467-024-49415-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 06/05/2024] [Indexed: 07/05/2024] Open
Abstract
Viral mutations frequently outpace technologies used to detect harmful variants. Given the continual emergence of SARS-CoV-2 variants, platforms that can identify the presence of a virus and its propensity for infection are needed. Our electronic biomembrane sensing platform recreates distinct SARS-CoV-2 host cell entry pathways and reports the progression of entry as electrical signals. We focus on two necessary entry processes mediated by the viral Spike protein: virus binding and membrane fusion, which can be distinguished electrically. We find that closely related variants of concern exhibit distinct fusion signatures that correlate with trends in cell-based infectivity assays, allowing us to report quantitative differences in their fusion characteristics and hence their infectivity potentials. We use SARS-CoV-2 as our prototype, but we anticipate that this platform can extend to other enveloped viruses and cell lines to quantifiably assess virus entry.
Collapse
Affiliation(s)
- Zhongmou Chao
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 124 Olin Hall, Ithaca, NY, 14853, USA
| | - Ekaterina Selivanovitch
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 124 Olin Hall, Ithaca, NY, 14853, USA
| | - Konstantinos Kallitsis
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Dr., Cambridge, CB3 0AS, UK
| | - Zixuan Lu
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Dr., Cambridge, CB3 0AS, UK
| | - Ambika Pachaury
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 124 Olin Hall, Ithaca, NY, 14853, USA
| | - Róisín Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Dr., Cambridge, CB3 0AS, UK
| | - Susan Daniel
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, 124 Olin Hall, Ithaca, NY, 14853, USA.
| |
Collapse
|
18
|
Salgado-Benvindo C, Tas A, Zevenhoven-Dobbe JC, van der Meer Y, Sidorov IA, Leijs AA, Wanningen P, Gelderloos AT, van Kasteren PB, Snijder EJ, van Hemert MJ. Characterization of SARS-CoV-2 replication in human H1299/ACE2 cells: A versatile and practical infection model for antiviral research and beyond. Antiviral Res 2024; 227:105903. [PMID: 38723907 DOI: 10.1016/j.antiviral.2024.105903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/12/2024] [Accepted: 05/06/2024] [Indexed: 05/18/2024]
Abstract
A range of cell culture infection models have been used to study SARS-CoV-2 and perform antiviral drug research. Commonly used African green monkey Vero, human lung-derived Calu-3 and ACE2+TMPRSS2-expressing A549 cells, each have their limitations. Here, we describe human ACE2-expressing H1299 lung cells as a more efficient and robust model for SARS-CoV-2 research. These cells are as easy to handle as Vero cells, support SARS-CoV-2 replication to high titers, display a functional innate immune response and are suitable for plaque assays, microscopy, the production of (genetically stable) virus stocks and antiviral assays. H1299/ACE2-based (CPE reduction) assays can be performed without adding a P-gP drug efflux pump inhibitor, which is often required in Vero-based assays. Moreover, H1299/ACE2 cells allowed us to perform CPE reduction assays with omicron variants that did not work in Vero-based assays. In summary, H1299/ACE2 cells are a versatile infection model to study SARS-CoV-2 replication in the context of antiviral drug development and virus-host interaction studies.
Collapse
Affiliation(s)
- Clarisse Salgado-Benvindo
- Molecular Virology Laboratory, Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Ali Tas
- Molecular Virology Laboratory, Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Jessika C Zevenhoven-Dobbe
- Molecular Virology Laboratory, Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Yvonne van der Meer
- Molecular Virology Laboratory, Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Igor A Sidorov
- Molecular Virology Laboratory, Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Anouk A Leijs
- Molecular Virology Laboratory, Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Patrick Wanningen
- Molecular Virology Laboratory, Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Anne T Gelderloos
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3720 BA, Bilthoven, the Netherlands
| | - Puck B van Kasteren
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3720 BA, Bilthoven, the Netherlands
| | - Eric J Snijder
- Molecular Virology Laboratory, Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
| | - Martijn J van Hemert
- Molecular Virology Laboratory, Center for Infectious Diseases, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands.
| |
Collapse
|
19
|
Osztie R, Czeglédi T, Ross S, Stipsicz B, Kalydi E, Béni S, Boldizsár I, Riethmüller E, Bősze SE, Alberti Á. Comprehensive Characterization of Phytochemical Composition, Membrane Permeability, and Antiproliferative Activity of Juglans nigra Polyphenols. Int J Mol Sci 2024; 25:6930. [PMID: 39000038 PMCID: PMC11241769 DOI: 10.3390/ijms25136930] [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: 05/31/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
The aim of our study was the detailed polyphenol profiling of Juglans nigra and the characterization of the membrane permeability and antiproliferative properties of its main phenolics. A total of 161 compounds were tentatively identified in J. nigra bark, leaf, and pericarp extracts by ultrahigh-performance liquid chromatography-high-resolution tandem mass spectrometry (UHPLC-HR-MS/MS). Eight compounds including myricetin-3-O-rhamnoside (86), quercetin-3-O-rhamnoside (106), quercetin-3-O-xyloside (74), juglone (141), 1,2,3,4-tetrahydro-7,8-dihydroxy-4-oxonaphthalen-1-yl-6-O-galloyl-glucoside (92), ellagic acid (143), gallic acid (14), and ethyl gallate (58) were isolated from J. nigra pericarp. The in vitro antiproliferative activity of the isolated compounds was investigated against three human cancer cell lines, confirming that juglone (141) inhibits cell proliferation in all of them, and has similar activity as the clinical standards. The permeability of the isolated compounds across biological membranes was evaluated by the parallel artificial membrane permeability assay (PAMPA). Both juglone (141) and ethyl-gallate (58) showed positive results in the blood-brain-barrier-specific PAMPA-BBB study. Juglone (141) also possesses logPe values which indicates that it may be able to cross both the GI and BBB membranes via passive diffusion.
Collapse
Affiliation(s)
- Rita Osztie
- Department of Pharmacognosy, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (R.O.); (T.C.); (I.B.); (E.R.)
| | - Tamás Czeglédi
- Department of Pharmacognosy, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (R.O.); (T.C.); (I.B.); (E.R.)
| | - Sarah Ross
- Department Pharmaceutical Biology, Institute for Drug Discovery, University of Leipzig, Eilenburger Str. 14, 04317 Leipzig, Germany;
| | - Bence Stipsicz
- Institute of Biology, Doctoral School of Biology, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary;
- HUN-REN-ELTE Research Group of Peptide Chemistry, Hungarian Research Network, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, 1117 Budapest, Hungary;
| | - Eszter Kalydi
- Institute of Organic Chemistry, Semmelweis University, Hőgyes Endre u. 7., 1092 Budapest, Hungary;
| | - Szabolcs Béni
- Institute of Chemistry, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, 1117 Budapest, Hungary;
| | - Imre Boldizsár
- Department of Pharmacognosy, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (R.O.); (T.C.); (I.B.); (E.R.)
- Department of Plant Anatomy, Institute of Biology, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/C, 1117 Budapest, Hungary
| | - Eszter Riethmüller
- Department of Pharmacognosy, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (R.O.); (T.C.); (I.B.); (E.R.)
| | - Szilvia E. Bősze
- HUN-REN-ELTE Research Group of Peptide Chemistry, Hungarian Research Network, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/A, 1117 Budapest, Hungary;
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4., 1089 Budapest, Hungary
| | - Ágnes Alberti
- Department of Pharmacognosy, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (R.O.); (T.C.); (I.B.); (E.R.)
| |
Collapse
|
20
|
Merigo F, Lagni A, Boschi F, Bernardi P, Conti A, Plebani R, Romano M, Sorio C, Lotti V, Sbarbati A. Loss of CFTR Reverses Senescence Hallmarks in SARS-CoV-2 Infected Bronchial Epithelial Cells. Int J Mol Sci 2024; 25:6185. [PMID: 38892373 PMCID: PMC11172982 DOI: 10.3390/ijms25116185] [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: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
SARS-CoV-2 infection has been recently shown to induce cellular senescence in vivo. A senescence-like phenotype has been reported in cystic fibrosis (CF) cellular models. Since the previously published data highlighted a low impact of SARS-CoV-2 on CFTR-defective cells, here we aimed to investigate the senescence hallmarks in SARS-CoV-2 infection in the context of a loss of CFTR expression/function. We infected WT and CFTR KO 16HBE14o-cells with SARS-CoV-2 and analyzed both the p21 and Ki67 expression using immunohistochemistry and viral and p21 gene expression using real-time PCR. Prior to SARS-CoV-2 infection, CFTR KO cells displayed a higher p21 and lower Ki67 expression than WT cells. We detected lipid accumulation in CFTR KO cells, identified as lipolysosomes and residual bodies at the subcellular/ultrastructure level. After SARS-CoV-2 infection, the situation reversed, with low p21 and high Ki67 expression, as well as reduced viral gene expression in CFTR KO cells. Thus, the activation of cellular senescence pathways in CFTR-defective cells was reversed by SARS-CoV-2 infection while they were activated in CFTR WT cells. These data uncover a different response of CF and non-CF bronchial epithelial cell models to SARS-CoV-2 infection and contribute to uncovering the molecular mechanisms behind the reduced clinical impact of COVID-19 in CF patients.
Collapse
Affiliation(s)
- Flavia Merigo
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (F.M.); (P.B.); (A.C.); (A.S.)
| | - Anna Lagni
- Microbiology Section, Department of Diagnostic and Public Health, University of Verona, 37134 Verona, Italy;
| | - Federico Boschi
- Department of Engineering for Innovation Medicine, University of Verona, 37134 Verona, Italy;
| | - Paolo Bernardi
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (F.M.); (P.B.); (A.C.); (A.S.)
| | - Anita Conti
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (F.M.); (P.B.); (A.C.); (A.S.)
| | - Roberto Plebani
- Laboratory of Molecular Medicine, Center for Advanced Studies and Technology (CAST), Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (R.P.); (M.R.)
| | - Mario Romano
- Laboratory of Molecular Medicine, Center for Advanced Studies and Technology (CAST), Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (R.P.); (M.R.)
| | - Claudio Sorio
- General Pathology Section, Department of Medicine, University of Verona, 37134 Verona, Italy;
| | - Virginia Lotti
- Microbiology Section, Department of Diagnostic and Public Health, University of Verona, 37134 Verona, Italy;
| | - Andrea Sbarbati
- Anatomy and Histology Section, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (F.M.); (P.B.); (A.C.); (A.S.)
| |
Collapse
|
21
|
Loveday EK, Welhaven H, Erdogan AE, Hain K, Chang CB, June RK, Taylor MP. Starve a cold or feed a fever? Identifying cellular metabolic changes following infection and exposure to SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595410. [PMID: 38826440 PMCID: PMC11142155 DOI: 10.1101/2024.05.22.595410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Viral infections induce major shifts in cellular metabolism elicited by active viral replication and antiviral responses. For the virus, harnessing cellular metabolism and evading changes that limit replication are essential for productive viral replication. In contrast, the cellular response to infection disrupts metabolic pathways to prevent viral replication and promote an antiviral state in the host cell and neighboring bystander cells. This competition between the virus and cell results in measurable shifts in cellular metabolism that differ depending on the virus, cell type, and extracellular environment. The resulting metabolic shifts can be observed and analyzed using global metabolic profiling techniques to identify pathways that are critical for either viral replication or cellular defense. SARS-CoV-2 is a respiratory virus that can exhibit broad tissue tropism and diverse, yet inconsistent, symptomatology. While the factors that determine the presentation and severity of SARS-CoV-2 infection remain unclear, metabolic syndromes are associated with more severe manifestations of SARS-CoV-2 disease. Despite these observations a critical knowledge gap remains between cellular metabolic responses and SARS-CoV-2 infection. Using a well-established untargeted metabolomics analysis workflow, we compared SARS-CoV-2 infection of human lung carcinoma cells. We identified significant changes in metabolic pathways that correlate with either productive or non-productive viral infection. This information is critical for characterizing the factors that contribute to SARS-CoV-2 replication that could be targeted for therapeutic interventions to limit viral disease.
Collapse
Affiliation(s)
- Emma Kate Loveday
- Center for Biofilm Engineering, Montana State University, Bozeman MT 59717
- Department of Chemical and Biological Engineering, Montana State University, Bozeman MT 59717
| | - Hope Welhaven
- Chemistry and Biochemistry, Montana State University, Bozeman MT 59717
| | - Ayten Ebru Erdogan
- Department of Chemical and Biological Engineering, Montana State University, Bozeman MT 59717
| | - Kyle Hain
- Microbiology and Cell Biology, Montana State University, Bozeman MT 59717
| | - Connie B. Chang
- Center for Biofilm Engineering, Montana State University, Bozeman MT 59717
- Department of Chemical and Biological Engineering, Montana State University, Bozeman MT 59717
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905
| | - Ronald K. June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman MT 59717
| | - Matthew P. Taylor
- Microbiology and Cell Biology, Montana State University, Bozeman MT 59717
| |
Collapse
|
22
|
Hartmann S, Radochonski L, Ye C, Martinez-Sobrido L, Chen J. SARS-CoV-2 ORF3a drives dynamic dense body formation for optimal viral infectivity. RESEARCH SQUARE 2024:rs.3.rs-4292014. [PMID: 38798602 PMCID: PMC11118709 DOI: 10.21203/rs.3.rs-4292014/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
SARS-CoV-2 uses the double-membrane vesicles as replication organelles. However, how virion assembly occurs has not been fully understood. Here we identified a SARS-CoV-2-driven membrane structure named the 3a dense body (3DB). 3DBs have unusual electron-dense and dynamic inner structures, and their formation is driven by the accessory protein ORF3a via hijacking a specific subset of the trans-Golgi network (TGN) and early endosomal membranes. 3DB formation is conserved in related bat and pangolin coronaviruses yet lost during the evolution to SARS-CoV. 3DBs recruit the viral structural proteins spike (S) and membrane (M) and undergo dynamic fusion/fission to facilitate efficient virion assembly. A recombinant SARS-CoV-2 virus with an ORF3a mutant specifically defective in 3DB formation showed dramatically reduced infectivity for both extracellular and cell-associated virions. Our study uncovers the crucial role of 3DB in optimal SARS-CoV-2 infectivity and highlights its potential as a target for COVID-19 prophylactics and therapeutics.
Collapse
Affiliation(s)
- Stella Hartmann
- Department of Microbiology, University of Chicago, Chicago, IL, USA 60637
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA 60439
| | - Lisa Radochonski
- Department of Microbiology, University of Chicago, Chicago, IL, USA 60637
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA 60439
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA 78227
| | | | - Jueqi Chen
- Department of Microbiology, University of Chicago, Chicago, IL, USA 60637
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA 60439
| |
Collapse
|
23
|
Keglevich G, Varga PR, Dinnyési E, Szalai Z, Bősze S, Rita OS, Drahos L, Karaghiosoff K. N-Functionalization of β-aminophosphonates: cytotoxic effects of the new derivatives. Org Biomol Chem 2024; 22:3940-3950. [PMID: 38682553 DOI: 10.1039/d4ob00243a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
β-Aminophosphonates obtained by the Michael addition of primary amines to the double bond of diethyl vinylphosphonate proved to be suitable starting materials (amine components) in the Kabachnik-Fields reaction with formaldehyde and dialkyl phosphites or secondary phosphine oxides to afford N-phosphonylmethyl- and N-phosphinoylmethyl-β-aminophosphonates. On the other hand, the starting aminophosphonates were modified by N-acylation using acid chlorides. The N-acyl products were found to exist in a dynamic equilibrium of two conformers as suggested by the broad NMR signals. At 26 °C, there may be rotation around the N-C axis of the acylamide function. At the same time, low-temperature NMR measurements at -5 °C revealed the presence of two distinct rotamers that could be characterized by 31P, 13C and 1H NMR data. The modified β-aminophosphonic derivatives were subjected to a comparative structure-activity analysis on MDA-MB-231, PC-3, A431 and Ebc-1 tumor cell lines, and in a few cases, significant activity was detected.
Collapse
Affiliation(s)
- György Keglevich
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary.
| | - Petra Regina Varga
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary.
| | - Emőke Dinnyési
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary.
| | - Zsuzsanna Szalai
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary.
| | - Szilvia Bősze
- HUN-REN-ELTE, Research Group of Peptide Chemistry, Hungarian Research Network, H-1117 Budapest, Hungary
| | - Oláhné Szabó Rita
- HUN-REN-ELTE, Research Group of Peptide Chemistry, Hungarian Research Network, H-1117 Budapest, Hungary
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary
| | - László Drahos
- MS Proteomics Research Group, Research Centre for Natural Sciences, H-1117 Budapest, Hungary
| | - Konstantin Karaghiosoff
- Department Chemie, Ludwig-Maximilians-Universitat München, Butenandtstr. 5-13, D-81377 München, Germany.
| |
Collapse
|
24
|
Carrascosa-Sàez M, Marqués MC, Geller R, Elena SF, Rahmeh A, Dufloo J, Sanjuán R. Cell type-specific adaptation of the SARS-CoV-2 spike. Virus Evol 2024; 10:veae032. [PMID: 38779130 PMCID: PMC11110937 DOI: 10.1093/ve/veae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/10/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) can infect various human tissues and cell types, principally via interaction with its cognate receptor angiotensin-converting enzyme-2 (ACE2). However, how the virus evolves in different cellular environments is poorly understood. Here, we used experimental evolution to study the adaptation of the SARS-CoV-2 spike to four human cell lines expressing different levels of key entry factors. After twenty passages of a spike-expressing recombinant vesicular stomatitis virus (VSV), cell-type-specific phenotypic changes were observed and sequencing allowed the identification of sixteen adaptive spike mutations. We used VSV pseudotyping to measure the entry efficiency, ACE2 affinity, spike processing, TMPRSS2 usage, and entry pathway usage of all the mutants, alone or in combination. The fusogenicity of the mutant spikes was assessed with a cell-cell fusion assay. Finally, mutant recombinant VSVs were used to measure the fitness advantage associated with selected mutations. We found that the effects of these mutations varied across cell types, both in terms of viral entry and replicative fitness. Interestingly, two spike mutations (L48S and A372T) that emerged in cells expressing low ACE2 levels increased receptor affinity, syncytia induction, and entry efficiency under low-ACE2 conditions. Our results demonstrate specific adaptation of the SARS-CoV-2 spike to different cell types and have implications for understanding SARS-CoV-2 tissue tropism and evolution.
Collapse
Affiliation(s)
- Marc Carrascosa-Sàez
- Institute for Integrative Systems Biology (I2SysBio). University of Valencia—CSIC, Paterna, 46980, Spain
| | - María-Carmen Marqués
- Institute for Integrative Systems Biology (I2SysBio). University of Valencia—CSIC, Paterna, 46980, Spain
| | - Ron Geller
- Institute for Integrative Systems Biology (I2SysBio). University of Valencia—CSIC, Paterna, 46980, Spain
- Instituto de Biomedicina de Valencia (IBV), CSIC and CIBER de Enfermedades Raras (CIBERER), Valencia 46010, Spain
| | - Santiago F Elena
- Institute for Integrative Systems Biology (I2SysBio). University of Valencia—CSIC, Paterna, 46980, Spain
- The Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Amal Rahmeh
- Departament de Medicina i Ciències de La Vida (MELIS), Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Jérémy Dufloo
- Institute for Integrative Systems Biology (I2SysBio). University of Valencia—CSIC, Paterna, 46980, Spain
| | - Rafael Sanjuán
- Institute for Integrative Systems Biology (I2SysBio). University of Valencia—CSIC, Paterna, 46980, Spain
| |
Collapse
|
25
|
Dufloo J, Sanjuán R. Temperature impacts SARS-CoV-2 spike fusogenicity and evolution. mBio 2024; 15:e0336023. [PMID: 38411986 PMCID: PMC11005339 DOI: 10.1128/mbio.03360-23] [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: 12/11/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024] Open
Abstract
SARS-CoV-2 infects both the upper and lower respiratory tracts, which are characterized by different temperatures (33°C and 37°C, respectively). In addition, fever is a common COVID-19 symptom. SARS-CoV-2 has been shown to replicate more efficiently at low temperatures, but the effect of temperature on different viral proteins remains poorly understood. Here, we investigate how temperature affects the SARS-CoV-2 spike function and evolution. We first observed that increasing temperature from 33°C to 37°C or 39°C increased spike-mediated cell-cell fusion. We then experimentally evolved a recombinant vesicular stomatitis virus expressing the SARS-CoV-2 spike at these different temperatures. We found that spike-mediated cell-cell fusion was maintained during evolution at 39°C but was lost in a high proportion of viruses that evolved at 33°C or 37°C. Consistently, sequencing of the spikes evolved at 33°C or 37°C revealed the accumulation of mutations around the furin cleavage site, a region that determines cell-cell fusion, whereas this did not occur in spikes evolved at 39°C. Finally, using site-directed mutagenesis, we found that disruption of the furin cleavage site had a temperature-dependent effect on spike-induced cell-cell fusion and viral fitness. Our results suggest that variations in body temperature may affect the activity and diversification of the SARS-CoV-2 spike. IMPORTANCE When it infects humans, SARS-CoV-2 is exposed to different temperatures (e.g., replication site and fever). Temperature has been shown to strongly impact SARS-CoV-2 replication, but how it affects the activity and evolution of the spike protein remains poorly understood. Here, we first show that high temperatures increase the SARS-CoV-2 spike fusogenicity. Then, we demonstrate that the evolution of the spike activity and variants depends on temperature. Finally, we show that the functional effect of specific spike mutations is temperature-dependent. Overall, our results suggest that temperature may be a factor influencing the activity and adaptation of the SARS-CoV-2 spike in vivo, which will help understanding viral tropism, pathogenesis, and evolution.
Collapse
Affiliation(s)
- Jérémy Dufloo
- Institute for Integrative Systems Biology, Consejo Superior de Investigaciones Científicas-Universitat de València, Paterna, València, Spain
| | - Rafael Sanjuán
- Institute for Integrative Systems Biology, Consejo Superior de Investigaciones Científicas-Universitat de València, Paterna, València, Spain
| |
Collapse
|
26
|
Minami S, Kotaki T, Sakai Y, Okamura S, Torii S, Ono C, Motooka D, Hamajima R, Nouda R, Nurdin JA, Yamasaki M, Kanai Y, Ebina H, Maeda Y, Okamoto T, Tachibana T, Matsuura Y, Kobayashi T. Vero cell-adapted SARS-CoV-2 strain shows increased viral growth through furin-mediated efficient spike cleavage. Microbiol Spectr 2024; 12:e0285923. [PMID: 38415690 PMCID: PMC10986611 DOI: 10.1128/spectrum.02859-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 02/01/2024] [Indexed: 02/29/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) utilizes several host proteases to cleave the spike (S) protein to enter host cells. SARS-CoV-2 S protein is cleaved into S1 and S2 subunits by furin, which is closely involved in the pathogenicity of SARS-CoV-2. However, the effects of the modulated protease cleavage activity due to S protein mutations on viral replication and pathogenesis remain unclear. Herein, we serially passaged two SARS-CoV-2 strains in Vero cells and characterized the cell-adapted SARS-CoV-2 strains in vitro and in vivo. The adapted strains showed high viral growth, effective S1/S2 cleavage of the S protein, and low pathogenicity compared with the wild-type strain. Furthermore, the viral growth and S1/S2 cleavage were enhanced by the combination of the Δ68-76 and H655Y mutations using recombinant SARS-CoV-2 strains generated by the circular polymerase extension reaction. The recombinant SARS-CoV-2 strain, which contained the mutation of the adapted strain, showed increased susceptibility to the furin inhibitor, suggesting that the adapted SARS-CoV-2 strain utilized furin more effectively than the wild-type strain. Pathogenicity was attenuated by infection with effectively cleaved recombinant SARS-CoV-2 strains, suggesting that the excessive cleavage of the S proteins decreases virulence. Finally, the high-growth-adapted SARS-CoV-2 strain could be used as the seed for a low-cost inactivated vaccine; immunization with this vaccine can effectively protect the host from SARS-CoV-2 variants. Our findings provide novel insights into the growth and pathogenicity of SARS-CoV-2 in the evolution of cell-cell transmission. IMPORTANCE The efficacy of the S protein cleavage generally differs among the SARS-CoV-2 variants, resulting in distinct viral characteristics. The relationship between a mutation and the entry of SARS-CoV-2 into host cells remains unclear. In this study, we analyzed the sequence of high-growth Vero cell-adapted SARS-CoV-2 and factors determining the enhancement of the growth of the adapted virus and confirmed the characteristics of the adapted strain by analyzing the recombinant SARS-CoV-2 strain. We successfully identified mutations Δ68-76 and H655Y, which enhance viral growth and the S protein cleavage by furin. Using recombinant viruses enabled us to conduct a virus challenge experiment in vivo. The pathogenicity of SARS-CoV-2 introduced with the mutations Δ68-76, H655Y, P812L, and Q853L was attenuated in hamsters, indicating the possibility of the attenuation of excessive cleaved SARS-CoV-2. These findings provide novel insights into the infectivity and pathogenesis of SARS-CoV-2 strains, thereby significantly contributing to the field of virology.
Collapse
Affiliation(s)
- Shohei Minami
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tomohiro Kotaki
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yusuke Sakai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shinya Okamura
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan
| | - Shiho Torii
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Chikako Ono
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Rina Hamajima
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Ryotaro Nouda
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Jeffery A. Nurdin
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Moeko Yamasaki
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yuta Kanai
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Hirotaka Ebina
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- The Research Foundation for Microbial Diseases of Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| | - Yusuke Maeda
- Laboratory of Viral Dynamism Research, Research Institute for Microbial Diseases Osaka University, Osaka, Japan
| | - Toru Okamoto
- Institute for Advanced Co-creation Studies, Research Institute for Microbial Diseases Osaka University, Osaka, Japan
| | - Taro Tachibana
- Cell Engineering Corporation, Osaka, Japan
- Department of Bioengineering, Graduate School of Engineering, Osaka Metropolitan University, Osaka, Japan
| | - Yoshiharu Matsuura
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| | - Takeshi Kobayashi
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| |
Collapse
|
27
|
Steiner S, Kratzel A, Barut GT, Lang RM, Aguiar Moreira E, Thomann L, Kelly JN, Thiel V. SARS-CoV-2 biology and host interactions. Nat Rev Microbiol 2024; 22:206-225. [PMID: 38225365 DOI: 10.1038/s41579-023-01003-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 01/17/2024]
Abstract
The zoonotic emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the ensuing coronavirus disease 2019 (COVID-19) pandemic have profoundly affected our society. The rapid spread and continuous evolution of new SARS-CoV-2 variants continue to threaten global public health. Recent scientific advances have dissected many of the molecular and cellular mechanisms involved in coronavirus infections, and large-scale screens have uncovered novel host-cell factors that are vitally important for the virus life cycle. In this Review, we provide an updated summary of the SARS-CoV-2 life cycle, gene function and virus-host interactions, including recent landmark findings on general aspects of coronavirus biology and newly discovered host factors necessary for virus replication.
Collapse
Affiliation(s)
- Silvio Steiner
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Annika Kratzel
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - G Tuba Barut
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Reto M Lang
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Etori Aguiar Moreira
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Lisa Thomann
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Jenna N Kelly
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland
- European Virus Bioinformatics Center, Jena, Germany
| | - Volker Thiel
- Institute of Virology and Immunology, Bern and Mittelhäusern, Bern, Switzerland.
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
- Multidisciplinary Center for Infectious Diseases, University of Bern, Bern, Switzerland.
- European Virus Bioinformatics Center, Jena, Germany.
| |
Collapse
|
28
|
Tomris I, van der Woude R, de Paiva Froes Rocha R, Torrents de la Peña A, Ward AB, de Vries RP. Viral envelope proteins fused to multiple distinct fluorescent reporters to probe receptor binding. Protein Sci 2024; 33:e4974. [PMID: 38533540 DOI: 10.1002/pro.4974] [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: 10/25/2023] [Revised: 03/04/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024]
Abstract
Enveloped viruses carry one or multiple proteins with receptor-binding functionalities. Functional receptors can be glycans, proteinaceous, or both; therefore, recombinant protein approaches are instrumental in attaining new insights regarding viral envelope protein receptor-binding properties. Visualizing and measuring receptor binding typically entails antibody detection or direct labeling, whereas direct fluorescent fusions are attractive tools in molecular biology. Here, we report a suite of distinct fluorescent fusions, both N- and C-terminal, for influenza A virus hemagglutinins and SARS-CoV-2 spike RBD. The proteins contained three or six fluorescent protein barrels and were applied directly to cells to assess receptor binding properties.
Collapse
Affiliation(s)
- Ilhan Tomris
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, The Netherlands
| | - Roosmarijn van der Woude
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, The Netherlands
| | - Rebeca de Paiva Froes Rocha
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Alba Torrents de la Peña
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Robert P de Vries
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, The Netherlands
| |
Collapse
|
29
|
Zitzmann C, Ke R, Ribeiro RM, Perelson AS. How robust are estimates of key parameters in standard viral dynamic models? PLoS Comput Biol 2024; 20:e1011437. [PMID: 38626190 PMCID: PMC11051641 DOI: 10.1371/journal.pcbi.1011437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 04/26/2024] [Accepted: 04/01/2024] [Indexed: 04/18/2024] Open
Abstract
Mathematical models of viral infection have been developed, fitted to data, and provide insight into disease pathogenesis for multiple agents that cause chronic infection, including HIV, hepatitis C, and B virus. However, for agents that cause acute infections or during the acute stage of agents that cause chronic infections, viral load data are often collected after symptoms develop, usually around or after the peak viral load. Consequently, we frequently lack data in the initial phase of viral growth, i.e., when pre-symptomatic transmission events occur. Missing data may make estimating the time of infection, the infectious period, and parameters in viral dynamic models, such as the cell infection rate, difficult. However, having extra information, such as the average time to peak viral load, may improve the robustness of the estimation. Here, we evaluated the robustness of estimates of key model parameters when viral load data prior to the viral load peak is missing, when we know the values of some parameters and/or the time from infection to peak viral load. Although estimates of the time of infection are sensitive to the quality and amount of available data, particularly pre-peak, other parameters important in understanding disease pathogenesis, such as the loss rate of infected cells, are less sensitive. Viral infectivity and the viral production rate are key parameters affecting the robustness of data fits. Fixing their values to literature values can help estimate the remaining model parameters when pre-peak data is missing or limited. We find a lack of data in the pre-peak growth phase underestimates the time to peak viral load by several days, leading to a shorter predicted growth phase. On the other hand, knowing the time of infection (e.g., from epidemiological data) and fixing it results in good estimates of dynamical parameters even in the absence of early data. While we provide ways to approximate model parameters in the absence of early viral load data, our results also suggest that these data, when available, are needed to estimate model parameters more precisely.
Collapse
Affiliation(s)
- Carolin Zitzmann
- Theoretical Biology and Biophysics Group, Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico
| | - Ruian Ke
- Theoretical Biology and Biophysics Group, Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico
| | - Ruy M. Ribeiro
- Theoretical Biology and Biophysics Group, Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico
| | - Alan S. Perelson
- Theoretical Biology and Biophysics Group, Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico
| |
Collapse
|
30
|
Mendonça SC, Gomes BA, Campos MF, da Fonseca TS, Esteves MEA, Andriolo BV, Cheohen CFDAR, Constant LEC, da Silva Costa S, Calil PT, Tucci AR, de Oliveira TKF, Rosa ADS, Ferreira VNDS, Lima JNH, Miranda MD, da Costa LJ, da Silva ML, Scotti MT, Allonso D, Leitão GG, Leitão SG. Myrtucommulones and Related Acylphloroglucinols from Myrtaceae as a Promising Source of Multitarget SARS-CoV-2 Cycle Inhibitors. Pharmaceuticals (Basel) 2024; 17:436. [PMID: 38675398 PMCID: PMC11054083 DOI: 10.3390/ph17040436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
The LABEXTRACT plant extract bank, featuring diverse members of the Myrtaceae family from Brazilian hot spot regions, provides a promising avenue for bioprospection. Given the pivotal roles of the Spike protein and 3CLpro and PLpro proteases in SARS-CoV-2 infection, this study delves into the correlations between the Myrtaceae species from the Atlantic Forest and these targets, as well as an antiviral activity through both in vitro and in silico analyses. The results uncovered notable inhibitory effects, with Eugenia prasina and E. mosenii standing out, while E. mosenii proved to be multitarget, presenting inhibition values above 72% in the three targets analyzed. All extracts inhibited viral replication in Calu-3 cells (EC50 was lower than 8.3 µg·mL-1). Chemometric analyses, through LC-MS/MS, encompassing prediction models and molecular networking, identified potential active compounds, such as myrtucommulones, described in the literature for their antiviral activity. Docking analyses showed that one undescribed myrtucommulone (m/z 841 [M - H]-) had a higher fitness score when interacting with the targets of this study, including ACE2, Spike, PLpro and 3CLpro of SARS-CoV-2. Also, the study concludes that Myrtaceae extracts, particularly from E. mosenii and E. prasina, exhibit promising inhibitory effects against crucial stages in SARS-CoV-2 infection. Compounds like myrtucommulones emerge as potential anti-SARS-CoV-2 agents, warranting further exploration.
Collapse
Affiliation(s)
- Simony Carvalho Mendonça
- Departamento de Produtos Naturais e Alimentos, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.C.M.); (B.A.G.); (M.F.C.)
| | - Brendo Araujo Gomes
- Departamento de Produtos Naturais e Alimentos, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.C.M.); (B.A.G.); (M.F.C.)
- Programa de Pós-Graduação em Biotecnologia Vegetal e Bioprocessos, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Mariana Freire Campos
- Departamento de Produtos Naturais e Alimentos, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.C.M.); (B.A.G.); (M.F.C.)
- Programa de Pós-Graduação em Biotecnologia Vegetal e Bioprocessos, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Thamirys Silva da Fonseca
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Maria Eduarda Alves Esteves
- Programa de Pós-Graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil; (M.E.A.E.); (M.L.d.S.)
| | - Bruce Veiga Andriolo
- Programa de Pós-Graduação em Biotecnologia, Instituto Nacional de Metrologia, Qualidade e Tecnologia, Duque de Caxias 25250-020, RJ, Brazil;
| | - Caio Felipe de Araujo Ribas Cheohen
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Centro de Ciências da Saúde, Instituto de Biodiversidade e Sustentabilidade NUPEM, Universidade Federal do Rio de Janeiro, Macaé 27965-045, RJ, Brazil;
| | - Larissa Esteves Carvalho Constant
- Programa de Pós-Graduação em Ciências Biológicas, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil; (L.E.C.C.); (S.d.S.C.); (D.A.)
| | - Stephany da Silva Costa
- Programa de Pós-Graduação em Ciências Biológicas, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil; (L.E.C.C.); (S.d.S.C.); (D.A.)
| | - Pedro Telles Calil
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil; (P.T.C.); (L.J.d.C.)
| | - Amanda Resende Tucci
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (T.K.F.d.O.); (A.d.S.R.); (V.N.d.S.F.); (J.N.H.L.); (M.D.M.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Thamara Kelcya Fonseca de Oliveira
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (T.K.F.d.O.); (A.d.S.R.); (V.N.d.S.F.); (J.N.H.L.); (M.D.M.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Alice dos Santos Rosa
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (T.K.F.d.O.); (A.d.S.R.); (V.N.d.S.F.); (J.N.H.L.); (M.D.M.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Vivian Neuza dos Santos Ferreira
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (T.K.F.d.O.); (A.d.S.R.); (V.N.d.S.F.); (J.N.H.L.); (M.D.M.)
| | - Julia Nilo Henrique Lima
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (T.K.F.d.O.); (A.d.S.R.); (V.N.d.S.F.); (J.N.H.L.); (M.D.M.)
| | - Milene Dias Miranda
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (T.K.F.d.O.); (A.d.S.R.); (V.N.d.S.F.); (J.N.H.L.); (M.D.M.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Luciana Jesus da Costa
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil; (P.T.C.); (L.J.d.C.)
| | - Manuela Leal da Silva
- Programa de Pós-Graduação em Biologia Computacional e Sistemas, Instituto Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil; (M.E.A.E.); (M.L.d.S.)
- Programa de Pós-Graduação em Biotecnologia, Instituto Nacional de Metrologia, Qualidade e Tecnologia, Duque de Caxias 25250-020, RJ, Brazil;
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas, Centro de Ciências da Saúde, Instituto de Biodiversidade e Sustentabilidade NUPEM, Universidade Federal do Rio de Janeiro, Macaé 27965-045, RJ, Brazil;
| | - Marcus Tullius Scotti
- Departamento de Química, Universidade Federal da Paraíba, João Pessoa 58033-455, PB, Brazil;
| | - Diego Allonso
- Programa de Pós-Graduação em Ciências Biológicas, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, RJ, Brazil; (L.E.C.C.); (S.d.S.C.); (D.A.)
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Gilda Guimarães Leitão
- Instituto de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Suzana Guimarães Leitão
- Departamento de Produtos Naturais e Alimentos, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.C.M.); (B.A.G.); (M.F.C.)
- Programa de Pós-Graduação em Biotecnologia Vegetal e Bioprocessos, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| |
Collapse
|
31
|
Jacob IB, Gemmiti A, Xiong W, Reynolds E, Nicholas B, Thangamani S, Jia H, Wang G. Human surfactant protein A inhibits SARS-CoV-2 infectivity and alleviates lung injury in a mouse infection model. Front Immunol 2024; 15:1370511. [PMID: 38596675 PMCID: PMC11002091 DOI: 10.3389/fimmu.2024.1370511] [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: 01/14/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
Introduction SARS coronavirus 2 (SARS-CoV-2) infects human angiotensin-converting enzyme 2 (hACE2)-expressing lung epithelial cells through its spike (S) protein. The S protein is highly glycosylated and could be a target for lectins. Surfactant protein A (SP-A) is a collagen-containing C-type lectin, expressed by mucosal epithelial cells and mediates its antiviral activities by binding to viral glycoproteins. Objective This study examined the mechanistic role of human SP-A in SARS-CoV-2 infectivity and lung injury in vitro and in vivo. Results Human SP-A can bind both SARS-CoV-2 S protein and hACE2 in a dose-dependent manner (p<0.01). Pre-incubation of SARS-CoV-2 (Delta) with human SP-A inhibited virus binding and entry and reduced viral load in human lung epithelial cells, evidenced by the dose-dependent decrease in viral RNA, nucleocapsid protein (NP), and titer (p<0.01). We observed significant weight loss, increased viral burden, and mortality rate, and more severe lung injury in SARS-CoV-2 infected hACE2/SP-A KO mice (SP-A deficient mice with hACE2 transgene) compared to infected hACE2/mSP-A (K18) and hACE2/hSP-A1 (6A2) mice (with both hACE2 and human SP-A1 transgenes) 6 Days Post-infection (DPI). Furthermore, increased SP-A level was observed in the saliva of COVID-19 patients compared to healthy controls (p<0.05), but severe COVID-19 patients had relatively lower SP-A levels than moderate COVID-19 patients (p<0.05). Discussion Collectively, human SP-A attenuates SARS-CoV-2-induced acute lung injury (ALI) by directly binding to the S protein and hACE2, and inhibiting its infectivity; and SP-A level in the saliva of COVID-19 patients might serve as a biomarker for COVID-19 severity.
Collapse
Affiliation(s)
- Ikechukwu B. Jacob
- Department of Surgery, the State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Amanda Gemmiti
- Department of Otolaryngology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Weichuan Xiong
- Department of Surgery, the State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Erin Reynolds
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Brian Nicholas
- Department of Otolaryngology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Saravanan Thangamani
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Hongpeng Jia
- Department of Surgery, Johns-Hopkins University, Baltimore, MD, United States
| | - Guirong Wang
- Department of Surgery, the State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
32
|
Faley SL, Boghdeh NA, Schaffer DK, Spivey EC, Alem F, Narayanan A, Wikswo JP, Brown JA. Gravity-perfused airway-on-a-chip optimized for quantitative BSL-3 studies of SARS-CoV-2 infection: barrier permeability, cytokine production, immunohistochemistry, and viral load assays. LAB ON A CHIP 2024; 24:1794-1807. [PMID: 38362777 PMCID: PMC10929697 DOI: 10.1039/d3lc00894k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/26/2024] [Indexed: 02/17/2024]
Abstract
Human microphysiological systems, such as organs on chips, are an emerging technology for modeling human physiology in a preclinical setting to understand the mechanism of action of drugs, to evaluate the efficacy of treatment options for human disease and impairment, and to assess drug toxicity. By using human cells co-cultured in three-dimensional constructs, organ chips can provide greater fidelity to the human cellular condition than their two-dimensional predecessors. However, with the rise of SARS-CoV-2 and the global COVID-19 pandemic, it became clear that many microphysiological systems were not compatible with or optimized for studies of infectious disease and operation in a Biosafety Level 3 (BSL-3) environment. Given that one of the early sites of SARS-CoV-2 infection is the airway, we created a human airway organ chip that could operate in a BSL-3 space with high throughput and minimal manipulation, while retaining the necessary physical and physiological components to recapitulate tissue response to infectious agents and the immune response to infection.
Collapse
Affiliation(s)
- Shannon L Faley
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
| | - Niloufar A Boghdeh
- Biomedical Research Laboratory, Institute of Biohealth Innovation, George Mason University, Manassas, VA 20110, USA
| | - David K Schaffer
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37235, USA
| | - Eric C Spivey
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
| | - Farhang Alem
- Biomedical Research Laboratory, Institute of Biohealth Innovation, George Mason University, Manassas, VA 20110, USA
| | - Aarthi Narayanan
- Biomedical Research Laboratory, Institute of Biohealth Innovation, George Mason University, Manassas, VA 20110, USA
- College of Science, Department of Biology, George Mason University, Fairfax, VA 22030, USA
| | - John P Wikswo
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA.
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37235, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Jacquelyn A Brown
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN 37235, USA
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN 37235, USA
| |
Collapse
|
33
|
van Huizen M, Bloeme - ter Horst JR, de Gruyter HLM, Geurink PP, van der Heden van Noort GJ, Knaap RCM, Nelemans T, Ogando NS, Leijs AA, Urakova N, Mark BL, Snijder EJ, Myeni SK, Kikkert M. Deubiquitinating activity of SARS-CoV-2 papain-like protease does not influence virus replication or innate immune responses in vivo. PLoS Pathog 2024; 20:e1012100. [PMID: 38527094 PMCID: PMC10994560 DOI: 10.1371/journal.ppat.1012100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/04/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024] Open
Abstract
The coronavirus papain-like protease (PLpro) is crucial for viral replicase polyprotein processing. Additionally, PLpro can subvert host defense mechanisms by its deubiquitinating (DUB) and deISGylating activities. To elucidate the role of these activities during SARS-CoV-2 infection, we introduced mutations that disrupt binding of PLpro to ubiquitin or ISG15. We identified several mutations that strongly reduced DUB activity of PLpro, without affecting viral polyprotein processing. In contrast, mutations that abrogated deISGylating activity also hampered viral polyprotein processing and when introduced into the virus these mutants were not viable. SARS-CoV-2 mutants exhibiting reduced DUB activity elicited a stronger interferon response in human lung cells. In a mouse model of severe disease, disruption of PLpro DUB activity did not affect lethality, virus replication, or innate immune responses in the lungs. This suggests that the DUB activity of SARS-CoV-2 PLpro is dispensable for virus replication and does not affect innate immune responses in vivo. Interestingly, the DUB mutant of SARS-CoV replicated to slightly lower titers in mice and elicited a diminished immune response early in infection, although lethality was unaffected. We previously showed that a MERS-CoV mutant deficient in DUB and deISGylating activity was strongly attenuated in mice. Here, we demonstrate that the role of PLpro DUB activity during infection can vary considerably between highly pathogenic coronaviruses. Therefore, careful considerations should be taken when developing pan-coronavirus antiviral strategies targeting PLpro.
Collapse
Affiliation(s)
- Mariska van Huizen
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Jonna R. Bloeme - ter Horst
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Heidi L. M. de Gruyter
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Paul P. Geurink
- Department of Cell and Chemical Biology, Division of Chemical Biology and Drug Discovery, Leiden University Medical Centre, Leiden, The Netherlands
| | - Gerbrand J. van der Heden van Noort
- Department of Cell and Chemical Biology, Division of Chemical Biology and Drug Discovery, Leiden University Medical Centre, Leiden, The Netherlands
| | - Robert C. M. Knaap
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Tessa Nelemans
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Natacha S. Ogando
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Anouk A. Leijs
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Nadya Urakova
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Brian L. Mark
- Department of Microbiology, University of Manitoba, Winnipeg, Canada
| | - Eric J. Snijder
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Sebenzile K. Myeni
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Marjolein Kikkert
- Molecular Virology Laboratory, Leiden University Center of Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
34
|
Lam FW, Brown CA, Ronca SE. Recombinant Rod Domain of Vimentin Reduces SARS-CoV-2 Viral Replication by Blocking Spike Protein-ACE2 Interactions. Int J Mol Sci 2024; 25:2477. [PMID: 38473724 PMCID: PMC10931652 DOI: 10.3390/ijms25052477] [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: 12/05/2023] [Revised: 01/18/2024] [Accepted: 01/26/2024] [Indexed: 03/14/2024] Open
Abstract
Although the SARS-CoV-2 vaccination is the primary preventive intervention, there are still few antiviral therapies available, with current drugs decreasing viral replication once the virus is intracellular. Adding novel drugs to target additional points in the viral life cycle is paramount in preventing future pandemics. The purpose of this study was to create and test a novel protein to decrease SARS-CoV-2 replication. We created the recombinant rod domain of vimentin (rhRod) in E. coli and used biolayer interferometry to measure its affinity to the SARS-CoV-2 S1S2 spike protein and the ability to block the SARS-CoV-2-ACE2 interaction. We performed plaque assays to measure rhRod's effect on SARS-CoV-2 replication in Vero E6 cells. Finally, we measured lung inflammation in SARS-CoV-2-exposed K18-hACE transgenic mice given intranasal and intraperitoneal rhRod. We found that rhRod has a high affinity for the S1S2 protein with a strong ability to block S1S2-ACE2 interactions. The daily addition of rhRod decreased viral replication in Vero E6 cells starting at 48 h at concentrations >1 µM. Finally, SARS-CoV-2-infected mice receiving rhRod had decreased lung inflammation compared to mock-treated animals. Based on our data, rhRod decreases SARS-CoV-2 replication in vitro and lung inflammation in vivo. Future studies will need to evaluate the protective effects of rhRod against additional viral variants and identify the optimal dosing scheme that both prevents viral replication and host lung injury.
Collapse
Affiliation(s)
- Fong Wilson Lam
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA (S.E.R.)
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX 77030, USA
| | - Cameron August Brown
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA (S.E.R.)
- Department of Pathology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shannon Elizabeth Ronca
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA (S.E.R.)
- William T. Shearer Center for Human Immunobiology, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX 77030, USA
| |
Collapse
|
35
|
Sarlo Davila KM, Nelli RK, Phadke KS, Ruden RM, Sang Y, Bellaire BH, Gimenez-Lirola LG, Miller LC. How do deer respiratory epithelial cells weather the initial storm of SARS-CoV-2 WA1/2020 strain? Microbiol Spectr 2024; 12:e0252423. [PMID: 38189329 PMCID: PMC10846091 DOI: 10.1128/spectrum.02524-23] [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: 06/16/2023] [Accepted: 11/28/2023] [Indexed: 01/09/2024] Open
Abstract
The potential infectivity of severe acute respiratory syndrome associated coronavirus-2 (SARS-CoV-2) in animals raises a public health and economic concern, particularly the high susceptibility of white-tailed deer (WTD) to SARS-CoV-2. The disparity in the disease outcome between humans and WTD is very intriguing, as the latter are often asymptomatic, subclinical carriers of SARS-CoV-2. To date, no studies have evaluated the innate immune factors responsible for the contrasting SARS-CoV-2-associated disease outcomes in these mammalian species. A comparative transcriptomic analysis in primary respiratory epithelial cells of human (HRECs) and WTD (Deer-RECs) infected with the SARS-CoV-2 WA1/2020 strain was assessed throughout 48 h post inoculation (hpi). Both HRECs and Deer-RECs were susceptible to virus infection, with significantly (P < 0.001) lower virus replication in Deer-RECs. The number of differentially expressed genes (DEG) gradually increased in Deer-RECs but decreased in HRECs throughout the infection. The ingenuity pathway analysis of DEGs further identified that genes commonly altered during SARS-CoV-2 infection mainly belong to cytokine and chemokine response pathways mediated via interleukin-17 (IL-17) and nuclear factor-κB (NF-κB) signaling pathways. Inhibition of the NF-κB signaling in the Deer-RECs pathway was predicted as early as 6 hpi. The findings from this study could explain the lack of clinical signs reported in WTD in response to SARS-CoV-2 infection as opposed to the severe clinical outcomes reported in humans.IMPORTANCEThis study demonstrated that human and white-tailed deer primary respiratory epithelial cells are susceptible to the SARS-CoV-2 WA1/2020 strain infection. However, the comparative transcriptomic analysis revealed that deer cells could limit viral replication without causing hypercytokinemia by downregulating IL-17 and NF-κB signaling pathways. Identifying differentially expressed genes in human and deer cells that modulate key innate immunity pathways during the early infection will lead to developing targeted therapies toward preventing or mitigating the "cytokine storm" often associated with severe cases of coronavirus disease 19 (COVID-19). Moreover, results from this study will aid in identifying novel prognostic biomarkers in predicting SARS-CoV-2 adaption and transmission in deer and associated cervids.
Collapse
Affiliation(s)
- Kaitlyn M. Sarlo Davila
- United States Department of Agriculture, Agricultural Research Service, Infectious Bacterial Disease Research Unit, National Animal Disease Center , Ames, Iowa, USA
| | - Rahul K. Nelli
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Kruttika S. Phadke
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Rachel M. Ruden
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Yongming Sang
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, Nashville, Tennessee, USA
| | - Bryan H. Bellaire
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Luis G. Gimenez-Lirola
- Department of Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | - Laura C. Miller
- United States Department of Agriculture, Agricultural Research Service, Virus and Prion Research Unit, National Animal Disease Center, Ames, Iowa, USA
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
36
|
Arumugam S, Vadivel K, Dhaked DK, Prasanna VS, Yellurkar ML, Das P, Manoharan R, Thomas AJ, Singh L, Singh S, Velayutham R. Antiviral Activity of Cinchona officinalis, a Homeopathic Medicine, against COVID-19. HOMEOPATHY 2024; 113:16-24. [PMID: 37673083 DOI: 10.1055/s-0043-1770339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is a potentially fatal disease caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Several studies have shown that hydroxychloroquine (HCQ) significantly inhibits SARS-CoV-2 infections in vitro. OBJECTIVE Since the phytoconstituents of Cinchona officinalis (CO) are similar to those of HCQ, the objective of this study was to test the antiviral potential of different homeopathic formulations of CO. METHODS An analysis of the molecular composition of CO was carried out using ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry, followed by a detailed docking study. The constituents of CO were docked against various targets of SARS-CoV-2, and the binding potential of the phytoconstituents was compared and quantified. The ligand with the lowest Glide docking score is considered to have the best binding affinity. The cytotoxicity of several homeopathic formulations, including CO mother tincture (CO-MT), was also checked on VeroE6 cells. A known antiviral, remdesivir, was used as a positive control for the in vitro assays to evaluate the effects of CO-MT against SARS-CoV-2-infected VeroE6 cells. RESULTS Molecular docking studies showed that constituents of CO exhibited binding potential to various targets of SARS-CoV-2, including Mpro, PLpro, RdRp, nucleocapsid protein, ACE2 (in host) and spike protein. Quinoline, one of the constituents of CO, can potentially bind the spike protein of SARS-CoV-2. Quinic acid showed better binding capabilities with Mpro, PLpro RdRp, nucleocapsid protein and ACE2 (allosteric site) than other constituents. Quinidine exhibited better binding to ACE2. Compared to HCQ, other phytoconstituents of CO had the equivalent potential to bind the RNA-dependent RNA polymerase, nucleocapsid protein, Mpro, PLpro and spike protein of SARS-CoV-2. In vitro assays showed that homeopathic CO-MT was not cytotoxic and that CO-MT and remdesivir respectively caused 89% and 99% inhibition of SARS-CoV-2 infection in VeroE6 cells. CONCLUSION Based on this in silico and in vitro evidence, we propose CO-MT as a promising antiviral medicine candidate for treating COVID-19. In vivo investigation is required to clarify the therapeutic potential of CO-MT in COVID-19.
Collapse
Affiliation(s)
- Somasundaram Arumugam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, Kolkata, West Bengal, India
| | - Kumaravel Vadivel
- Department of Pathology and Microbiology, National Institute of Homeopathy (NIH), Kolkata, West Bengal, India
| | - Devendra Kumar Dhaked
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, Kolkata, West Bengal, India
| | - Vani Sai Prasanna
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, Kolkata, West Bengal, India
| | - Manoj Limbraj Yellurkar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, Kolkata, West Bengal, India
| | - Pamelika Das
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, Kolkata, West Bengal, India
| | - Raja Manoharan
- Department of Homeopathic Pharmacy, National Institute of Homeopathy (NIH), Kolkata, West Bengal, India
| | | | - Lalith Singh
- National Institute of Homeopathy (NIH), Kolkata, West Bengal, India
| | - Subhas Singh
- Department of Organon of Medicine, National Institute of Homeopathy (NIH), Kolkata, West Bengal, India
| | - Ravichandiran Velayutham
- National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, West Bengal, India
| |
Collapse
|
37
|
Ellis S, Way R, Nel M, Burleigh A, Doykov I, Kembou-Ringert J, Woodall M, Masonou T, Case KM, Ortez AT, McHugh TD, Casal A, McCoy LE, Murdan S, Hynds RE, Gilmour KC, Grandjean L, Cortina-Borja M, Heywood WE, Mills K, Smith CM. Salivary IgA and vimentin differentiate in vitro SARS-CoV-2 infection: A study of 290 convalescent COVID-19 patients. Mucosal Immunol 2024; 17:124-136. [PMID: 38007005 PMCID: PMC11139657 DOI: 10.1016/j.mucimm.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
SARS-CoV-2 initially infects cells in the nasopharynx and oral cavity. The immune system at these mucosal sites plays a crucial role in minimizing viral transmission and infection. To develop new strategies for preventing SARS-CoV-2 infection, this study aimed to identify proteins that protect against viral infection in saliva. We collected 551 saliva samples from 290 healthcare workers who had tested positive for COVID-19, before vaccination, between June and December 2020. The samples were categorized based on their ability to block or enhance infection using in vitro assays. Mass spectrometry and enzyme-linked immunosorbent assay experiments were used to identify and measure the abundance of proteins that specifically bind to SARS-CoV-2 antigens. Immunoglobulin (Ig)A specific to SARS-CoV-2 antigens was detectable in over 83% of the convalescent saliva samples. We found that concentrations of anti-receptor-binding domain IgA >500 pg/µg total protein in saliva correlate with reduced viral infectivity in vitro. However, there is a dissociation between the salivary IgA response to SARS-CoV-2, and systemic IgG titers in convalescent COVID-19 patients. Then, using an innovative technique known as spike-baited mass spectrometry, we identified novel spike-binding proteins in saliva, most notably vimentin, which correlated with increased viral infectivity in vitro and could serve as a therapeutic target against COVID-19.
Collapse
Affiliation(s)
- Samuel Ellis
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Rosie Way
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Miranda Nel
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alice Burleigh
- UCL Great Ormond Street Institute of Child Health, London, UK; Centre for Adolescent Rheumatology, University College London, London, UK
| | - Ivan Doykov
- UCL Great Ormond Street Institute of Child Health, London, UK
| | | | | | - Tereza Masonou
- UCL Great Ormond Street Institute of Child Health, London, UK
| | | | | | - Timothy D McHugh
- UCL Centre for Clinical Microbiology, Royal Free Hospital, London, UK
| | - Antonio Casal
- Department of Pharmaceutics, UCL School of Pharmacy, London, UK
| | - Laura E McCoy
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | | | - Robert E Hynds
- Epithelial Cell Biology in ENT Research (EpiCENTR) Group, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Kimberly C Gilmour
- UCL Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Louis Grandjean
- UCL Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | | | - Wendy E Heywood
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Kevin Mills
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Claire M Smith
- UCL Great Ormond Street Institute of Child Health, London, UK.
| |
Collapse
|
38
|
Avila-Herrera A, Kimbrel JA, Manuel Martí J, Thissen J, Saada EA, Weisenberger T, Arrildt KT, Segelke BW, Allen JE, Zemla A, Borucki MK. Differential laboratory passaging of SARS-CoV-2 viral stocks impacts the in vitro assessment of neutralizing antibodies. PLoS One 2024; 19:e0289198. [PMID: 38271318 PMCID: PMC10810540 DOI: 10.1371/journal.pone.0289198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Viral populations in natural infections can have a high degree of sequence diversity, which can directly impact immune escape. However, antibody potency is often tested in vitro with a relatively clonal viral populations, such as laboratory virus or pseudotyped virus stocks, which may not accurately represent the genetic diversity of circulating viral genotypes. This can affect the validity of viral phenotype assays, such as antibody neutralization assays. To address this issue, we tested whether recombinant virus carrying SARS-CoV-2 spike (VSV-SARS-CoV-2-S) stocks could be made more genetically diverse by passage, and if a stock passaged under selective pressure was more capable of escaping monoclonal antibody (mAb) neutralization than unpassaged stock or than viral stock passaged without selective pressures. We passaged VSV-SARS-CoV-2-S four times concurrently in three cell lines and then six times with or without polyclonal antiserum selection pressure. All three of the monoclonal antibodies tested neutralized the viral population present in the unpassaged stock. The viral inoculum derived from serial passage without antiserum selection pressure was neutralized by two of the three mAbs. However, the viral inoculum derived from serial passage under antiserum selection pressure escaped neutralization by all three mAbs. Deep sequencing revealed the rapid acquisition of multiple mutations associated with antibody escape in the VSV-SARS-CoV-2-S that had been passaged in the presence of antiserum, including key mutations present in currently circulating Omicron subvariants. These data indicate that viral stock that was generated under polyclonal antiserum selection pressure better reflects the natural environment of the circulating virus and may yield more biologically relevant outcomes in phenotypic assays. Thus, mAb assessment assays that utilize a more genetically diverse, biologically relevant, virus stock may yield data that are relevant for prediction of mAb efficacy and for enhancing biosurveillance.
Collapse
Affiliation(s)
- Aram Avila-Herrera
- Lawrence Livermore National Laboratory, Computing Directorate, Global Security Computing Applications Division, Livermore, California, United States of America
| | - Jeffrey A. Kimbrel
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| | - Jose Manuel Martí
- Lawrence Livermore National Laboratory, Computing Directorate, Global Security Computing Applications Division, Livermore, California, United States of America
| | - James Thissen
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| | - Edwin A. Saada
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| | - Tracy Weisenberger
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| | - Kathryn T. Arrildt
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| | - Brent W. Segelke
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| | - Jonathan E. Allen
- Lawrence Livermore National Laboratory, Computing Directorate, Global Security Computing Applications Division, Livermore, California, United States of America
| | - Adam Zemla
- Lawrence Livermore National Laboratory, Computing Directorate, Global Security Computing Applications Division, Livermore, California, United States of America
| | - Monica K. Borucki
- Lawrence Livermore National Laboratory, Physical and Life Sciences Directorate, Biosciences and Biotechnology Division, Livermore, California, United States of America
| |
Collapse
|
39
|
Bgatova N, Savchenko S, Lamanov A, Taskaeva I, Ayzikovich B, Gritcinger V, Letyagin A, Korolev M. Intracellular organelles remodeling in myocardial endotheliocytes in COVID-19: an autopsy-based study. Ultrastruct Pathol 2024; 48:66-74. [PMID: 38007715 DOI: 10.1080/01913123.2023.2286977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
It is known that the unfavorable outcome in patients infected with SARS-CoV-2 may be associated with the development of complications caused by heart damage due to the direct virus action. The mechanism of these cardiovascular injuries caused by SARS-CoV-2 infection has not been fully understood; however, the study of COVID-19-associated myocardial microcirculatory dysfunction can represent the useful strategy to solving this challenge. Thus, here we aimed to study the ultrastructural organization of endothelial cells of myocardial capillaries in patients with COVID-19. The morphology of endotheliocytes of the myocardial blood capillaries in patients with COVID-19 was studied on cardiac autopsy material using transmission electron microscopy. The endotheliocytes of myocardial capillaries in patients with COVID-19 were characterized by the abundant rough endoplasmic reticulum (ER) membranes, the Golgi complex, and free polysomal complexes of ribosomes and lipids. The presence of double membrane vesicles with virions and zippered ER was detected in the cytoplasm of endotheliocytes. The revealed endothelial ultrastructural changes indicate the remodeling of intracellular membranes during SARS-CoV-2 infection. Our findings confirm the formation of virus-induced structures in myocardial endothelial cells considered critical for viral replication and assembly. The data may elucidate the mechanisms of endothelial dysfunction development in patients with COVID-19 to provide potential targets for drug therapy.
Collapse
Affiliation(s)
- Nataliya Bgatova
- Research Institute of Clinical and Experimental Lymphology - Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Sergey Savchenko
- Department of Forensic Medicine, Novosibirsk State Medical University, Novosibirsk, Russia
| | - Alexei Lamanov
- Department of Forensic Medicine, Novosibirsk State Medical University, Novosibirsk, Russia
| | - Iuliia Taskaeva
- Research Institute of Clinical and Experimental Lymphology - Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Boris Ayzikovich
- Department of Forensic Medicine, Novosibirsk State Medical University, Novosibirsk, Russia
| | - Valentina Gritcinger
- Department of Forensic Medicine, Novosibirsk State Medical University, Novosibirsk, Russia
| | - Andrey Letyagin
- Research Institute of Clinical and Experimental Lymphology - Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Maksim Korolev
- Research Institute of Clinical and Experimental Lymphology - Branch of the Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
40
|
Baid K, Chiok KR, Banerjee A. Median Tissue Culture Infectious Dose 50 (TCID 50) Assay to Determine Infectivity of Cytopathic Viruses. Methods Mol Biol 2024; 2813:117-123. [PMID: 38888774 DOI: 10.1007/978-1-0716-3890-3_8] [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] [Indexed: 06/20/2024]
Abstract
The emergence of zoonotic viruses like severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and SARS-CoV-2 have significantly impacted global health and economy. The discovery of other viruses in wildlife reservoir species present a threat for future emergence in humans and animals. Therefore, assays that are less reliant on virus-specific information, such as neutralization assays, are crucial to rapidly develop diagnostics, understand virus replication and pathogenicity, and assess the efficacy of therapeutics against newly emerging viruses. Here, we describe the discontinuous median tissue culture infectious dose 50 (TCID50) assay to quantitatively determine the titer of any virus that can produce a visible cytopathic effect in infected cells.
Collapse
Affiliation(s)
- Kaushal Baid
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - Kim R Chiok
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada
| | - Arinjay Banerjee
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, SK, Canada.
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK, Canada.
- Department of Biology, University of Waterloo, Waterloo, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
41
|
Baran J, Kuryk Ł, Szczepińska T, Łaźniewski M, Garofalo M, Mazurkiewicz-Pisarek A, Mikiewicz D, Mazurkiewicz A, Trzaskowski M, Wieczorek M, Pancer K, Hallmann E, Brydak L, Plewczynski D, Ciach T, Mierzejewska J, Staniszewska M. In vitro immune evaluation of adenoviral vector-based platform for infectious diseases. BIOTECHNOLOGIA 2023; 104:403-419. [PMID: 38213479 PMCID: PMC10777723 DOI: 10.5114/bta.2023.132775] [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: 04/03/2023] [Revised: 09/12/2023] [Accepted: 09/29/2023] [Indexed: 01/13/2024] Open
Abstract
New prophylactic vaccine platforms are imperative to combat respiratory infections. The efficacy of T and B memory cell-mediated protection, generated through the adenoviral vector, was tested to assess the effectiveness of the new adenoviral-based platforms for infectious diseases. A combination of adenovirus AdV1 (adjuvant), armed with costimulatory ligands (ICOSL and CD40L), and rRBD (antigen: recombinant nonglycosylated spike protein rRBD) was used to promote the differentiation of T and B lymphocytes. Adenovirus AdV2 (adjuvant), without ligands, in combination with rRBD, served as a control. In vitro T-cell responses to the AdV1+rRBD combination revealed that CD8+ platform-specific T-cells increased (37.2 ± 0.7% vs. 23.1 ± 2.1%), and T-cells acted against SARS-CoV-2 via CD8+TEMRA (50.0 ± 1.3% vs. 36.0 ± 3.2%). Memory B cells were induced after treatment with either AdV1+rRBD (84.1 ± 0.8% vs. 82.3 ± 0.4%) or rRBD (94.6 ± 0.3% vs. 82.3 ± 0.4%). Class-switching from IgM and IgD to isotype IgG following induction with rRBD+Ab was observed. RNA-seq profiling identified gene expression patterns related to T helper cell differentiation that protect against pathogens. The analysis determined signaling pathways controlling the induction of protective immunity, including the MAPK cascade, adipocytokine, cAMP, TNF, and Toll-like receptor signaling pathway. The AdV1+rRBD formulation induced IL-6, IL-8, and TNF. RNA-seq of the VERO E6 cell line showed differences in the apoptosis gene expression stimulated with the platforms vs. mock. In conclusion, AdV1+rRBD effectively generates T and B memory cell-mediated protection, presenting promising results in producing CD8+ platform-specific T cells and isotype-switched IgG memory B cells. The platform induces protective immunity by controlling the Th1, Th2, and Th17 cell differentiation gene expression patterns. Further studies are required to confirm its effectiveness.
Collapse
Affiliation(s)
- Joanna Baran
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Łukasz Kuryk
- National Institute of Public Health, Warsaw, Poland
| | - Teresa Szczepińska
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Michał Łaźniewski
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | | | | | - Diana Mikiewicz
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Alina Mazurkiewicz
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | - Maciej Trzaskowski
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| | | | | | | | - Lidia Brydak
- National Institute of Public Health, Warsaw, Poland
| | - Dariusz Plewczynski
- Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
- Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Tomasz Ciach
- Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | | | - Monika Staniszewska
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Warsaw, Poland
| |
Collapse
|
42
|
Grootemaat AE, Wiersma N, van der Niet S, Schimmel IM, Florquin S, Reits EA, Miller SE, van der Wel NN. Nucleocapsid protein accumulates in renal tubular epithelium of a post-COVID-19 patient. Microbiol Spectr 2023; 11:e0302923. [PMID: 37975661 PMCID: PMC10715010 DOI: 10.1128/spectrum.03029-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/17/2023] [Indexed: 11/19/2023] Open
Abstract
IMPORTANCE Even though the coronavirus disease 2019 (COVID-19) pandemic is slowly developing into a conventional infectious disease, the long-term effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus infection are still not well understood. One of the problems is that many COVID-19 cases develop acute kidney injuries. Still, it is heavily debated whether SARS-CoV-2 virus enters and actively replicates in kidney tissue and if SARS-CoV-2 virus particles can be detected in kidney during or post-infection. Here, we demonstrated that nucleocapsid N protein was detected in kidney tubular epithelium of patients that already recovered form COVID-19. The presence of the abundantly produced N protein without signs of viral replication could have implications for the recurrence of kidney disease and have a continuing effect on the immune system.
Collapse
Affiliation(s)
- Anita E. Grootemaat
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre AMC, Amsterdam, the Netherlands
| | - Niek Wiersma
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre AMC, Amsterdam, the Netherlands
| | - Sanne van der Niet
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre AMC, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Irene M. Schimmel
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre AMC, Amsterdam, the Netherlands
| | - Sandrine Florquin
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Pathology, Amsterdam University Medical Centers (location University of Amsterdam), Amsterdam, the Netherlands
| | - Eric A. Reits
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre AMC, Amsterdam, the Netherlands
| | - Sara E. Miller
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Nicole N. van der Wel
- Electron Microscopy Centre Amsterdam, Medical Biology, Amsterdam University Medical Centre AMC, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| |
Collapse
|
43
|
Stewart H, Palmulli R, Johansen KH, McGovern N, Shehata OM, Carnell GW, Jackson HK, Lee JS, Brown JC, Burgoyne T, Heeney JL, Okkenhaug K, Firth AE, Peden AA, Edgar JR. Tetherin antagonism by SARS-CoV-2 ORF3a and spike protein enhances virus release. EMBO Rep 2023; 24:e57224. [PMID: 37818801 PMCID: PMC10702813 DOI: 10.15252/embr.202357224] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/23/2023] [Accepted: 09/21/2023] [Indexed: 10/13/2023] Open
Abstract
The antiviral restriction factor, tetherin, blocks the release of several different families of enveloped viruses, including the Coronaviridae. Tetherin is an interferon-induced protein that forms parallel homodimers between the host cell and viral particles, linking viruses to the surface of infected cells and inhibiting their release. We demonstrate that SARS-CoV-2 infection causes tetherin downregulation and that tetherin depletion from cells enhances SARS-CoV-2 viral titres. We investigate the potential viral proteins involved in abrogating tetherin function and find that SARS-CoV-2 ORF3a reduces tetherin localisation within biosynthetic organelles where Coronaviruses bud, and increases tetherin localisation to late endocytic organelles via reduced retrograde recycling. We also find that expression of Spike protein causes a reduction in cellular tetherin levels. Our results confirm that tetherin acts as a host restriction factor for SARS-CoV-2 and highlight the multiple distinct mechanisms by which SARS-CoV-2 subverts tetherin function.
Collapse
Affiliation(s)
- Hazel Stewart
- Department of PathologyUniversity of CambridgeCambridgeUK
| | | | - Kristoffer H Johansen
- Department of PathologyUniversity of CambridgeCambridgeUK
- Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMDUSA
| | - Naomi McGovern
- Department of PathologyUniversity of CambridgeCambridgeUK
| | - Ola M Shehata
- Department of Biomedical ScienceUniversity of Sheffield, Firth CourtSheffieldUK
| | - George W Carnell
- Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| | | | - Jin S Lee
- Department of PathologyUniversity of CambridgeCambridgeUK
| | | | - Thomas Burgoyne
- Royal Brompton HospitalGuy's and St Thomas' NHS Foundation TrustLondonUK
- UCL Institute of OphthalmologyUniversity College LondonLondonUK
| | | | | | - Andrew E Firth
- Department of PathologyUniversity of CambridgeCambridgeUK
| | - Andrew A Peden
- Department of Biomedical ScienceUniversity of Sheffield, Firth CourtSheffieldUK
| | - James R Edgar
- Department of PathologyUniversity of CambridgeCambridgeUK
| |
Collapse
|
44
|
Termansen MB, Frische S. Fecal-oral transmission of SARS-CoV-2: A systematic review of evidence from epidemiological and experimental studies. Am J Infect Control 2023; 51:1430-1437. [PMID: 37121473 PMCID: PMC10141930 DOI: 10.1016/j.ajic.2023.04.170] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/21/2023] [Accepted: 04/21/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND SARS-CoV-2 ribonucleic acid (RNA) has been detected in feces, but RNA is not infectious. This systematic review aims to answer if fecal SARS-CoV-2 is experimentally infectious and if evidence of human fecal-oral SARS-CoV-2 transmission exists. METHODS On September 19, 2022, we searched PubMed, Embase, Web of Science, medRxiv, and bioRxiv. Biomedical studies inoculating SARS-CoV-2 from feces, rectal, or anal swabs in cells, tissue, organoids, or animals were included. Epidemiological studies of groups differing in exposure to fecal SARS-CoV-2 were included. Risk of bias was assessed using standardized tools. Results were summarized by vote counting, tabulation, and a harvest plot. PROSPERO registration no. CRD42020221719. RESULTS A total of 4,874 studies were screened; 26 studies were included; and 13 out of 23 biomedical studies (56.5%) succeeded in infection. Two (66.7%) epidemiological studies found limited evidence suggesting fecal-oral transmission. All studies had concerns about the risk of bias. CONCLUSIONS It is possible to experimentally infect cell cultures, organoids, and animals with fecal SARS-CoV-2. No strong epidemiologic evidence was found to support human fecal-oral transmission. We advise future research to study fecal infectivity at different time points during infection, apply appropriate controls, use in vivo models, and study fecal exposure as a risk factor of transmission in human populations.
Collapse
|
45
|
Lu S, Luo S, Liu C, Li M, An X, Li M, Hou J, Fan H, Mao P, Tong Y, Song L. Induction of significant neutralizing antibodies against SARS-CoV-2 by a highly attenuated pangolin coronavirus variant with a 104nt deletion at the 3'-UTR. Emerg Microbes Infect 2023; 12:2151383. [PMID: 36453209 PMCID: PMC9769135 DOI: 10.1080/22221751.2022.2151383] [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] [Indexed: 12/05/2022]
Abstract
SARS-CoV-2 related coronaviruses (SARS-CoV-2r) from Guangdong and Guangxi pangolins have been implicated in the emergence of SARS-CoV-2 and future pandemics. We previously reported the culture of a SARS-CoV-2r GX_P2V from Guangxi pangolins. Here we report the GX_P2V isolate rapidly adapted to Vero cells by acquiring two genomic mutations: an alanine to valine substitution in the nucleoprotein and a 104-nucleotide deletion in the hypervariable region (HVR) of the 3'-terminus untranslated region (3'-UTR). We further report the characterization of the GX_P2V variant (renamed GX_P2V(short_3UTR)) in in vitro and in vivo infection models. In cultured Vero, BGM and Calu-3 cells, GX_P2V(short_3UTR) had similar robust replication kinetics, and consistently produced minimum cell damage. GX_P2V(short_3UTR) infected golden hamsters and BALB/c mice but was highly attenuated. Golden hamsters infected intranasally had a short duration of productive infection in pulmonary, not extrapulmonary, tissues. These productive infections induced neutralizing antibodies against pseudoviruses of GX_P2V and SARS-CoV-2. Collectively, our data show that the GX_P2V(short_3UTR) is highly attenuated in in vitro and in vivo infection models. Attenuation of the variant is likely partially due to the 104-nt deletion in the HVR in the 3'-UTR. This study furthers our understanding of pangolin coronaviruses pathogenesis and provides novel insights for the design of live attenuated vaccines against SARS-CoV-2.
Collapse
Affiliation(s)
- Shanshan Lu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Shengdong Luo
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Chang Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Muli Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, People’s Republic of China
| | - Xiaoping An
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Mengzhe Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Jun Hou
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Huahao Fan
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China,Huahao Fan Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Panyong Mao
- Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China,Panyong Mao Department of Infectious Diseases, Fifth Medical Center of Chinese PLA General Hospital, Beijing, People’s Republic of China
| | - Yigang Tong
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China,Yigang Tong Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Lihua Song
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China, Lihua Song Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| |
Collapse
|
46
|
Castro V, Pérez-Berna AJ, Calvo G, Pereiro E, Gastaminza P. Three-Dimensional Remodeling of SARS-CoV2-Infected Cells Revealed by Cryogenic Soft X-ray Tomography. ACS NANO 2023; 17:22708-22721. [PMID: 37939169 PMCID: PMC10690842 DOI: 10.1021/acsnano.3c07265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/10/2023]
Abstract
Plus-strand RNA viruses are proficient at remodeling host cell membranes for optimal viral genome replication and the production of infectious progeny. These ultrastructural alterations result in the formation of viral membranous organelles and may be observed by different imaging techniques, providing nanometric resolution. Guided by confocal and electron microscopy, this study describes the generation of wide-field volumes using cryogenic soft-X-ray tomography (cryo-SXT) on SARS-CoV-2-infected human lung adenocarcinoma cells. Confocal microscopy showed accumulation of double-stranded RNA (dsRNA) and nucleocapsid (N) protein in compact perinuclear structures, preferentially found around centrosomes at late stages of the infection. Transmission electron microscopy (TEM) showed accumulation of membranous structures in the vicinity of the infected cell nucleus, forming a viral replication organelle containing characteristic double-membrane vesicles and virus-like particles within larger vesicular structures. Cryo-SXT revealed viral replication organelles very similar to those observed by TEM but indicated that the vesicular organelle observed in TEM sections is indeed a vesiculo-tubular network that is enlarged and elongated at late stages of the infection. Overall, our data provide additional insight into the molecular architecture of the SARS-CoV-2 replication organelle.
Collapse
Affiliation(s)
- Victoria Castro
- Centro
Nacional de Biotecnología. Calle Darwin, 3, 28049 Madrid, Spain
| | | | - Gema Calvo
- Centro
Nacional de Biotecnología. Calle Darwin, 3, 28049 Madrid, Spain
| | - Eva Pereiro
- ALBA
Synchrotron Light Source, Carrer de la Llum 2-26, 08290 Cerdanyola del Valles, Spain
| | - Pablo Gastaminza
- Centro
Nacional de Biotecnología. Calle Darwin, 3, 28049 Madrid, Spain
| |
Collapse
|
47
|
Ali M, Park IH, Kim J, Kim G, Oh J, You JS, Kim J, Shin JS, Yoon SS. How Deep Learning in Antiviral Molecular Profiling Identified Anti-SARS-CoV-2 Inhibitors. Biomedicines 2023; 11:3134. [PMID: 38137356 PMCID: PMC10740425 DOI: 10.3390/biomedicines11123134] [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: 10/18/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
The integration of artificial intelligence (AI) into drug discovery has markedly advanced the search for effective therapeutics. In our study, we employed a comprehensive computational-experimental approach to identify potential anti-SARS-CoV-2 compounds. We developed a predictive model to assess the activities of compounds based on their structural features. This model screened a library of approximately 700,000 compounds, culminating in the selection of the top 100 candidates for experimental validation. In vitro assays on human intestinal epithelial cells (Caco-2) revealed that 19 of these compounds exhibited inhibitory activity. Notably, eight compounds demonstrated dose-dependent activity in Vero cell lines, with half-maximal effective concentration (EC50) values ranging from 1 μM to 7 μM. Furthermore, we utilized a clustering approach to pinpoint potential nucleoside analog inhibitors, leading to the discovery of two promising candidates: azathioprine and its metabolite, thioinosinic acid. Both compounds showed in vitro activity against SARS-CoV-2, with thioinosinic acid also significantly reducing viral loads in mouse lungs. These findings underscore the utility of AI in accelerating drug discovery processes.
Collapse
Affiliation(s)
- Mohammed Ali
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - In Ho Park
- Department of Biomedical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Junebeom Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Gwanghee Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jooyeon Oh
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jin Sun You
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jieun Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jeon-Soo Shin
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sang Sun Yoon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (M.A.); (J.K.); (G.K.); (J.O.); (J.S.Y.); (J.K.)
- Brain Korea 21 Project for Medical Sciences, Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Institute of Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- BioMe Inc., Seoul 02455, Republic of Korea
| |
Collapse
|
48
|
Liu W, Zhang M, Hu C, Song H, Mei Y, Liu Y, Zhang Q. Remdesivir Derivative VV116 Is a Potential Broad-Spectrum Inhibitor of Both Human and Animal Coronaviruses. Viruses 2023; 15:2295. [PMID: 38140536 PMCID: PMC10748125 DOI: 10.3390/v15122295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
Coronaviruses represent a significant threat to both human and animal health, encompassing a range of pathogenic strains responsible for illnesses, from the common cold to more severe diseases. VV116 is a deuterated derivative of Remdesivir with oral bioavailability that was found to potently inhibit SARS-CoV-2. In this work, we investigated the broad-spectrum antiviral activity of VV116 against a variety of human and animal coronaviruses. We examined the inhibitory effects of VV116 on the replication of the human coronaviruses HCoV-NL63, HCoV-229E, and HCoV-OC43, as well as the animal coronaviruses MHV, FIPV, FECV, and CCoV. The findings reveal that VV116 effectively inhibits viral replication across these strains without exhibiting cytotoxicity, indicating its potential for safe therapeutic use. Based on the results of a time-of-addition assay and an rNTP competitive inhibition assay, it is speculated that the inhibitory mechanism of VV116 against HCoV-NL63 is consistent with its inhibition of SARS-CoV-2. Our work presents VV116 as a promising candidate for broad-spectrum anti-coronavirus therapy, with implications for both human and animal health, and supports the expansion of its therapeutic applications as backed by detailed experimental data.
Collapse
Affiliation(s)
- Weiyong Liu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (W.L.); (C.H.); (H.S.)
| | - Min Zhang
- Department of Emergency Medicine, The Third People’s Hospital of Hubei Province, Wuhan 430033, China;
| | - Chengxiu Hu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (W.L.); (C.H.); (H.S.)
| | - Huijuan Song
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (W.L.); (C.H.); (H.S.)
| | - Yi Mei
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China;
| | - Yingle Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China;
| | - Qi Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China;
| |
Collapse
|
49
|
Rudramurthy GR, Naveenkumar CN, Bharathkumar K, Shandil RK, Narayanan S. Genomic Mutations in SARS-CoV-2 Genome following Infection in Syrian Golden Hamster and Associated Lung Pathologies. Pathogens 2023; 12:1328. [PMID: 38003792 PMCID: PMC10674674 DOI: 10.3390/pathogens12111328] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
The continuous evolution of the SARS-CoV-2 virus led to constant developments and efforts in understanding the significance and impacts of SARS-CoV-2 variants on human health. Our study aimed to determine the accumulation of genetic mutations and associated lung pathologies in male and female hamsters infected with the ancestral Wuhan strain of SARS-CoV-2. The present study showed no significant difference in the viral load between male and female hamsters and peak infection was found to be on day four post infection in both sexes of the animals. Live virus particles were detected up to 5 days post infection (dpi) through the TCID-50 assay, while qRT-PCR could detect viral RNA up to 14 dpi from all the infected animals. Further, the determination of the neutralizing antibody titer showed the onset of the humoral immune response as early as 4 dpi in both sexes against SARS-CoV-2, and a significant cross-protection against the delta variant of SARS-CoV-2 was observed. Histopathology showed edema, inflammation, inflammatory cell infiltration, necrosis, and degeneration of alveolar and bronchial epithelium cells from 3 dpi to 14 dpi in both sexes. Furthermore, next-generation sequencing (NGS) showed up to 10 single-nucleotide polymorphisms (SNPs) in the SARS-CoV-2 (ancestral Wuhan strain) genome isolated from both male and female hamsters. The mutation observed at the 23014 position (Glu484Asp) in the SARS-CoV-2 genome isolated from both sexes of the hamsters plays a significant role in the antiviral efficacy of small molecules, vaccines, and the Mabs-targeting S protein. The present study shows that either of the genders can be used in the pre-clinical efficacy of antiviral agents against SARS-CoV-2 in hamsters. However, considering the major mutation in the S protein, the understanding of the genetic mutation in SARS-CoV-2 after passing through hamsters is crucial in deciding the efficacy of the antiviral agents targeting the S protein. Importance: Our study findings indicate the accumulation of genomic mutations in SARS-CoV-2 after passing through the Syrian golden hamsters. Understanding the genomic mutations showed that either of the hamster genders can be used in the pre-clinical efficacy of antiviral agents and vaccines.
Collapse
Affiliation(s)
- Gudepalya Renukaiah Rudramurthy
- Foundation for Neglected Disease Research (FNDR), Plot No. 20A, KIADB Industrial Area, Bengaluru 561203, Karnataka, India; (C.N.N.); (K.B.); (R.K.S.); (S.N.)
| | | | | | | | | |
Collapse
|
50
|
Miyashita L, Foley G, Semple S, Gibbons JM, Pade C, McKnight Á, Grigg J. Curbside particulate matter and susceptibility to SARS-CoV-2 infection. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100141. [PMID: 37781647 PMCID: PMC10509961 DOI: 10.1016/j.jacig.2023.100141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/22/2023] [Accepted: 06/05/2023] [Indexed: 10/03/2023]
Abstract
Background Biologic plausibility for the association between exposure to particulate matter (PM) less than 10 μm in aerodynamic diameter (PM10) and coronavirus disease 2019 (COVID-19) morbidity in epidemiologic studies has not been determined. The upregulation of angiotensin-converting enzyme 2 (ACE2), the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) entry receptor on host cells, by PM10 is a putative mechanism. Objective We sought to assess the effect of PM10 on SARS-CoV-2 infection of cells in vitro. Methods PM10 from the curbside of London's Marylebone Road and from exhaust emissions was collected by cyclone. A549 cells, human primary nasal epithelial cells (HPNEpCs), SARS-CoV-2-susceptible Vero-E6 and Calu3 cells were cultured with PM10. ACE2 expression (as determined by median fluorescent intensity) was assessed by flow cytometry, and ACE2 mRNA transcript level was assessed by PCR. The role of oxidative stress was determined by N-acetyl cysteine. The cytopathic effect of SARS-CoV-2 (percentage of infection enhancement) and expression of SARS-CoV-2 genes' open reading frame (ORF) 1ab, S protein, and N protein (focus-forming units/mL) were assessed in Vero-E6 cells. Data were analyzed by either the Mann-Whitney U test or Kruskal-Wallis test with the Dunn multiple comparisons test. Results Curbside PM10 at concentrations of 10 μg/mL or more increased ACE2 expression in A549 cells (P = .0021). Both diesel PM10 and curbside PM10 in a concentration of 10 μg/mL increased ACE2 expression in HPNEpCs (P = .0022 and P = .0072, respectively). ACE2 expression simulated by curbside PM10 was attenuated by N-acetyl cysteine in HPNEpCs (P = .0464). Curbside PM10 increased ACE2 expression in Calu3 cells (P = .0256). In Vero-E6 cells, curbside PM10 increased ACE2 expression (P = .0079), ACE2 transcript level (P = .0079), SARS-CoV-2 cytopathic effect (P = .0002), and expression of the SARS-CoV-2 genes' ORF1ab, S protein, and N protein (P = .0079). Conclusions Curbside PM10 increases susceptibility to SARS-COV-2 infection in vitro.
Collapse
Affiliation(s)
- Lisa Miyashita
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Gary Foley
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Sean Semple
- Institute for Social Marketing, University of Stirling, Stirling, United Kingdom
| | - Joseph M. Gibbons
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Corinna Pade
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Áine McKnight
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Jonathan Grigg
- Blizard Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|