1
|
Reuss D, Brown JC, Sukhova K, Furnon W, Cowton V, Patel AH, Palmarini M, Thompson C, Barclay WS. Interference between SARS-CoV-2 and influenza B virus during coinfection is mediated by induction of specific interferon responses in the lung epithelium. Virology 2025; 608:110556. [PMID: 40318419 DOI: 10.1016/j.virol.2025.110556] [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: 02/20/2025] [Revised: 04/15/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Coinfections with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza virus have represented a major health concern since the beginning of the COVID-19 pandemic. The continued spread and constant emergence of new SARS-CoV-2 variants mean that cocirculation and coinfection with seasonal respiratory viruses will continue. Despite the considerable contribution of influenza B virus (IBV) infections to global disease burdens, its interactions with SARS-CoV-2 remain largely unstudied. In this study, we sequentially coinfected lung epithelial cells with representative SARS-CoV-2 variants and IBV strains. We found that prior infection with IBV impaired SARS-CoV-2 D614G, Delta and Omicron BA.1 replication, but did not affect replication of the more recent Omicron EG.5.1 variant. We additionally show that pre-infection with SARS-CoV-2 reduces live attenuated influenza vaccine (LAIV) replication, suggesting vaccine effectiveness in children carrying SARS-CoV-2 pre-infections can be negatively impacted in coinfection. Both SARS-CoV-2 and IBV induced strong type III interferon (IFN) responses, whereas SARS-CoV-2 drove type I IFN production not seen in IBV infection, suggesting viral interference through specific IFN responses. Treatment with innate immune response inhibitors BX795 and Ruxolitinib abrogated viral interference between IBV and SARS-CoV-2 in coinfection, demonstrating that IFN-stimulated gene (ISG) responses play a vital role in viral interference. More specifically, we show that the magnitude and timing of ISG expression, triggered by the primary infecting virus in sequential coinfection, facilitates viral interference between IBV and SARS-CoV-2.
Collapse
Affiliation(s)
- Dorothee Reuss
- Department of Infectious Disease, Imperial College London, UK.
| | | | - Ksenia Sukhova
- Department of Infectious Disease, Imperial College London, UK
| | - Wilhelm Furnon
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Vanessa Cowton
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Arvind H Patel
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | | | - Wendy S Barclay
- Department of Infectious Disease, Imperial College London, UK.
| |
Collapse
|
2
|
Michalak KP, Michalak AZ, Brenk-Krakowska A. Acute COVID-19 and LongCOVID syndrome - molecular implications for therapeutic strategies - review. Front Immunol 2025; 16:1582783. [PMID: 40313948 PMCID: PMC12043656 DOI: 10.3389/fimmu.2025.1582783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/28/2025] [Indexed: 05/03/2025] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has been recognized not only for its acute effects but also for its ability to cause LongCOVID Syndrome (LCS), a condition characterized by persistent symptoms affecting multiple organ systems. This review examines the molecular and immunological mechanisms underlying LCS, with a particular focus on autophagy inhibition, chronic inflammation, oxidative, nitrosative and calcium stress, viral persistence and autoimmunology. Potential pathophysiological mechanisms involved in LCS include (1) autoimmune activation, (2) latent viral persistence, where SARS-CoV-2 continues to influence host metabolism, (3) reactivation of latent pathogens such as Epstein-Barr virus (EBV) or cytomegalovirus (CMV), exacerbating immune and metabolic dysregulation, and (4) possible persistent metabolic and inflammatory dysregulation, where the body fails to restore post-infection homeostasis. The manipulation of cellular pathways by SARS-CoV-2 proteins is a critical aspect of the virus' ability to evade immune clearance and establish long-term dysfunction. Viral proteins such as NSP13, ORF3a and ORF8 have been shown to disrupt autophagy, thereby impairing viral clearance and promoting immune evasion. In addition, mitochondrial dysfunction, dysregulated calcium signaling, oxidative stress, chronic HIF-1α activation and Nrf2 inhibition create a self-sustaining inflammatory feedback loop that contributes to tissue damage and persistent symptoms. Therefore understanding the molecular basis of LCS is critical for the development of effective therapeutic strategies. Targeting autophagy and Nrf2 activation, glycolysis inhibition, and restoration calcium homeostasis may provide novel strategies to mitigate the long-term consequences of SARS-CoV-2 infection. Future research should focus on personalized therapeutic interventions based on the dominant molecular perturbations in individual patients.
Collapse
Affiliation(s)
- Krzysztof Piotr Michalak
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | | - Alicja Brenk-Krakowska
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| |
Collapse
|
3
|
Janevska M, Naessens E, Verhasselt B. Impact of SARS-CoV-2 Wuhan and Omicron Variant Proteins on Type I Interferon Response. Viruses 2025; 17:569. [PMID: 40285011 PMCID: PMC12031613 DOI: 10.3390/v17040569] [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: 03/08/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
SARS-CoV-2 has demonstrated a remarkable capacity for immune evasion. While initial studies focused on the Wuhan variant and adaptive immunity, later emerging strains such as Omicron exhibit mutations that may alter their immune-modulatory properties. We performed a comprehensive review of immune evasion mechanisms associated with SARS-CoV-2 viral proteins to focus on the evolutionary dynamics of immune modulation. We systematically analyzed and compared the impact of all currently known Wuhan and Omicron SARS-CoV-2 proteins on type I interferon (IFN) responses using a dual-luciferase reporter assay carrying an interferon-inducible promoter. Results revealed that Nsp1, Nsp5, Nsp14, and ORF6 are potent type I IFN inhibitors conserved across Wuhan and Omicron strains. Notably, we identified strain-specific differences, with Nsp6 and Spike proteins exhibiting enhanced IFN suppression in Omicron, whereas the Envelope protein largely retained this function. To extend these findings, we investigated selected proteins in primary human endothelial cells and also observed strain-specific differences in immune response with higher type I IFN response in cells expressing the Wuhan strain variant, suggesting that Omicron's adaptational mutations may contribute to a damped type I IFN response in the course of the pandemic's trajectory.
Collapse
Affiliation(s)
- Marija Janevska
- Department of Diagnostic Sciences, Ghent University, B9000 Ghent, Belgium;
| | - Evelien Naessens
- Department of Laboratory Medicine, Ghent University Hospital, B9000 Ghent, Belgium;
| | - Bruno Verhasselt
- Department of Diagnostic Sciences, Ghent University, B9000 Ghent, Belgium;
- Department of Laboratory Medicine, Ghent University Hospital, B9000 Ghent, Belgium;
| |
Collapse
|
4
|
Khatun O, Kaur S, Tripathi S. Anti-interferon armamentarium of human coronaviruses. Cell Mol Life Sci 2025; 82:116. [PMID: 40074984 PMCID: PMC11904029 DOI: 10.1007/s00018-025-05605-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/15/2024] [Accepted: 01/23/2025] [Indexed: 03/14/2025]
Abstract
Cellular innate immune pathways are formidable barriers against viral invasion, creating an environment unfavorable for virus replication. Interferons (IFNs) play a crucial role in driving and regulating these cell-intrinsic innate antiviral mechanisms through the action of interferon-stimulated genes (ISGs). The host IFN response obstructs viral replication at every stage, prompting viruses to evolve various strategies to counteract or evade this response. Understanding the interplay between viral proteins and cell-intrinsic IFN-mediated immune mechanisms is essential for developing antiviral and anti-inflammatory strategies. Human coronaviruses (HCoVs), including SARS-CoV-2, MERS-CoV, SARS-CoV, and seasonal coronaviruses, encode a range of proteins that, through shared and distinct mechanisms, inhibit IFN-mediated innate immune responses. Compounding the issue, a dysregulated early IFN response can lead to a hyper-inflammatory immune reaction later in the infection, resulting in severe disease. This review provides a brief overview of HCoV replication and a detailed account of its interaction with host cellular innate immune pathways regulated by IFN.
Collapse
Affiliation(s)
- Oyahida Khatun
- Emerging Viral Pathogens Laboratory, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, India
- Microbiology & Cell Biology Department, Biological Sciences Division, Indian Institute of Science, Bengaluru, India
| | - Sumandeep Kaur
- Emerging Viral Pathogens Laboratory, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, India
- Microbiology & Cell Biology Department, Biological Sciences Division, Indian Institute of Science, Bengaluru, India
| | - Shashank Tripathi
- Emerging Viral Pathogens Laboratory, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, India.
- Microbiology & Cell Biology Department, Biological Sciences Division, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
5
|
Pagani I, Venturini A, Capurro V, Nonis A, Ghezzi S, Lena M, Alcalá-Franco B, Gianferro F, Guidone D, Colombo C, Pedemonte N, Bragonzi A, Cigana C, Galietta LJV, Vicenzi E. Distinct Responses of Cystic Fibrosis Epithelial Cells to SARS-CoV-2 and Influenza A Virus. Am J Respir Cell Mol Biol 2025; 72:308-319. [PMID: 39311876 PMCID: PMC11890075 DOI: 10.1165/rcmb.2024-0213oc] [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: 05/06/2024] [Accepted: 09/23/2024] [Indexed: 03/01/2025] Open
Abstract
The coronavirus disease (COVID-19) pandemic has underscored the impact of viral infections on individuals with cystic fibrosis (CF). Initial observations suggested lower COVID-19 rates among CF populations; however, subsequent clinical data have presented a more complex scenario. This study aimed to investigate how bronchial epithelial cells from individuals with and without CF, including various CFTR (CF transmembrane conductance regulator) mutations, respond to in vitro infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants and SARS-CoV. Comparisons with the influenza A virus (IAV) were included based on evidence that patients with CF experience heightened morbidity from IAV infection. Our findings showed that CF epithelial cells exhibited reduced replication of SARS-CoV-2, regardless of the type of CFTR mutation or SARS-CoV-2 variant, as well as the original 2003 SARS-CoV. In contrast, these cells displayed more efficient IAV replication than non-CF cells. Interestingly, the reduced susceptibility to SARS-CoV-2 in CF was not linked to the expression of ACE2 (angiotensin-converting enzyme 2) receptor or to CFTR dysfunction, as pharmacological treatments to restore CFTR function did not normalize the viral response. Both SARS-CoV-2 infection and CFTR function influenced the concentrations of certain cytokines and chemokines, although these effects were not correlated. Overall, this study reveals a unique viral response in CF epithelial cells, characterized by reduced replication for some viruses like SARS-CoV-2, while showing increased susceptibility to others, such as IAV. This research offers a new perspective on CF and viral interactions, emphasizing the need for further investigation into the mechanisms underlying these differences.
Collapse
Affiliation(s)
| | - Arianna Venturini
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Department of Translational Medical Sciences, University of Napoli Federico II, Naples, Italy
| | - Valeria Capurro
- Unit of Medical Genetics (UOC), IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Alessandro Nonis
- University Centre for Statistics in the Biomedical Sciences (CUSSB), Vita-Salute San Raffaele University, Milan, Italy
| | | | - Mariateresa Lena
- Unit of Medical Genetics (UOC), IRCCS Giannina Gaslini Institute, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy; and
| | - Beatriz Alcalá-Franco
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabrizio Gianferro
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Guidone
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Department of Translational Medical Sciences, University of Napoli Federico II, Naples, Italy
| | - Carla Colombo
- Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Nicoletta Pedemonte
- Unit of Medical Genetics (UOC), IRCCS Giannina Gaslini Institute, Genoa, Italy
| | - Alessandra Bragonzi
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Cigana
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luis J. V. Galietta
- Telethon Institute of Genetics and Medicine, Pozzuoli, Italy
- Department of Translational Medical Sciences, University of Napoli Federico II, Naples, Italy
| | | |
Collapse
|
6
|
de Oliveira Silva Pinto M, de Paula Pereira L, de Mendonça Angelo ALP, Xavier MAP, de Magalhães Vieira Machado A, Russo RC. Dissecting the COVID-19 Immune Response: Unraveling the Pathways of Innate Sensing and Response to SARS-CoV-2 Structural Proteins. J Mol Recognit 2025; 38:e70002. [PMID: 39905998 DOI: 10.1002/jmr.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV), the virus responsible for COVID-19, interacts with the host immune system through complex mechanisms that significantly influence disease outcomes, affecting both innate and adaptive immunity. These interactions are crucial in determining the disease's severity and the host's ability to clear the virus. Given the virus's substantial socioeconomic impact, high morbidity and mortality rates, and public health importance, understanding these mechanisms is essential. This article examines the diverse innate immune responses triggered by SARS-CoV-2's structural proteins, including the spike (S), membrane (M), envelope (E), and nucleocapsid (N) proteins, along with nonstructural proteins (NSPs) and open reading frames. These proteins play pivotal roles in immune modulation, facilitating viral replication, evading immune detection, and contributing to severe inflammatory responses such as cytokine storms and acute respiratory distress syndrome (ARDS). The virus employs strategies like suppressing type I interferon production and disrupting key antiviral pathways, including MAVS, OAS-RNase-L, and PKR. This study also explores the immune pathways that govern the activation and suppression of immune responses throughout COVID-19. By analyzing immune sensing receptors and the responses initiated upon recognizing SARS-CoV-2 structural proteins, this review elucidates the complex pathways associated with the innate immune response in COVID-19. Understanding these mechanisms offers valuable insights for therapeutic interventions and informs public health strategies, contributing to a deeper understanding of COVID-19 immunopathogenesis.
Collapse
Affiliation(s)
- Matheus de Oliveira Silva Pinto
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
- Viral Disease Immunology Group, Fundação Osvaldo Cruz, Instituto René Rachou, Belo Horizonte, Minas Gerais, Brazil
| | - Leonardo de Paula Pereira
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
- Viral Disease Immunology Group, Fundação Osvaldo Cruz, Instituto René Rachou, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
7
|
Ammara M, Samiry I, Zaid Y, Oudghiri M, Naya A. A Pilot Study on the Role of TRAFs in the Development of SARS-CoV-2 Infection Before and After Immunization with AstraZeneca Chadox1 in Mice. Curr Issues Mol Biol 2025; 47:165. [PMID: 40136419 PMCID: PMC11941553 DOI: 10.3390/cimb47030165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
The TRAF family of molecules are intracellular signaling adaptors that regulate various signaling pathways. These pathways are not only mediated by the TNFR superfamily and the Toll-like receptor/IL-1 receptor superfamily but also by unconventional cytokine receptors like IL-6 and IL-17 receptors. Overactive immune responses caused by TRAF signaling following the activation of these receptors frequently result in inflammatory and autoimmune diseases such as rheumatoid arthritis, inflammatory bowel disease, psoriasis, and autoinflammatory syndromes. Therefore, it is crucial to comprehend the signaling processes controlled by TRAFs, which have a significant influence on the determination of cell fate (life or death) and the functioning, specialization, and endurance of cells in the innate and adaptive immune systems. Our data indicate that the dysregulation of cellular expression and/or signaling of TRAFs leads to the excessive production of pro-inflammatory cytokines, hence promoting abnormal activation of immune cells. The objective of our investigation was to comprehend the function of these molecules in SARS-CoV-2 infection both prior to and during SARS-CoV-2 vaccination. Our results demonstrate a clear inactivation of the TRAF5 and TRAF6 genes when infection occurs after immunization, in contrast to infection without prior vaccination. This can bolster the belief that immunization is essential while also demonstrating the involvement of these molecules in the pathogenesis of SARS-CoV-2.
Collapse
Affiliation(s)
- Mounia Ammara
- Immunology and Biodiversity Laboratory, Department of Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20000, Morocco; (M.A.); (I.S.); (Y.Z.); (M.O.)
| | - Inass Samiry
- Immunology and Biodiversity Laboratory, Department of Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20000, Morocco; (M.A.); (I.S.); (Y.Z.); (M.O.)
| | - Younes Zaid
- Immunology and Biodiversity Laboratory, Department of Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20000, Morocco; (M.A.); (I.S.); (Y.Z.); (M.O.)
- Materials, Nanotechnologies and Environment Laboratory, Department of Biology, Faculty of Sciences, Mohammed V University, Rabat 10000, Morocco
| | - Mounia Oudghiri
- Immunology and Biodiversity Laboratory, Department of Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20000, Morocco; (M.A.); (I.S.); (Y.Z.); (M.O.)
| | - Abdallah Naya
- Immunology and Biodiversity Laboratory, Department of Biology, Faculty of Sciences Ain Chock, Hassan II University, Casablanca 20000, Morocco; (M.A.); (I.S.); (Y.Z.); (M.O.)
| |
Collapse
|
8
|
San Felipe CJ, Batra J, Muralidharan M, Malpotra S, Anand D, Bauer R, Verba KA, Swaney DL, Krogan NJ, Grabe M, Fraser JS. Coupled equilibria of dimerization and lipid binding modulate SARS Cov 2 Orf9b interactions and interferon response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.16.638509. [PMID: 40027672 PMCID: PMC11870501 DOI: 10.1101/2025.02.16.638509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Open Reading Frame 9b (Orf9b), an accessory protein of SARS-CoV and -2, is involved in innate immune suppression through its binding to the mitochondrial receptor Translocase of Outer Membrane 70 (Tom70). Previous structural studies of Orf9b in isolation revealed a β-sheet-rich homodimer, however, structures of Orf9b in complex with Tom70 revealed a monomeric helical fold. Here, we developed a biophysical model that quantifies how Orf9b switches between these conformations and binds to Tom70, a requirement for suppressing the type 1 interferon response. We used this model to characterize the effect of lipid binding and mutations in variants of concern to the Orf9b:Tom70 equilibrium. We found that the binding of a lipid to the Orf9b homodimer biases the Orf9b monomer:dimer equilibrium towards the dimer by reducing the dimer dissociation rate ∼100-fold. We also found that mutations in variants of concern can alter different microscopic rate constants without significantly affecting binding to Tom70. Together our results highlight how perturbations to different steps in these coupled equilibria can affect the apparent affinity of Orf9b to Tom70, with potential downstream implications for interferon signaling in coronavirus infection.
Collapse
Affiliation(s)
- CJ San Felipe
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158
| | - Jyoti Batra
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA 94158
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, 94158, California,USA
- Department of Cellular and Molecular Pharmacology,University of California San Francisco, San Francisco, CA 94158
| | - Monita Muralidharan
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA 94158
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, 94158, California,USA
- Department of Cellular and Molecular Pharmacology,University of California San Francisco, San Francisco, CA 94158
| | - Shivali Malpotra
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA 94158
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, 94158, California,USA
- Department of Cellular and Molecular Pharmacology,University of California San Francisco, San Francisco, CA 94158
| | - Durga Anand
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA 94158
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, 94158, California,USA
- Department of Cellular and Molecular Pharmacology,University of California San Francisco, San Francisco, CA 94158
| | - Rachel Bauer
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA 94158
- Department of Cellular and Molecular Pharmacology,University of California San Francisco, San Francisco, CA 94158
| | - Kliment A Verba
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA 94158
- Department of Cellular and Molecular Pharmacology,University of California San Francisco, San Francisco, CA 94158
| | - Danielle L. Swaney
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA 94158
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, 94158, California,USA
- Department of Cellular and Molecular Pharmacology,University of California San Francisco, San Francisco, CA 94158
| | - Nevan J. Krogan
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA 94158
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, 94158, California,USA
- Department of Cellular and Molecular Pharmacology,University of California San Francisco, San Francisco, CA 94158
| | - Michael Grabe
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158
| | - James S. Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158
- Quantitative Biosciences Institute, University of California San Francisco, San Francisco, CA 94158
| |
Collapse
|
9
|
Zodda E, Pons M, DeMoya-Valenzuela N, Calvo-González C, Benítez-Rodríguez C, López-Ayllón BD, Hibot A, Zuin A, Crosas B, Cascante M, Montoya M, Pujol MD, Rubio-Martínez J, Thomson TM. Induction of the Inflammasome by the SARS-CoV-2 Accessory Protein ORF9b, Abrogated by Small-Molecule ORF9b Homodimerization Inhibitors. J Med Virol 2025; 97:e70145. [PMID: 39902616 DOI: 10.1002/jmv.70145] [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: 06/11/2024] [Revised: 10/07/2024] [Accepted: 11/07/2024] [Indexed: 02/05/2025]
Abstract
Viral accessory proteins play critical roles in viral escape from host innate immune responses and in viral inflammatory pathogenesis. Here we show that the SARS-CoV-2 accessory protein, ORF9b, but not other SARS-CoV-2 accessory proteins (ORF3a, ORF3b, ORF6, ORF7, ORF8, ORF9c, and ORF10), strongly activates inflammasome-dependent caspase-1 in A549 lung carcinoma cells and THP-1 monocyte-macrophage cells. Exposure to lipopolysaccharide (LPS) and ATP additively enhanced the activation of caspase-1 by ORF9b, suggesting that ORF9b and LPS follow parallel pathways in the activation of the inflammasome and caspase-1. Following rational in silico approaches, we have designed small molecules capable of inhibiting the homodimerization of ORF9b, which experimentally inhibited ORF9b-ORF9b homotypic interactions, caused mitochondrial eviction of ORF9b, inhibited ORF9b-induced activation of caspase-1 in A549 and THP-1 cells, cytokine release in THP-1 cells, and restored type I interferon (IFN-I) signaling suppressed by ORF9b in both cell models. These small molecules are first-in-class compounds targeting a viral accessory protein critical for viral-induced exacerbated inflammation and escape from innate immune responses, with the potential of mitigating the severe immunopathogenic damage induced by highly pathogenic coronaviruses and restoring antiviral innate immune responses curtailed by viral infection.
Collapse
Grants
- This work was funded by the Spanish National Research Council (CSIC, project numbers CSIC-COV19-006, CSIC-COV-19-201, CSIC-COV-19-117, SGL2103019, SGL2103015, 202020E079 and 202320E187 and LINCGLOBAL INCGL20009), the Catalan Agency for Management of University and Research Grants (AGAUR, 2020PANDE00048, 2021SGR1490, 2021SGR00350), the Spanish Ministry of Science (PID2021-123399OB-I00), the CSIC's Global Health Platform (PTI Salud Global), The Networked Center for Biomedical Research in Liver and Digestive Diseases (CIBER-EHD), the Spanish Structures and Excellence María de Maeztu program (CEX2021-001202-M), the European Commission-Next Generation EU (Regulation EU 2020/2094), and INDICASAT-AIP.
Collapse
Affiliation(s)
- Erika Zodda
- Laboratory of Cell Signaling and Cancer, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
| | - Mònica Pons
- Laboratory of Cell Signaling and Cancer, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
| | - Natàlia DeMoya-Valenzuela
- Department of Materials Science and Physical Chemistry, University of Barcelona, Barcelona, Spain
- Theoretical and Computational Chemistry Research Institute (IQTCUB), Barcelona, Spain
| | - Cristina Calvo-González
- Laboratory of Cell Signaling and Cancer, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
| | - Cristina Benítez-Rodríguez
- Laboratory of Cell Signaling and Cancer, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
| | - Blanca D López-Ayllón
- Viral immunology Lab, Molecular Biomedicine Department, Margarita Salas Center for Biological Research (CIB-CSIC), Madrid, Spain
| | - Achraf Hibot
- Laboratory of Pharmaceutical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Alice Zuin
- Regulation of the Proteasome Laboratory, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
| | - Bernat Crosas
- Regulation of the Proteasome Laboratory, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Barcelona, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBER-EHD), Madrid, Spain
| | - María Montoya
- Viral immunology Lab, Molecular Biomedicine Department, Margarita Salas Center for Biological Research (CIB-CSIC), Madrid, Spain
| | - María D Pujol
- Laboratory of Pharmaceutical Chemistry, School of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Jaime Rubio-Martínez
- Department of Materials Science and Physical Chemistry, University of Barcelona, Barcelona, Spain
- Theoretical and Computational Chemistry Research Institute (IQTCUB), Barcelona, Spain
| | - Timothy M Thomson
- Laboratory of Cell Signaling and Cancer, Barcelona Institute for Molecular Biology, Spanish National Scientific Research Council (IBMB-CSIC), Barcelona, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBER-EHD), Madrid, Spain
- High-Altitude Research Institute (IIA), Universidad Peruana Cayetano Heredia, Lima, Peru
- Instituto de Investigaciones Científicas y Servicio de Alta Tecnología (INDICASAT AIP), Panama City, Panama
| |
Collapse
|
10
|
Albalawi W, Thomas J, Mughal F, Kotsiri A, Roper KJ, Alshehri A, Kelbrick M, Pollakis G, Paxton WA. SARS-CoV-2 S, M, and E Structural Glycoproteins Differentially Modulate Endoplasmic Reticulum Stress Responses. Int J Mol Sci 2025; 26:1047. [PMID: 39940816 PMCID: PMC11816748 DOI: 10.3390/ijms26031047] [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: 12/19/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
We have previously shown that the hepatitis C virus (HCV) E1E2 envelope glycoprotein can regulate HIV-1 long-terminal repeat (LTR) activity through disruption to NF-κB activation. This response is associated with upregulation of the endoplasmic reticulum (ER) stress response pathway. Here, we demonstrate that the SARS-CoV-2 S, M, and E but not the N structural protein can perform similar downmodulation of HIV-1 LTR activation, and in a dose-dependent manner, in both HEK293 and lung BEAS-2B cell lines. This effect is highest with the SARS-CoV-2 Wuhan S strain and decreases over time for the subsequent emerging variants of concern (VOC), with Omicron providing the weakest effect. We developed pseudo-typed viral particle (PVP) viral tools that allowed for the generation of cell lines constitutively expressing the four SARS-CoV-2 structural proteins and utilising the VSV-g envelope protein to deliver the integrated gene construct. Differential gene expression analysis (DGEA) was performed on cells expressing S, E, M, or N to determine cell activation status. Gene expression differences were found in a number of interferon-stimulated genes (ISGs), including IF16, IFIT1, IFIT2, and ISG15, as well as for a number of heat shock protein (HSP) genes, including HSPH1, HSPA6, and HSPBP1, with all four SARS-CoV-2 structural proteins. There were also differences observed in expression patterns of transcription factors, with both SP1 and MAVS upregulated in the presence of S, M, and E but not the N protein. Collectively, the results indicate that gene expression patterns associated with ER stress pathways can be activated by SARS-CoV-2 envelope glycoprotein expression. The results suggest the SARS-CoV-2 infection can modulate an array of cell pathways, resulting in disruption to NF-κB signalling, hence providing alterations to multiple physiological responses of SARS-CoV-2-infected cells.
Collapse
Affiliation(s)
- Wejdan Albalawi
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool L69 7BE, UK; (W.A.); (J.T.); (F.M.); (A.K.); (K.J.R.); (A.A.); (M.K.)
- Department Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Aljouf, Sakakah 72388, Saudi Arabia
| | - Jordan Thomas
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool L69 7BE, UK; (W.A.); (J.T.); (F.M.); (A.K.); (K.J.R.); (A.A.); (M.K.)
| | - Farah Mughal
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool L69 7BE, UK; (W.A.); (J.T.); (F.M.); (A.K.); (K.J.R.); (A.A.); (M.K.)
| | - Aurelia Kotsiri
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool L69 7BE, UK; (W.A.); (J.T.); (F.M.); (A.K.); (K.J.R.); (A.A.); (M.K.)
| | - Kelly J. Roper
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool L69 7BE, UK; (W.A.); (J.T.); (F.M.); (A.K.); (K.J.R.); (A.A.); (M.K.)
- Virology Department, Animal and Plant Health Agency (APHA-Weybridge), Addlestone KT15 3NB, UK
| | - Abdullateef Alshehri
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool L69 7BE, UK; (W.A.); (J.T.); (F.M.); (A.K.); (K.J.R.); (A.A.); (M.K.)
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, P.O. Box 1988, Najran 61441, Saudi Arabia
| | - Matthew Kelbrick
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool L69 7BE, UK; (W.A.); (J.T.); (F.M.); (A.K.); (K.J.R.); (A.A.); (M.K.)
| | - Georgios Pollakis
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool L69 7BE, UK; (W.A.); (J.T.); (F.M.); (A.K.); (K.J.R.); (A.A.); (M.K.)
| | - William A. Paxton
- Department of Clinical Infection, Microbiology and Immunology (CIMI), Institute of Infection, Veterinary and Ecological Sciences (IVES), University of Liverpool, Liverpool L69 7BE, UK; (W.A.); (J.T.); (F.M.); (A.K.); (K.J.R.); (A.A.); (M.K.)
| |
Collapse
|
11
|
Wu Y, He L, Li R, Li J, Zhao Q, Shao B. A20 as a Potential Therapeutic Target for COVID-19. Immun Inflamm Dis 2025; 13:e70127. [PMID: 39853876 PMCID: PMC11760982 DOI: 10.1002/iid3.70127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 11/29/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major concern due to its astonishing prevalence and high fatality rate, especially among elderly people. Patients suffering from COVID-19 may exhibit immunosuppression in the initial stage of infection, while a cytokine storm can occur when the disease progresses to a severe stage. This inopportune immune rhythm not only makes patients more susceptible to the virus but also leads to numerous complications resulting from the excessive production of inflammatory factors. A20, which is widely accepted as a pivotal regulator of inflammation, has been shown to be implicated in the processes of antiviral responses and immunosuppression. Thus, A20 may participate in regulating the pathological processes of COVID-19. METHODS This narrative literature review summarizes recent evidence on the mechanisms of A20 in regulating the pathological processes of COVID-19. We also downloaded single-cell RNA-seq data sets from healthy individuals and patients with varying severities of COVID-19 from the NCBI GEO database to further dissect A20's regulatory mechanisms of these intricate cytokine pathways that are closely associated with SARS-CoV-2 infection. RESULTS A20 might be one of the most critical anti-infectious and anti-inflammatory factors involved in the pathogenesis of COVID-19. It effectively suppresses the immune damage and inflammatory storm caused by viral infection. CONCLUSIONS Understanding the relationship between A20-regulated signaling pathways and pathological processes of COVID-19 can provide insight into potential targets for intervention. Precise regulation of A20 to induce antiviral activity and an anti-inflammatory response could mediate the pathogenesis of COVID-19 and could become an effective treatment.
Collapse
Affiliation(s)
- Yongyao Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Lilan He
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Rong Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Jiuxuan Li
- Laboratory of Radiation Biology, Laboratory Medicine Centre, Department of Blood TransfusionThe Second Affiliated HospitalArmy Military Medical UniversityChongqingChina
| | - Qing Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Bin Shao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
12
|
Sugiura Y, Shimizu K, Takahashi T, Ueno S, Tanigou H, Amarbayasgalan S, Kamitani W. Amino acid T25 in the substrate-binding domain of SARS-CoV-2 nsp5 is involved in viral replication in the mouse lung. PLoS One 2024; 19:e0312800. [PMID: 39642113 PMCID: PMC11623800 DOI: 10.1371/journal.pone.0312800] [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: 07/01/2024] [Accepted: 10/15/2024] [Indexed: 12/08/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) non-structural protein 5 (nsp5) is a cysteine protease involved in viral replication and suppression of the host immune system. The substrate-binding domain of nsp5 is important for its protease activity. However, the relationship between nsp5 protease activity and viral replication remains unclear. We confirmed the importance of amino acid T25 in the nsp5 substrate-binding domain for viral replication using a split luciferase assay. By generating recombinant viruses using bacterial artificial chromosomes, we found that the proliferation of viruses with the T25I mutation in nsp5 was cell-dependent in culture. Furthermore, mice infected with the T25I mutant recombinant virus with a mouse acclimation backbone showed weight loss and increased lung viral load, similar to the wild-type (WT) infected group, up to 3 days after infection. However, after day 4, the lung viral load was significantly reduced in the T25I-infected group compared to that in the WT-infected group. This suggests that nsp5 T25 is involved in the pathogenesis of SARS-CoV-2.
Collapse
Affiliation(s)
- Yoshiro Sugiura
- Department of Infectious Disease and Host Defense, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Kenta Shimizu
- Department of Infectious Disease and Host Defense, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Tatsuki Takahashi
- Department of Infectious Disease and Host Defense, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Shiori Ueno
- Department of Infectious Disease and Host Defense, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Haruka Tanigou
- Department of Infectious Disease and Host Defense, Graduate School of Medicine, Gunma University, Gunma, Japan
| | | | - Wataru Kamitani
- Department of Infectious Disease and Host Defense, Graduate School of Medicine, Gunma University, Gunma, Japan
| |
Collapse
|
13
|
Zoler E, Meyer T, Bellón JS, Mönnig M, Sun B, Piehler J, Schreiber G. Promiscuous Janus kinase binding to cytokine receptors modulates signaling efficiencies and contributes to cytokine pleiotropy. Sci Signal 2024; 17:eadl1892. [PMID: 39561221 DOI: 10.1126/scisignal.adl1892] [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: 10/04/2023] [Revised: 04/09/2024] [Accepted: 10/08/2024] [Indexed: 11/21/2024]
Abstract
Janus kinases (JAKs) bind to class I and II cytokine receptors, activating signaling and regulating gene transcription through signal transducer and activator of transcription (STAT) proteins. Type I interferons (IFNs) require the JAK members TYK2 and JAK1, which bind to the receptor subunits IFNAR1 and IFNAR2, respectively. We investigated the role of JAKs in regulating IFNAR signaling activity. Synthetic IFNARs in which the extracellular domains of IFNAR1 and IFNAR2 are replaced with nanobodies had near-native type I IFN signaling, whereas the homomeric variant of IFNAR2 initiated much weaker signaling, despite harboring docking sites for JAKs and STATs. Cells with JAK1 and TYK2 knockout (KO) showed residual signaling, suggesting partial complementation by the remaining JAKs, particularly when they were overexpressed. Live-cell micropatterning experiments confirmed the promiscuous binding of JAK1, JAK2, and TYK2 to IFNAR1 and IFNAR2, and their recruitment correlated with their relative cellular abundances. However, each JAK had a different efficacy in inducing cross-phosphorylation and downstream signaling. JAK binding was also promiscuous for other cytokine receptors, including IFN-L1, IL-10Rβ, TPOR, and GHR, but not for EPOR, which activated different downstream signaling pathways. These findings suggest that competitive binding of JAKs to cytokine receptors together with the varying absolute and relative abundances of the JAKs in different cell types can account for the cell type-dependent signaling pleiotropy of cytokine receptors.
Collapse
Affiliation(s)
- Eyal Zoler
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Thomas Meyer
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Junel Sotolongo Bellón
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Mia Mönnig
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Boyue Sun
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Jacob Piehler
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Gideon Schreiber
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
14
|
Nguyen MH, Palfy G, Fogeron ML, Ninot Pedrosa M, Zehnder J, Rimal V, Callon M, Lecoq L, Barnes A, Meier BH, Böckmann A. Analysis of the structure and interactions of the SARS-CoV-2 ORF7b accessory protein. Proc Natl Acad Sci U S A 2024; 121:e2407731121. [PMID: 39508769 PMCID: PMC11573672 DOI: 10.1073/pnas.2407731121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/03/2024] [Indexed: 11/15/2024] Open
Abstract
SARS-CoV-2 carries a sizeable number of proteins that are accessory to replication but may be essential for virus-host interactions and modulation of the host immune response. Here, we investigated the structure and interactions of the largely unknown ORF7b, a small membranous accessory membrane protein of SARS-CoV-2. We show that structural predictions indicate a transmembrane (TM) leucine zipper for ORF7b, and experimentally confirm the predominantly α-helical secondary structure within a phospholipid membrane mimetic by solid-state NMR. We also show that ORF7b forms heterogeneous higher-order multimers. We determined ORF7b interactions with cellular TM leucine zipper proteins using both biochemical and NMR approaches, providing evidence for ORF7b interaction with the TM domains of E-cadherin, as well as phospholamban. Our results place ORF7b as a hypothetical interferer in cellular processes that utilize leucine zipper motifs in transmembrane multimerization domains.
Collapse
Affiliation(s)
- Minh-Ha Nguyen
- Molecular Microbiology and Structural Biochemistry, Unité Mixte de Recherche 5086 CNRS/Université de Lyon, 69367 Lyon, France
| | - Gyula Palfy
- Department of Chemistry and Applied Biosciences, Institute of Molecular Physical Sciences, Eidgenössische Technische Hochschule Zurich, 8093 Zurich, Switzerland
| | - Marie-Laure Fogeron
- Molecular Microbiology and Structural Biochemistry, Unité Mixte de Recherche 5086 CNRS/Université de Lyon, 69367 Lyon, France
| | - Martí Ninot Pedrosa
- Molecular Microbiology and Structural Biochemistry, Unité Mixte de Recherche 5086 CNRS/Université de Lyon, 69367 Lyon, France
| | - Johannes Zehnder
- Department of Chemistry and Applied Biosciences, Institute of Molecular Physical Sciences, Eidgenössische Technische Hochschule Zurich, 8093 Zurich, Switzerland
| | - Vaclav Rimal
- Department of Chemistry and Applied Biosciences, Institute of Molecular Physical Sciences, Eidgenössische Technische Hochschule Zurich, 8093 Zurich, Switzerland
| | - Morgane Callon
- Molecular Microbiology and Structural Biochemistry, Unité Mixte de Recherche 5086 CNRS/Université de Lyon, 69367 Lyon, France
- Department of Chemistry and Applied Biosciences, Institute of Molecular Physical Sciences, Eidgenössische Technische Hochschule Zurich, 8093 Zurich, Switzerland
| | - Lauriane Lecoq
- Molecular Microbiology and Structural Biochemistry, Unité Mixte de Recherche 5086 CNRS/Université de Lyon, 69367 Lyon, France
| | - Alexander Barnes
- Department of Chemistry and Applied Biosciences, Institute of Molecular Physical Sciences, Eidgenössische Technische Hochschule Zurich, 8093 Zurich, Switzerland
| | - Beat H Meier
- Department of Chemistry and Applied Biosciences, Institute of Molecular Physical Sciences, Eidgenössische Technische Hochschule Zurich, 8093 Zurich, Switzerland
| | - Anja Böckmann
- Molecular Microbiology and Structural Biochemistry, Unité Mixte de Recherche 5086 CNRS/Université de Lyon, 69367 Lyon, France
| |
Collapse
|
15
|
Sanislav O, Tetaj R, Metali, Ratcliffe J, Phillips W, Klein AR, Sethi A, Zhou J, Mezzenga R, Saxer SS, Charnley M, Annesley SJ, Reynolds NP. Cell invasive amyloid assemblies from SARS-CoV-2 peptides can form multiple polymorphs with varying neurotoxicity. NANOSCALE 2024; 16:19814-19827. [PMID: 39363846 DOI: 10.1039/d4nr03030c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
The neurological symptoms of COVID-19, often referred to as neuro-COVID include neurological pain, memory loss, cognitive and sensory disruption. These neurological symptoms can persist for months and are known as Post-Acute Sequalae of COVID-19 (PASC). The molecular origins of neuro-COVID, and how it contributes to PASC are unknown, however a growing body of research highlights that the self-assembly of protein fragments from SARS-CoV-2 into amyloid nanofibrils may play a causative role. Previously, we identified two fragments from the SARS-CoV-2 proteins, Open Reading Frame (ORF) 6 and ORF10, that self-assemble into neurotoxic amyloid assemblies. Here we further our understanding of the self-assembly mechanisms and nano-architectures formed by these fragments and their biological responses. By solubilising the peptides in a fluorinated solvent, we eliminate insoluble aggregates in the starting materials (seeds) that change the polymorphic landscape of the assemblies. The resultant assemblies are dominated by structures with higher free energies (e.g. ribbons and amorphous aggregates) that are less toxic to cultured neurons but do affect their mitochondrial respiration. We also show the first direct evidence of cellular uptake of viral amyloids. This work highlights the importance of understanding the polymorphic behaviour of amyloids and the correlation to neurotoxicity, particularly in the context of neuro-COVID and PASC.
Collapse
Affiliation(s)
- Oana Sanislav
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Rina Tetaj
- Institute for Chemistry and Bioanalytics, School of Life Sciences, FHNW, Muttenz, 4132, Switzerland
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, Victoria 3086, Australia.
| | - Metali
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, Victoria 3086, Australia.
| | - Julian Ratcliffe
- Bio Imaging Platform, La Trobe University, Melbourne, Victoria 3086, Australia
| | - William Phillips
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, Victoria 3086, Australia.
| | - Annaleise R Klein
- Australian Nuclear Science and Technology Organisation (ANSTO), Australian Synchrotron, Clayton, Victoria 3168, Australia
| | - Ashish Sethi
- Australian Nuclear Science and Technology Organisation (ANSTO), Australian Synchrotron, Clayton, Victoria 3168, Australia
| | - Jiangtao Zhou
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, LFO, E23, 8092, Zurich, Switzerland
| | - Raffaele Mezzenga
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, LFO, E23, 8092, Zurich, Switzerland
- Department of Materials, ETH Zurich, Zurich, 8093, Switzerland
| | - Sina S Saxer
- Institute for Chemistry and Bioanalytics, School of Life Sciences, FHNW, Muttenz, 4132, Switzerland
| | - Mirren Charnley
- Optical Sciences Centre, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Parkville, Victoria 3000, Australia
| | - Sarah J Annesley
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Nicholas P Reynolds
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, Victoria 3086, Australia.
- The Biomedical and Environmental Sensor Technology (BEST) Research Centre, Biosensors Program, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria 3086, Australia
| |
Collapse
|
16
|
Wickenhagen A, van Tol S, Munster V. Molecular determinants of cross-species transmission in emerging viral infections. Microbiol Mol Biol Rev 2024; 88:e0000123. [PMID: 38912755 PMCID: PMC11426021 DOI: 10.1128/mmbr.00001-23] [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] [Indexed: 06/25/2024] Open
Abstract
SUMMARYSeveral examples of high-impact cross-species transmission of newly emerging or re-emerging bat-borne viruses, such as Sudan virus, Nipah virus, and severe acute respiratory syndrome coronavirus 2, have occurred in the past decades. Recent advancements in next-generation sequencing have strengthened ongoing efforts to catalog the global virome, in particular from the multitude of different bat species. However, functional characterization of these novel viruses and virus sequences is typically limited with regard to assessment of their cross-species potential. Our understanding of the intricate interplay between virus and host underlying successful cross-species transmission has focused on the basic mechanisms of entry and replication, as well as the importance of host innate immune responses. In this review, we discuss the various roles of the respective molecular mechanisms underlying cross-species transmission using different recent bat-borne viruses as examples. To delineate the crucial cellular and molecular steps underlying cross-species transmission, we propose a framework of overall characterization to improve our capacity to characterize viruses as benign, of interest, or of concern.
Collapse
Affiliation(s)
- Arthur Wickenhagen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Sarah van Tol
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Vincent Munster
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| |
Collapse
|
17
|
Ivanov KI, Yang H, Sun R, Li C, Guo D. The emerging role of SARS-CoV-2 nonstructural protein 1 (nsp1) in epigenetic regulation of host gene expression. FEMS Microbiol Rev 2024; 48:fuae023. [PMID: 39231808 PMCID: PMC11418652 DOI: 10.1093/femsre/fuae023] [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: 06/19/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/06/2024] Open
Abstract
Infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes widespread changes in epigenetic modifications and chromatin architecture in the host cell. Recent evidence suggests that SARS-CoV-2 nonstructural protein 1 (nsp1) plays an important role in driving these changes. Previously thought to be primarily involved in host translation shutoff and cellular mRNA degradation, nsp1 has now been shown to be a truly multifunctional protein that affects host gene expression at multiple levels. The functions of nsp1 are surprisingly diverse and include not only the downregulation of cellular mRNA translation and stability, but also the inhibition of mRNA export from the nucleus, the suppression of host immune signaling, and, most recently, the epigenetic regulation of host gene expression. In this review, we first summarize the current knowledge on SARS-CoV-2-induced changes in epigenetic modifications and chromatin structure. We then focus on the role of nsp1 in epigenetic reprogramming, with a particular emphasis on the silencing of immune-related genes. Finally, we discuss potential molecular mechanisms underlying the epigenetic functions of nsp1 based on evidence from SARS-CoV-2 interactome studies.
Collapse
Affiliation(s)
- Konstantin I Ivanov
- Guangzhou National Laboratory, Guangzhou, 510320, China
- Department of Microbiology, University of Helsinki, Helsinki, 00014, Finland
| | - Haibin Yang
- MOE Key Laboratory of Tropical Disease Control, Center for Infection and Immunity Studies (CIIS), School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Ruixue Sun
- Guangzhou National Laboratory, Guangzhou, 510320, China
| | - Chunmei Li
- MOE Key Laboratory of Tropical Disease Control, Center for Infection and Immunity Studies (CIIS), School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Deyin Guo
- Guangzhou National Laboratory, Guangzhou, 510320, China
- MOE Key Laboratory of Tropical Disease Control, Center for Infection and Immunity Studies (CIIS), School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, 518107, China
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, China
| |
Collapse
|
18
|
Puray-Chavez M, Eschbach JE, Xia M, LaPak KM, Zhou Q, Jasuja R, Pan J, Xu J, Zhou Z, Mohammed S, Wang Q, Lawson DQ, Djokic S, Hou G, Ding S, Brody SL, Major MB, Goldfarb D, Kutluay SB. A basally active cGAS-STING pathway limits SARS-CoV-2 replication in a subset of ACE2 positive airway cell models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.07.574522. [PMID: 38260460 PMCID: PMC10802478 DOI: 10.1101/2024.01.07.574522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Host factors that define the cellular tropism of SARS-CoV-2 beyond the cognate ACE2 receptor are poorly defined. Here we report that SARS-CoV-2 replication is restricted at a post-entry step in a number of ACE2-positive airway-derived cell lines due to tonic activation of the cGAS-STING pathway mediated by mitochondrial DNA leakage and naturally occurring cGAS and STING variants. Genetic and pharmacological inhibition of the cGAS-STING and type I/III IFN pathways as well as ACE2 overexpression overcome these blocks. SARS-CoV-2 replication in STING knockout cell lines and primary airway cultures induces ISG expression but only in uninfected bystander cells, demonstrating efficient antagonism of the type I/III IFN-pathway in productively infected cells. Pharmacological inhibition of STING in primary airway cells enhances SARS-CoV-2 replication and reduces virus-induced innate immune activation. Together, our study highlights that tonic activation of the cGAS-STING and IFN pathways can impact SARS-CoV-2 cellular tropism in a manner dependent on ACE2 expression levels.
Collapse
Affiliation(s)
- Maritza Puray-Chavez
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jenna E. Eschbach
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ming Xia
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kyle M. LaPak
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Qianzi Zhou
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ria Jasuja
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jiehong Pan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jian Xu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Zixiang Zhou
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shawn Mohammed
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Qibo Wang
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Dana Q. Lawson
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sanja Djokic
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gaopeng Hou
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Steven L. Brody
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael B. Major
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, USA
| | - Dennis Goldfarb
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- Institute for Informatics, Data Science & Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Sebla B. Kutluay
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
19
|
Chiu HP, Yeo YY, Lai TY, Hung CT, Kowdle S, Haas GD, Jiang S, Sun W, Lee B. SARS-CoV-2 Nsp15 antagonizes the cGAS-STING-mediated antiviral innate immune responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611469. [PMID: 39282446 PMCID: PMC11398466 DOI: 10.1101/2024.09.05.611469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Coronavirus (CoV) Nsp15 is a viral endoribonuclease (EndoU) with a preference for uridine residues. CoV Nsp15 is an innate immune antagonist which prevents dsRNA sensor recognition and stress granule formation by targeting viral and host RNAs. SARS-CoV-2 restricts and delays the host antiviral innate immune responses through multiple viral proteins, but the role of SARS-CoV-2 Nsp15 in innate immune evasion is not completely understood. Here, we generate an EndoU activity knockout rSARS-CoV-2Nsp15-H234A to elucidate the biological functions of Nsp15. Relative to wild-type rSARS-CoV-2, replication of rSARS-CoV-2Nsp15-H234A was significantly decreased in IFN-responsive A549-ACE2 cells but not in its STAT1 knockout counterpart. Transcriptomic analysis revealed upregulation of innate immune response genes in cells infected with rSARS-CoV-2Nsp15-H234A relative to wild-type virus, including cGAS-STING, cytosolic DNA sensors activated by both DNA and RNA viruses. Treatment with STING inhibitors H-151 and SN-011 rescued the attenuated phenotype of rSARS-CoV-2Nsp15-H234A. SARS-CoV-2 Nsp15 inhibited cGAS-STING-mediated IFN-β promoter and NF-κB reporter activity, as well as facilitated the replication of EV-D68 and NDV by diminishing cGAS and STING expression and downstream innate immune responses. Notably, the decline in cGAS and STING was also apparent during SARS-CoV-2 infection. The EndoU activity was essential for SARS-CoV-2 Nsp15-mediated cGAS and STING downregulation, but not all HCoV Nsp15 share the consistent substrate selectivity. In the hamster model, rSARS-CoV-2Nsp15-H234A replicated to lower titers in the nasal turbinates and lungs and induced higher innate immune responses. Collectively, our findings exhibit that SARS-CoV-2 Nsp15 serves as a host innate immune antagonist by targeting host cGAS and STING.
Collapse
Affiliation(s)
- Hsin-Ping Chiu
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yao Yu Yeo
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Program in Virology, Division of Medical Sciences, Harvard Medical School, Boston, MA, United States
| | - Tsoi Ying Lai
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Chuan-Tien Hung
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Shreyas Kowdle
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Griffin D Haas
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sizun Jiang
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Program in Virology, Division of Medical Sciences, Harvard Medical School, Boston, MA, United States
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Department of Pathology, Dana Farber Cancer Institute, Boston, MA, United States
- Broad Institute of Harvard and MIT, Cambridge, MA, United States
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
20
|
Aktepe TE, Deerain JM, Hyde JL, Fritzlar S, Mead EM, Carrera Montoya J, Hachani A, Pearson JS, White PA, Mackenzie JM. Norovirus-mediated translation repression promotes macrophage cell death. PLoS Pathog 2024; 20:e1012480. [PMID: 39226332 PMCID: PMC11398682 DOI: 10.1371/journal.ppat.1012480] [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: 12/19/2023] [Revised: 09/13/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024] Open
Abstract
Norovirus infection is characterised by a rapid onset of disease and the development of debilitating symptoms including projectile vomiting and diffuse diarrhoea. Vaccines and antivirals are sorely lacking and developments in these areas are hampered by the lack of an adequate cell culture system to investigate human norovirus replication and pathogenesis. Herein, we describe how the model norovirus, Mouse norovirus (MNV), produces a viral protein, NS3, with the functional capacity to attenuate host protein translation which invokes the activation of cell death via apoptosis. We show that this function of NS3 is conserved between human and mouse viruses and map the protein domain attributable to this function. Our study highlights a critical viral protein that mediates crucial activities during replication, potentially identifying NS3 as a worthy target for antiviral drug development.
Collapse
Affiliation(s)
- Turgut E Aktepe
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Joshua M Deerain
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Jennifer L Hyde
- Department of Microbiology, School of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Svenja Fritzlar
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Eleanor M Mead
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Julio Carrera Montoya
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Abderrahman Hachani
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Jaclyn S Pearson
- The Hudson Institute of Medical Research, Centre for Innate Immunity and Infectious Diseases, Melbourne, Australia
| | - Peter A White
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Jason M Mackenzie
- Department of Microbiology and Immunology, at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| |
Collapse
|
21
|
Omler A, Mutso M, Vaher M, Freitas JR, Taylor A, David CT, Moseley GW, Liu X, Merits A, Mahalingam S. Exploring Barmah Forest virus pathogenesis: molecular tools to investigate non-structural protein 3 nuclear localization and viral genomic determinants of replication. mBio 2024; 15:e0099324. [PMID: 38953633 PMCID: PMC11323547 DOI: 10.1128/mbio.00993-24] [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: 04/13/2024] [Accepted: 05/03/2024] [Indexed: 07/04/2024] Open
Abstract
Barmah Forest virus (BFV) is a mosquito-borne virus that causes arthralgia with accompanying rash, fever, and myalgia in humans. The virus is mainly found in Australia and has caused outbreaks associated with significant health concerns. As the sole representative of the Barmah Forest complex within the genus Alphavirus, BFV is not closely related genetically to other alphaviruses. Notably, basic knowledge of BFV molecular virology has not been well studied due to a lack of critical investigative tools such as an infectious clone. Here we describe the construction of an infectious BFV cDNA clone based on Genbank sequence and demonstrate that the clone-derived virus has in vitro and in vivo properties similar to naturally occurring virus, BFV field isolate 2193 (BFV2193-FI). A substitution in nsP4, V1911D, which was identified in the Genbank reference sequence, was found to inhibit virus rescue and replication. T1325P substitution in nsP2 selected during virus passaging was shown to be an adaptive mutation, compensating for the inhibitory effect of nsP4-V1911D. The two mutations were associated with changes in viral non-structural polyprotein processing and type I interferon (IFN) induction. Interestingly, a nuclear localization signal, active in mammalian but not mosquito cells, was identified in nsP3. A point mutation abolishing nsP3 nuclear localization attenuated BFV replication. This effect was more prominent in the presence of type I interferon signaling, suggesting nsP3 nuclear localization might be associated with IFN antagonism. Furthermore, abolishing nsP3 nuclear localization reduced virus replication in mice but did not significantly affect disease.IMPORTANCEBarmah Forest virus (BFV) is Australia's second most prevalent arbovirus, with approximately 1,000 cases reported annually. The clinical symptoms of BFV infection include rash, polyarthritis, arthralgia, and myalgia. As BFV is not closely related to other pathogenic alphaviruses or well-studied model viruses, our understanding of its molecular virology and mechanisms of pathogenesis is limited. There is also a lack of molecular tools essential for corresponding studies. Here we describe the construction of an infectious clone of BFV, variants harboring point mutations, and sequences encoding marker protein. In infected mammalian cells, nsP3 of BFV was located in the nuclei. This finding extends our understanding of the diverse mechanisms used by alphavirus replicase proteins to interact with host cells. Our novel observations highlight the complex synergy through which the viral replication machinery evolves to correct mutation errors within the viral genome.
Collapse
Affiliation(s)
- Ailar Omler
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
- Institute of Bioengineering, University of Tartu, Tartu, Estonia
| | - Margit Mutso
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Mihkel Vaher
- The Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Joseph R. Freitas
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
- Global Virus Network (GVN) Centre for Excellence in Arboviruses, Griffith University, Gold Coast, Queensland, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Adam Taylor
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
- Global Virus Network (GVN) Centre for Excellence in Arboviruses, Griffith University, Gold Coast, Queensland, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Cassandra T. David
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Gregory W. Moseley
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Xiang Liu
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
- Global Virus Network (GVN) Centre for Excellence in Arboviruses, Griffith University, Gold Coast, Queensland, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Andres Merits
- Institute of Bioengineering, University of Tartu, Tartu, Estonia
| | - Suresh Mahalingam
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
- Global Virus Network (GVN) Centre for Excellence in Arboviruses, Griffith University, Gold Coast, Queensland, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
22
|
Gonzalez-Orozco M, Rodriguez-Salazar CA, Giraldo MI. The Dual Role of TRIM7 in Viral Infections. Viruses 2024; 16:1285. [PMID: 39205259 PMCID: PMC11360163 DOI: 10.3390/v16081285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
The E3 ubiquitin ligase TRIM7 is known to have dual roles during viral infections. Like other TRIM proteins, TRIM7 can regulate the IFN pathway via the regulation of the cytosolic receptors RIG-I or MDA-5, which promote the production of type I interferons (IFN-I) and antiviral immune responses. Alternatively, under certain infectious conditions, TRIM7 can negatively regulate IFN-I signaling, resulting in increased virus replication. A growing body of evidence has also shown that TRIM7 can, in some cases, ubiquitinate viral proteins to promote viral replication and pathogenesis, while in other cases it can promote degradation of viral proteins through the proteasome, reducing virus infection. TRIM7 can also regulate the host inflammatory response and modulate the production of inflammatory cytokines, which can lead to detrimental inflammation. TRIM7 can also protect the host during infection by reducing cellular apoptosis. Here, we discuss the multiple functions of TRIM7 during viral infections and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Maria Gonzalez-Orozco
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (M.G.-O.); (C.A.R.-S.)
| | - Carlos A. Rodriguez-Salazar
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (M.G.-O.); (C.A.R.-S.)
- Molecular Biology and Virology Laboratory, Faculty of Medicine and Health Sciences, Corporación Universitaria Empresarial Alexander von Humboldt, Armenia 630003, Colombia
| | - Maria I. Giraldo
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (M.G.-O.); (C.A.R.-S.)
| |
Collapse
|
23
|
Magnen M, You R, Rao AA, Davis RT, Rodriguez L, Bernard O, Simoneau CR, Hysenaj L, Hu KH, Maishan M, Conrad C, Gbenedio OM, Samad B, Consortium TUCSFCOMET, Love C, Woodruff PG, Erle DJ, Hendrickson CM, Calfee CS, Matthay MA, Roose JP, Sil A, Ott M, Langelier CR, Krummel MF, Looney MR. Immediate myeloid depot for SARS-CoV-2 in the human lung. SCIENCE ADVANCES 2024; 10:eadm8836. [PMID: 39083602 PMCID: PMC11290487 DOI: 10.1126/sciadv.adm8836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 05/20/2024] [Indexed: 08/02/2024]
Abstract
In the pathogenesis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, epithelial populations in the distal lung expressing Angiotensin-converting enzyme 2 (ACE2) are infrequent, and therefore, the model of viral expansion and immune cell engagement remains incompletely understood. Using human lungs to investigate early host-viral pathogenesis, we found that SARS-CoV-2 had a rapid and specific tropism for myeloid populations. Human alveolar macrophages (AMs) reliably expressed ACE2 allowing both spike-ACE2-dependent viral entry and infection. In contrast to Influenza A virus, SARS-CoV-2 infection of AMs was productive, amplifying viral titers. While AMs generated new viruses, the interferon responses to SARS-CoV-2 were muted, hiding the viral dissemination from specific antiviral immune responses. The reliable and veiled viral depot in myeloid cells in the very early phases of SARS-CoV-2 infection of human lungs enables viral expansion in the distal lung and potentially licenses subsequent immune pathologies.
Collapse
Affiliation(s)
- Mélia Magnen
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ran You
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arjun A. Rao
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
- CoLabs Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ryan T. Davis
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lauren Rodriguez
- CoLabs Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Olivier Bernard
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Camille R. Simoneau
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Lisiena Hysenaj
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kenneth H. Hu
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mazharul Maishan
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Catharina Conrad
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Oghenekevwe M. Gbenedio
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bushra Samad
- CoLabs Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - The UCSF COMET Consortium
- All UCSF COMET Consortium collaborators are affiliated with the University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christina Love
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Prescott G. Woodruff
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David J. Erle
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Carolyn M. Hendrickson
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Carolyn S. Calfee
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael A. Matthay
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeroen P. Roose
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Anita Sil
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Melanie Ott
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Gladstone Institutes, San Francisco, CA 94158, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Charles R. Langelier
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Matthew F. Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
24
|
Leekha A, Saeedi A, Kumar M, Sefat KMSR, Martinez-Paniagua M, Meng H, Fathi M, Kulkarni R, Reichel K, Biswas S, Tsitoura D, Liu X, Cooper LJN, Sands CM, Das VE, Sebastian M, Hurst BL, Varadarajan N. An intranasal nanoparticle STING agonist protects against respiratory viruses in animal models. Nat Commun 2024; 15:6053. [PMID: 39025863 PMCID: PMC11258242 DOI: 10.1038/s41467-024-50234-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 07/04/2024] [Indexed: 07/20/2024] Open
Abstract
Respiratory viral infections cause morbidity and mortality worldwide. Despite the success of vaccines, vaccination efficacy is weakened by the rapid emergence of viral variants with immunoevasive properties. The development of an off-the-shelf, effective, and safe therapy against respiratory viral infections is thus desirable. Here, we develop NanoSTING, a nanoparticle formulation of the endogenous STING agonist, 2'-3' cGAMP, to function as an immune activator and demonstrate its safety in mice and rats. A single intranasal dose of NanoSTING protects against pathogenic strains of SARS-CoV-2 (alpha and delta VOC) in hamsters. In transmission experiments, NanoSTING reduces the transmission of SARS-CoV-2 Omicron VOC to naïve hamsters. NanoSTING also protects against oseltamivir-sensitive and oseltamivir-resistant strains of influenza in mice. Mechanistically, NanoSTING upregulates locoregional interferon-dependent and interferon-independent pathways in mice, hamsters, as well as non-human primates. Our results thus implicate NanoSTING as a broad-spectrum immune activator for controlling respiratory virus infection.
Collapse
Affiliation(s)
- Ankita Leekha
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Arash Saeedi
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Monish Kumar
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - K M Samiur Rahman Sefat
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Melisa Martinez-Paniagua
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Hui Meng
- College of Optometry, University of Houston, Houston, TX, USA
| | - Mohsen Fathi
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Rohan Kulkarni
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Kate Reichel
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Sujit Biswas
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | | | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA
| | | | | | - Vallabh E Das
- College of Optometry, University of Houston, Houston, TX, USA
| | | | - Brett L Hurst
- Institute for Antiviral Research, Utah State University, Logan, UT, USA
| | - Navin Varadarajan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA.
| |
Collapse
|
25
|
Singh J, Anantharaj A, Kumar P, Pandey R, Pandey AK, Medigeshi GR. The Effective Inhibitory Concentration of Interferon-β Correlates with Infectivity and Replication Fitness of SARS-CoV-2 Variants. J Interferon Cytokine Res 2024; 44:325-333. [PMID: 38557204 DOI: 10.1089/jir.2024.0016] [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: 04/04/2024] Open
Abstract
India saw a spike in COVID-19 cases in early 2023, and this wave of infection was attributed to XBB sublineages of SARS-CoV-2 Omicron variant. The impact of XBB wave was significantly shorter with low burden of severe cases or hospitalization as compared with previous SARS-CoV-2 variants of concern. Although a combination of old and new mutations in the spike region of XBB.1.16 variant led to a drastic reduction in the ability of antibodies from prior immunity to neutralize this virus, additional nonspike mutations suggested a possible change in its ability to suppress innate immune responses. In this study, we tested the sensitivity of Delta, BA.2.75, and XBB.1.16 variants to interferon-β (IFN-β) treatment and found that XBB.1.16 variant was most sensitive to IFN-β. We next tested the ability of serum antibodies from healthy individuals to neutralize XBB.1.16. We showed that most of the individuals with hybrid immunity maintained a low but significant level of neutralizing antibodies to XBB.1.16 variant. Therefore, our observations indicated that both hybrid immunity because of natural infection and enhanced sensitivity to IFNs may have contributed to the low impact of XBB.1.16 infections in India.
Collapse
Affiliation(s)
- Janmejay Singh
- Bioassay Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Anbalagan Anantharaj
- Bioassay Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Parveen Kumar
- Bioassay Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Rajesh Pandey
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Anil Kumar Pandey
- Academic Block, Employees State Insurance Corporation Medical College and Hospital, Faridabad, Haryana, India
| | - Guruprasad R Medigeshi
- Bioassay Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Department of Biology, Indian Institute of Science Education and Research, Tirupati, Andhra Pradesh, India
| |
Collapse
|
26
|
Williams BD, Ferede D, Abdelaal HFM, Berube BJ, Podell BK, Larsen SE, Baldwin SL, Coler RN. Protective interplay: Mycobacterium tuberculosis diminishes SARS-CoV-2 severity through innate immune priming. Front Immunol 2024; 15:1424374. [PMID: 38966641 PMCID: PMC11222399 DOI: 10.3389/fimmu.2024.1424374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/06/2024] [Indexed: 07/06/2024] Open
Abstract
At the beginning of the COVID-19 pandemic those with underlying chronic lung conditions, including tuberculosis (TB), were hypothesized to be at higher risk of severe COVID-19 disease. However, there is inconclusive clinical and preclinical data to confirm the specific risk SARS-CoV-2 poses for the millions of individuals infected with Mycobacterium tuberculosis (M.tb). We and others have found that compared to singly infected mice, mice co-infected with M.tb and SARS-CoV-2 leads to reduced SARS-CoV-2 severity compared to mice infected with SARS-CoV-2 alone. Consequently, there is a large interest in identifying the molecular mechanisms responsible for the reduced SARS-CoV-2 infection severity observed in M.tb and SARS-CoV-2 co-infection. To address this, we conducted a comprehensive characterization of a co-infection model and performed mechanistic in vitro modeling to dynamically assess how the innate immune response induced by M.tb restricts viral replication. Our study has successfully identified several cytokines that induce the upregulation of anti-viral genes in lung epithelial cells, thereby providing protection prior to challenge with SARS-CoV-2. In conclusion, our study offers a comprehensive understanding of the key pathways induced by an existing bacterial infection that effectively restricts SARS-CoV-2 activity and identifies candidate therapeutic targets for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Brittany D. Williams
- Department of Global Health, University of Washington, Seattle, WA, United States
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle Children’s, Seattle, WA, United States
| | - Debora Ferede
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle Children’s, Seattle, WA, United States
| | - Hazem F. M. Abdelaal
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle Children’s, Seattle, WA, United States
| | - Bryan J. Berube
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle Children’s, Seattle, WA, United States
- HDT Bio Corp, Seattle, WA, United States
| | - Brendan K. Podell
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Sasha E. Larsen
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle Children’s, Seattle, WA, United States
| | - Susan L. Baldwin
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle Children’s, Seattle, WA, United States
| | - Rhea N. Coler
- Department of Global Health, University of Washington, Seattle, WA, United States
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle Children’s, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
27
|
Xiao X, Fu Y, You W, Huang C, Zeng F, Gu X, Sun X, Li J, Zhang Q, Du W, Cheng G, Liu Z, Liu L. Inhibition of the RLR signaling pathway by SARS-CoV-2 ORF7b is mediated by MAVS and abrogated by ORF7b-homologous interfering peptide. J Virol 2024; 98:e0157323. [PMID: 38572974 PMCID: PMC11092349 DOI: 10.1128/jvi.01573-23] [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: 10/09/2023] [Accepted: 03/15/2024] [Indexed: 04/05/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and characterized by dysregulated immune response. Studies have shown that the SARS-CoV-2 accessory protein ORF7b induces host cell apoptosis through the tumor necrosis factor alpha (TNF-α) pathway and blocks the production of interferon beta (IFN-β). The underlying mechanism remains to be investigated. In this study, we found that ORF7b facilitated viral infection and production, and inhibited the RIG-I-like receptor (RLR) signaling pathway through selectively interacting with mitochondrial antiviral-signaling protein (MAVS). MAVS439-466 region and MAVS Lys461 were essential for the physical association between MAVS and ORF7b, and the inhibition of the RLR signaling pathway by ORF7b. MAVSK461/K63 ubiquitination was essential for the RLR signaling regulated by the MAVS-ORF7b complex. ORF7b interfered with the recruitment of tumor necrosis factor receptor-related factor 6 (TRAF6) and the activation of the RLR signaling pathway by MAVS. Furthermore, interfering peptides targeting the ORF7b complex reversed the ORF7b-suppressed MAVS-RLR signaling pathway. The most potent interfering peptide V disrupts the formation of ORF7b tetramers, reverses the levels of the ORF7b-inhibited physical association between MAVS and TRAF6, leading to the suppression of viral growth and infection. Overall, this study provides a mechanism for the suppression of innate immunity by SARS-CoV-2 infection and the mechanism-based approach via interfering peptides to potentially prevent SARS-CoV-2 infection.IMPORTANCEThe pandemic coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and continues to be a threat to public health. It is imperative to understand the biology of SARS-CoV-2 infection and find approaches to prevent SARS-CoV-2 infection and ameliorate COVID-19. Multiple SARS-CoV-2 proteins are known to function on the innate immune response, but the underlying mechanism remains unknown. This study shows that ORF7b inhibits the RIG-I-like receptor (RLR) signaling pathway through the physical association between ORF7b and mitochondrial antiviral-signaling protein (MAVS), impairing the K63-linked MAVS polyubiquitination and its recruitment of tumor necrosis factor receptor-related factor 6 (TRAF6) to MAVS. The most potent interfering peptide V targeting the ORF7b-MAVS complex may reverse the suppression of the MAVS-mediated RLR signaling pathway by ORF7b and prevent viral infection and production. This study may provide new insights into the pathogenic mechanism of SARS-CoV-2 and a strategy to develop new drugs to prevent SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Institute of Virology, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Yanan Fu
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Institute of Virology, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Wanling You
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Institute of Virology, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Congcong Huang
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Institute of Virology, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Feng Zeng
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Institute of Virology, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Xinsheng Gu
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Xiaoguang Sun
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Institute of Virology, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Jian Li
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Institute of Virology, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Qiwei Zhang
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Weixing Du
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Zhixin Liu
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Institute of Virology, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Long Liu
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Institute of Virology, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
28
|
Viox EG, Bosinger SE, Douek DC, Schreiber G, Paiardini M. Harnessing the power of IFN for therapeutic approaches to COVID-19. J Virol 2024; 98:e0120423. [PMID: 38651899 PMCID: PMC11092331 DOI: 10.1128/jvi.01204-23] [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] [Indexed: 04/25/2024] Open
Abstract
Interferons (IFNs) are essential for defense against viral infections but also drive recruitment of inflammatory cells to sites of infection, a key feature of severe COVID-19. Here, we explore the complexity of the IFN response in COVID-19, examine the effects of manipulating IFN on SARS-CoV-2 viral replication and pathogenesis, and highlight pre-clinical and clinical studies evaluating the therapeutic efficacy of IFN in limiting COVID-19 severity.
Collapse
Affiliation(s)
- Elise G. Viox
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Steven E. Bosinger
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Emory NPRC Genomics Core Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Daniel C. Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Gideon Schreiber
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
29
|
Kong X, Wang Q, Wang X, Yang K, Nie S, Li Y, Lao W, Yu X, Zhang Y, Li Z, Liu Y, Ning J, Wang Y, Bi C, Wu C, Zhai A. LINC01002 functions as a ceRNA to regulate FRMD8 by sponging miR-4324 for the development of COVID-19. Virol J 2024; 21:109. [PMID: 38734674 PMCID: PMC11088083 DOI: 10.1186/s12985-024-02382-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 05/03/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Syndrome coronavirus-2 (SARS-CoV-2) has developed various strategies to evade the antiviral impact of type I IFN. Non-structural proteins and auxiliary proteins have been extensively researched on their role in immune escape. Nevertheless, the detailed mechanisms of structural protein-induced immune evasion have not been well elucidated. METHODS Human alveolar basal epithelial carcinoma cell line (A549) was stimulated with polyinosinic-polycytidylic acid (PIC) and independently transfected with four structural proteins expression plasmids, including nucleocapsid (N), spike (S), membrane (M) and envelope (E) proteins. By RT-qPCR and ELISA, the structural protein with the most pronounced inhibitory effects on IFN-β induction was screened. RNA-sequencing (RNA-Seq) and two differential analysis strategies were used to obtain differentially expressed genes associated with N protein inhibition of IFN-β induction. Based on DIANA-LncBase and StarBase databases, the interactive competitive endogenous RNA (ceRNA) network for N protein-associated genes was constructed. By combining single-cell sequencing data (GSE158055), lncRNA-miRNA-mRNA axis was further determined. Finally, RT-qPCR was utilized to illustrate the regulatory functions among components of the ceRNA axis. RESULTS SARS-CoV-2 N protein inhibited IFN-β induction in human alveolar epithelial cells most significantly compared with other structural proteins. RNA-Seq data analysis revealed genes related to N protein inhibiting IFNs induction. The obtained 858 differentially expressed genes formed the reliable ceRNA network. The function of LINC01002-miR-4324-FRMD8 axis in the IFN-dominated immune evasion was further demonstrated through integrating single-cell sequencing data. Moreover, we validated that N protein could reverse the effect of PIC on LINC01002, FRMD8 and miR-4324 expression, and subsequently on IFN-β expression level. And LINC01002 could regulate the production of FRMD8 by inhibiting miR-4324. CONCLUSION SARS-CoV-2 N protein suppressed the induction of IFN-β by regulating LINC01002 which was as a ceRNA, sponging miR-4324 and participating in the regulation of FRMD8 mRNA. Our discovery provides new insights into early intervention therapy and drug development on SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xinyi Kong
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Qinjin Wang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Xumeng Wang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
- Department of Microbiology, Harbin Medical University, Harbin, 150081, China
| | - Kaming Yang
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Shuping Nie
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Yuetong Li
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Wanwen Lao
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Xin Yu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Yanping Zhang
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Zhenlin Li
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Yang Liu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Jie Ning
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Yan Wang
- Department of Microbiology, Harbin Medical University, Harbin, 150081, China.
| | - Changlong Bi
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China.
| | - Chao Wu
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China.
| | - Aixia Zhai
- Department of Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China.
| |
Collapse
|
30
|
Gori Savellini G, Anichini G, Manetti F, Trivisani CI, Cusi MG. Deletion of 82-85 N-Terminal Residues in SARS-CoV-2 Nsp1 Restricts Virus Replication. Viruses 2024; 16:689. [PMID: 38793572 PMCID: PMC11125901 DOI: 10.3390/v16050689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Non-structural protein 1 (Nsp1) represents one of the most crucial SARS-CoV-2 virulence factors by inhibiting the translation of host mRNAs and promoting their degradation. We selected naturally occurring virus lineages with specific Nsp1 deletions located at both the N- and C-terminus of the protein. Our data provide new insights into how Nsp1 coordinates these functions on host and viral mRNA recognition. Residues 82-85 in the N-terminal part of Nsp1 likely play a role in docking the 40S mRNA entry channel, preserving the inhibition of host gene expression without affecting cellular mRNA decay. Furthermore, this domain prevents viral mRNAs containing the 5'-leader sequence to escape translational repression. These findings support the presence of distinct domains within the Nsp1 protein that differentially modulate mRNA recognition, translation and turnover. These insights have implications for the development of drugs targeting viral proteins and provides new evidences of how specific mutations in SARS-CoV-2 Nsp1 could attenuate the virus.
Collapse
Affiliation(s)
| | - Gabriele Anichini
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
| | - Fabrizio Manetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy (C.I.T.)
| | | | - Maria Grazia Cusi
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
| |
Collapse
|
31
|
Zhang Z. The Initial COVID-19 Reliable Interactive DNA Methylation Markers and Biological Implications. BIOLOGY 2024; 13:245. [PMID: 38666857 PMCID: PMC11048280 DOI: 10.3390/biology13040245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/22/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
Earlier research has established the existence of reliable interactive genomic biomarkers. However, reliable DNA methylation biomarkers, not to mention interactivity, have yet to be identified at the epigenetic level. This study, drawing from 865,859 methylation sites, discovered two miniature sets of Infinium MethylationEPIC sites, each having eight CpG sites (genes) to interact with each other and disease subtypes. They led to the nearly perfect (96.87-100% accuracy) prediction of COVID-19 patients from patients with other diseases or healthy controls. These CpG sites can jointly explain some post-COVID-19-related conditions. These CpG sites and the optimally performing genomic biomarkers reported in the literature become potential druggable targets. Among these CpG sites, cg16785077 (gene MX1), cg25932713 (gene PARP9), and cg22930808 (gene PARP9) at DNA methylation levels indicate that the initial SARS-CoV-2 virus may be better treated as a transcribed viral DNA into RNA virus, i.e., not as an RNA virus that has concerned scientists in the field. Such a discovery can significantly change the scientific thinking and knowledge of viruses.
Collapse
Affiliation(s)
- Zhengjun Zhang
- School of Computer, Data and Information Sciences, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
32
|
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
|
33
|
Scutari R, Fox V, Fini V, Granaglia A, Vittucci AC, Smarrazzo A, Lancella L, Calo' Carducci F, Romani L, Cursi L, Bernaschi P, Russo C, Campana A, Bernardi S, Villani A, Perno CF, Alteri C. Molecular characterization of SARS-CoV-2 Omicron clade and clinical presentation in children. Sci Rep 2024; 14:5325. [PMID: 38438451 PMCID: PMC10912656 DOI: 10.1038/s41598-024-55599-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024] Open
Abstract
Since its emergence, SARS-CoV-2 Omicron clade has shown a marked degree of variability and different clinical presentation compared with previous clades. Here we demonstrate that at least four Omicron lineages circulated in children since December 2021, and studied until November 2022: BA.1 (33.6%), BA.2 (40.6%), BA.5 (23.7%) and BQ.1 (2.1%). At least 70% of infections concerned children under 1 year, most of them being infected with BA.2 lineages (n = 201, 75.6%). Looking at SARS-CoV-2 genetic variability, 69 SNPs were found to be significantly associated in pairs, (phi < - 0.3 or > 0.3 and p-value < 0.001). 16 SNPs were involved in 4 distinct clusters (bootstrap > 0.75). One of these clusters (A23040G, A27259C, T23617G, T23620G) was also positively associated with moderate/severe COVID-19 presentation (AOR [95% CI] 2.49 [1.26-4.89] p-value: 0.008) together with comorbidities (AOR [95% CI] 2.67 [1.36-5.24] p-value: 0.004). Overall, these results highlight the extensive SARS-CoV-2 Omicron circulation in children, mostly aged < 1 year, and provide insights on viral diversification even considering low-abundant SNPs, finally suggesting the potential contribution of viral diversification in affecting disease severity.
Collapse
Affiliation(s)
- Rossana Scutari
- Multimodal Laboratory Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Major School in Microbiology and Virology, Campus Bio-Medico University, Rome, Italy
| | - Valeria Fox
- Multimodal Laboratory Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Vanessa Fini
- Multimodal Laboratory Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Annarita Granaglia
- Multimodal Laboratory Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Anna Chiara Vittucci
- Academic Department of Pediatrics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Smarrazzo
- Academic Department of Pediatrics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Laura Lancella
- Academic Department of Pediatrics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Lorenza Romani
- Academic Department of Pediatrics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Laura Cursi
- Academic Department of Pediatrics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paola Bernaschi
- Microbiology and Diagnostics in Immunology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Cristina Russo
- Microbiology and Diagnostics in Immunology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Campana
- Academic Department of Pediatrics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Stefania Bernardi
- Academic Department of Pediatrics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Alberto Villani
- Academic Department of Pediatrics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Carlo Federico Perno
- Multimodal Laboratory Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
- Microbiology and Diagnostics in Immunology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Claudia Alteri
- Multimodal Laboratory Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
34
|
Edalat F, Khakpour N, Heli H, Letafati A, Ramezani A, Hosseini SY, Moattari A. Immunological mechanisms of the nucleocapsid protein in COVID-19. Sci Rep 2024; 14:3711. [PMID: 38355695 PMCID: PMC10867304 DOI: 10.1038/s41598-024-53906-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 02/06/2024] [Indexed: 02/16/2024] Open
Abstract
The emergence of corona virus disease 2019 (COVID-19), resulting from Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has left an indelible mark on a global scale, causing countless infections and fatalities. This investigation delves into the role of the SARS-CoV-2 nucleocapsid (N) protein within the HEK293 cells, shedding light on its influence over apoptosis, interferon signaling, and cytokines production. The N gene was amplified, inserted into the pAdTrack-CMV vector, and then transfected to the HEK293 cells. Changes in the expression of IRF3, IRF7, IFN-β, BAK, BAX, and BCL-2 genes were evaluated. The levels of proinflammatory cytokines of IL-6, IL-12, IL-1β, and TNF-α were also determined. The N protein exhibited an anti-apoptotic effect by modulating critical genes associated with apoptosis, including BAK, BAX, and BCL-2. This effect potentially prolonged the survival of infected cells. The N protein also played a role in immune evasion by suppressing the interferon pathway, evidenced by the downregulation of essential interferon regulatory factors of IRF3 and IRF7, and IFN-β expression. The N protein expression led to a substantial increase in the production of proinflammatory cytokines of IL-6, IL-12, IL-1β, and TNF-α. The N protein emerged as a versatile factor and was exerted over apoptosis, interferon signaling, and cytokine production. These findings carry potential implications for the development of targeted therapies to combat COVID-19 and mitigate its global health impact.
Collapse
Affiliation(s)
- Fahime Edalat
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Niloofar Khakpour
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Heli
- Nanomedicine and Nanobiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arash Letafati
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Science, Shiraz, Iran
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Afagh Moattari
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
35
|
Gao W, Wang L, Cui W, Wang H, Huang G, Li Z, Li G, Zhang W. Deubiquitinase USP1 regulates sarbecovirus ORF6 protein function. J Virol 2024; 98:e0143723. [PMID: 38084957 PMCID: PMC10804995 DOI: 10.1128/jvi.01437-23] [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: 09/14/2023] [Accepted: 11/27/2023] [Indexed: 01/24/2024] Open
Abstract
SARS-CoV-2 belongs to the subgenus Sarbecovirus, which universally encodes the accessory protein ORF6. SARS-CoV-2 ORF6 is an antagonist of the interferon (IFN)-mediated antiviral response and plays an important role in viral infections. However, the mechanism by which the host counteracts the function of ORF6 to restrict viral replication remains unclear. In this study, we found that most ORF6 proteins encoded by sarbecoviruses could be ubiquitinated and subsequently degraded via the proteasome pathway. Through extensive screening, we identified that the deubiquitinase USP1, which effectively and broadly deubiquitinates sarbecovirus ORF6 proteins, stabilizes ORF6 proteins, resulting in enhanced viral replication. Therefore, ubiquitination and deubiquitination of ORF6 are important for antagonizing IFN-mediated antiviral signaling and influencing the virulence of SARS-CoV-2. These findings highlight an essential molecular mechanism and may provide a novel target for therapeutic interventions against viral infections.IMPORTANCEThe ORF6 proteins encoded by sarbecoviruses are essential for effective viral replication and infection and are important targets for developing effective intervention strategies. In this study, we confirmed that sarbecovirus ORF6 proteins are important antagonists of the host immune response and identified the regulatory mechanisms of ubiquitination and deubiquitination of most sarbecovirus ORF6 proteins. Moreover, we revealed that DUB USP1 prevents the proteasomal degradation of all ORF6 proteins, thereby promoting the virulence of SARS-CoV-2. Thus, impeding ORF6 function is helpful for attenuating the virulence of sarbecoviruses. Therefore, our findings provide a deeper understanding of the molecular mechanisms underlying sarbecovirus infections and offer potential new therapeutic targets for the prevention and treatment of these infections.
Collapse
Affiliation(s)
- Wenying Gao
- Center for Pathogen Biology and Infectious Diseases, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Liuli Wang
- College of Medicine, Jilin University, Changchun, China
| | - Wenzhe Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Hongfei Wang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Guofeng Huang
- Center for Pathogen Biology and Infectious Diseases, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Zhaolong Li
- Center for Pathogen Biology and Infectious Diseases, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Guangquan Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, China
| | - Wenyan Zhang
- Center for Pathogen Biology and Infectious Diseases, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
36
|
Suleman M, Ishaq I, Khan H, Ullah khan S, Masood R, Albekairi NA, Alshammari A, Crovella S. Elucidating the binding mechanism of SARS-CoV-2 NSP6-TBK1 and structure-based designing of phytocompounds inhibitors for instigating the host immune response. Front Chem 2024; 11:1346796. [PMID: 38293247 PMCID: PMC10824840 DOI: 10.3389/fchem.2023.1346796] [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: 11/29/2023] [Accepted: 12/31/2023] [Indexed: 02/01/2024] Open
Abstract
SARS-CoV-2, also referred to as severe acute respiratory syndrome coronavirus 2, is the virus responsible for causing COVID-19, an infectious disease that emerged in Wuhan, China, in December 2019. Among its crucial functions, NSP6 plays a vital role in evading the human immune system by directly interacting with a receptor called TANK-binding kinase (TBK1), leading to the suppression of IFNβ production. Consequently, in the present study we used the structural and biophysical approaches to analyze the effect of newly emerged mutations on the binding of NSP6 and TBK1. Among the identified mutations, four (F35G, L37F, L125F, and I162T) were found to significantly destabilize the structure of NSP6. Furthermore, the molecular docking analysis highlighted that the mutant NSP6 displayed its highest binding affinity with TBK1, exhibiting docking scores of -1436.2 for the wildtype and -1723.2, -1788.6, -1510.2, and -1551.7 for the F35G, L37F, L125F, and I162T mutants, respectively. This suggests the potential for an enhanced immune system evasion capability of NSP6. Particularly, the F35G mutation exhibited the strongest binding affinity, supported by a calculated binding free energy of -172.19 kcal/mol. To disrupt the binding between NSP6 and TBK1, we conducted virtual drug screening to develop a novel inhibitor derived from natural products. From this screening, we identified the top 5 hit compounds as the most promising candidates with a docking score of -6.59 kcal/mol, -6.52 kcal/mol, -6.32 kcal/mol, -6.22 kcal/mol, and -6.21 kcal/mol. The molecular dynamic simulation of top 3 hits further verified the dynamic stability of drugs-NSP6 complexes. In conclusion, this study provides valuable insight into the higher infectivity of the SARS-CoV-2 new variants and a strong rationale for the development of novel drugs against NSP6.
Collapse
Affiliation(s)
- Muhammad Suleman
- Laboratory of Animal Research Center (LARC), Qatar University, Doha, Qatar
- Center for Biotechnology and Microbiology, University of Swat, Swat, Pakistan
| | - Iqra Ishaq
- Center for Biotechnology and Microbiology, University of Swat, Swat, Pakistan
| | - Haji Khan
- Center for Biotechnology and Microbiology, University of Swat, Swat, Pakistan
| | - Safir Ullah khan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Rehana Masood
- Department of Biochemistry, Shaheed Benazir Bhutto Women University, Peshawar, Pakistan
| | - Norah A. Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sergio Crovella
- Laboratory of Animal Research Center (LARC), Qatar University, Doha, Qatar
| |
Collapse
|
37
|
Hsieh MK, Klauda JB. Multiscale Molecular Dynamics Simulations of the Homodimer Accessory Protein ORF7b of SARS-CoV-2. J Phys Chem B 2024; 128:150-162. [PMID: 38147592 DOI: 10.1021/acs.jpcb.3c07105] [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: 12/28/2023]
Abstract
The SARS-CoV-2 ORF7b protein has drawn attention for its potential role in viral pathogenesis, but its structural details and lateral membrane associations remain elusive. In this study, we conducted multiscale molecular dynamics simulations to provide detailed molecular insights of the protein's dimerization, which is crucial for unraveling its structural model of protein-protein interface important to regulating cellular immune response. To gain a deeper understanding of homodimer configurations, we employed a machine learning algorithm for structural-based clustering. Clusters were categorized into three distinct groups for both parallel and antiparallel orientations, highlighting the influence of the initial monomer conformation on dimer configurations. Analysis of hydrogen bonding and π-π and π-cation stacking interactions within clusters revealed variations in interactions between clusters. In parallel dimers, weak stacking interactions in the transmembrane (TM) region were observed. In contrast, antiparallel dimers exhibited strong hydrogen bonding and stacking interactions contributing to tight dimeric packing, both within and outside the TM domain. Overall, our study provides a comprehensive view of the structural dynamics of ORF7b homodimerization in both parallel and antiparallel orientations. These findings shed light on the molecular interactions involved in ORF7b dimerization, which are crucial for understanding its potential roles in SARS-CoV-2 pathogenesis. This knowledge could inform future research and therapeutic strategies targeting this viral protein.
Collapse
Affiliation(s)
- Min-Kang Hsieh
- Department of Chemical and Biomolecular Engineering, University of Maryland College Park, College Park, Maryland 20742, United States
| | - Jeffery B Klauda
- Department of Chemical and Biomolecular Engineering, University of Maryland College Park, College Park, Maryland 20742, United States
- Institute for Physical Science and Technology, Biophysics Program, University of Maryland College Park, College Park, Maryland 20742, United States
| |
Collapse
|
38
|
Chau CW, Sugimura R. Organoids in COVID-19: can we break the glass ceiling? J Leukoc Biol 2024; 115:85-99. [PMID: 37616269 DOI: 10.1093/jleuko/qiad098] [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: 01/30/2023] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
COVID-19 emerged in September 2020 as a disease caused by the virus SARS-CoV-2. The disease presented as pneumonia at first but later was shown to cause multisystem infections and long-term complications. Many efforts have been put into discovering the exact pathogenesis of the disease. In this review, we aim to discuss an emerging tool in disease modeling, organoids, in the investigation of COVID-19. This review will introduce some methods and breakthroughs achieved by organoids and the limitations of this system.
Collapse
Affiliation(s)
- Chiu Wang Chau
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, 21 Sassoon Rd, Pokfulam 99077, Hong Kong
| | - Ryohichi Sugimura
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, 21 Sassoon Rd, Pokfulam 99077, Hong Kong
- Centre for Translational Stem Cell Biology, 17 Science Park W Ave, Science Park 999077, Hong Kong
| |
Collapse
|
39
|
Li W, Li G, Sun Y, Zhang L, Cui X, Jia Y, Zhao T. Prediction of SARS-CoV-2 Infection Phosphorylation Sites and Associations of these Modifications with Lung Cancer Development. Curr Gene Ther 2024; 24:239-248. [PMID: 37957848 DOI: 10.2174/0115665232268074231026111634] [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: 06/20/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 11/15/2023]
Abstract
INTRODUCTION Since the emergence of SARS-CoV-2 viruses, multiple mutant strains have been identified. Infection with SARS-CoV-2 virus leads to alterations in host cell phosphorylation signal, which systematically modulates the immune response. METHODS Identification and analysis of SARS-CoV-2 virus infection phosphorylation sites enable insight into the mechanisms of viral infection and effects on host cells, providing important fundamental data for the study and development of potent drugs for the treatment of immune inflammatory diseases. In this paper, we have analyzed the SARS-CoV-2 virus-infected phosphorylation region and developed a transformer-based deep learning-assisted identification method for the specific identification of phosphorylation sites in SARS-CoV-2 virus-infected host cells. RESULTS Furthermore, through association analysis with lung cancer, we found that SARS-CoV-2 infection may affect the regulatory role of the immune system, leading to an abnormal increase or decrease in the immune inflammatory response, which may be associated with the development and progression of cancer. CONCLUSION We anticipate that this study will provide an important reference for SARS-CoV-2 virus evolution as well as immune-related studies and provide a reliable complementary screening tool for anti-SARS-CoV-2 virus drug and vaccine design.
Collapse
Affiliation(s)
- Wei Li
- Institute of Bioinformatics, Harbin Institute of Technology, Harbin, China
| | - Gen Li
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuzhi Sun
- Institute for Bioinformatics, School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Liyuan Zhang
- Institute for Bioinformatics, School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xinran Cui
- Institute for Bioinformatics, School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yuran Jia
- Institute for Bioinformatics, School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Tianyi Zhao
- School of Medicine and Health, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
40
|
Lee JS, Dittmar M, Miller J, Li M, Ayyanathan K, Ferretti M, Hulahan J, Whig K, Etwebi Z, Griesman T, Schultz DC, Cherry S. Evolutionary arms race between SARS-CoV-2 and interferon signaling via dynamic interaction with autophagy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.13.566859. [PMID: 38014114 PMCID: PMC10680587 DOI: 10.1101/2023.11.13.566859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
SARS-CoV-2 emerged, and is evolving to efficiently infect humans worldwide. SARS-CoV-2 evades early innate recognition, interferon signaling activated only in bystander cells. This balance of innate activation and viral evasion has important consequences, but the pathways involved are incompletely understood. Here we find that autophagy genes regulate innate immune signaling, impacting the basal set point of interferons, and thus permissivity to infection. Mechanistically, autophagy genes negatively regulate MAVS, and this low basal level of MAVS is efficiently antagonized by SARS-CoV-2 ORF9b, blocking interferon activation in infected cells. However, upon loss of autophagy increased MAVS overcomes ORF9b-mediated antagonism suppressing infection. This has led to the evolution of SARS-CoV-2 variants to express higher levels of ORF9b, allowing SARS-CoV-2 to replicate under conditions of increased MAVS signaling. Altogether, we find a critical role of autophagy in the regulation of innate immunity and uncover an evolutionary trajectory of SARS-CoV-2 ORF9b to overcome host defenses.
Collapse
|
41
|
Bosquillon de Jarcy L, Akbil B, Mhlekude B, Leyens J, Postmus D, Harnisch G, Jansen J, Schmidt ML, Aigner A, Pott F, Chua RL, Krist L, Gentile R, Mühlemann B, Jones TC, Niemeyer D, Fricke J, Keil T, Pischon T, Janke J, Conrad C, Iacobelli S, Drosten C, Corman VM, Ralser M, Eils R, Kurth F, Sander L, Goffinet C. 90K/LGALS3BP expression is upregulated in COVID-19 but may not restrict SARS-CoV-2 infection. Clin Exp Med 2023; 23:3689-3700. [PMID: 37162650 PMCID: PMC10170455 DOI: 10.1007/s10238-023-01077-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/12/2023] [Indexed: 05/11/2023]
Abstract
Glycoprotein 90K, encoded by the interferon-stimulated gene LGALS3BP, displays broad antiviral activity. It reduces HIV-1 infectivity by interfering with Env maturation and virion incorporation, and increases survival of Influenza A virus-infected mice via antiviral innate immune signaling. Its antiviral potential in SARS-CoV-2 infection remains largely unknown. Here, we analyzed the expression of 90K/LGALS3BP in 44 hospitalized COVID-19 patients at multiple levels. We quantified 90K protein concentrations in serum and PBMCs as well as LGALS3BP mRNA levels. Complementary, we analyzed two single cell RNA-sequencing datasets for expression of LGALS3BP in respiratory specimens and PBMCs from COVID-19 patients. Finally, we analyzed the potential of 90K to interfere with SARS-CoV-2 infection of HEK293T/ACE2, Calu-3 and Caco-2 cells using authentic virus. 90K protein serum concentrations were significantly elevated in COVID-19 patients compared to uninfected sex- and age-matched controls. Furthermore, PBMC-associated concentrations of 90K protein were overall reduced by SARS-CoV-2 infection in vivo, suggesting enhanced secretion into the extracellular space. Mining of published PBMC scRNA-seq datasets uncovered monocyte-specific induction of LGALS3BP mRNA expression in COVID-19 patients. In functional assays, neither 90K overexpression in susceptible cell lines nor exogenous addition of purified 90K consistently inhibited SARS-CoV-2 infection. Our data suggests that 90K/LGALS3BP contributes to the global type I IFN response during SARS-CoV-2 infection in vivo without displaying detectable antiviral properties in vitro.
Collapse
Affiliation(s)
- Laure Bosquillon de Jarcy
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 , Berlin, Germany
- Speciality Network: Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Bengisu Akbil
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 , Berlin, Germany
| | - Baxolele Mhlekude
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 , Berlin, Germany
| | - Johanna Leyens
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Dylan Postmus
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 , Berlin, Germany
| | - Greta Harnisch
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jenny Jansen
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Marie L Schmidt
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Annette Aigner
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Fabian Pott
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 , Berlin, Germany
| | - Robert Lorenz Chua
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany
| | - Lilian Krist
- Institute of Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | | | - Barbara Mühlemann
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Terence C Jones
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Zoology, Centre for Pathogen Evolution, University of Cambridge, Downing St., Cambridge, CB2 3EJ, UK
| | - Daniela Niemeyer
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Infection Research, Associated Partner Charité, Berlin, Germany
| | - Julia Fricke
- Institute of Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Thomas Keil
- Institute of Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Josef-Schneiderstr. 2, 97080, Würzburg, Germany
- State Institute of Health, Bavarian Health and Food Safety Authority, Eggenreuther Weg 43, 91058, Erlangen, Germany
| | - Tobias Pischon
- Molecular Epidemiology Research Group, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
- Biobank Technology Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
- Core Facility Biobank, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10178, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, 10117, Berlin, Germany
| | - Jürgen Janke
- Biobank Technology Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany
| | - Christian Conrad
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany
| | | | - Christian Drosten
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Infection Research, Associated Partner Charité, Berlin, Germany
| | - Victor M Corman
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Infection Research, Associated Partner Charité, Berlin, Germany
| | - Markus Ralser
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, NW11AT, UK
| | - Roland Eils
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 , Berlin, Germany
- Center for Digital Health, Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Lung Research (DZL), 35392, Gießen, Germany
- Health Data Science Unit, Heidelberg University Hospital and BioQuant, 69120, Heidelberg, Germany
| | - Florian Kurth
- Speciality Network: Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, 20359, Hamburg, Germany
- Department of Medicine, University Medical Center, Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Leif Sander
- Speciality Network: Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Lung Research (DZL), 35392, Gießen, Germany
| | - Christine Goffinet
- Institute of Virology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 , Berlin, Germany.
| |
Collapse
|
42
|
Zhang D, Ji L, Chen X, He Y, Sun Y, Ji L, Zhang T, Shen Q, Wang X, Wang Y, Yang S, Zhang W, Zhou C. SARS-CoV-2 Nsp15 suppresses type I interferon production by inhibiting IRF3 phosphorylation and nuclear translocation. iScience 2023; 26:107705. [PMID: 37680466 PMCID: PMC10480782 DOI: 10.1016/j.isci.2023.107705] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/23/2023] [Accepted: 08/21/2023] [Indexed: 09/09/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes 2019 coronavirus disease (COVID-19), poses a significant threat to global public health security. Like other coronaviruses, SARS-CoV-2 has developed various strategies to inhibit the production of interferon (IFN). Here, we have discovered that SARS-CoV-2 Nsp15 obviously reduces the expression of IFN-β and IFN-stimulated genes (ISG56, CXCL10), and also inhibits IRF3 phosphorylation and nuclear translocation by antagonizing the RLR-mediated antiviral signaling pathway. Mechanically, we found that the poly-U-specific endonuclease domain (EndoU) of Nsp15 directly associates with the kinase domain (KD) of TBK1 to interfere TBK1 interacting with IRF3 and the flowing TBK1-mediated IRF3 phosphorylation. Furthermore, Nsp15 also prevented nuclear translocation of phosphorylated IRF3 via binding to the nuclear import adaptor karyopherin α1 (KPNA1) and promoting it autophagy-dependent degradation. These findings collectively reveal a novel mechanism by which Nsp15 antagonizes host's innate immune response.
Collapse
Affiliation(s)
- Dianqi Zhang
- Clinical Laboratory Center, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
- Department of Clinical Laboratory, The Affiliated Yixing Hospital of Jiangsu University, Yixing, Jiangsu 214221, China
| | - Likai Ji
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Xu Chen
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
- Department of Laboratory Medicine and Pathology, Jiangsu Provincial Corps Hospital of Chinese People’s Armed Police Force, Yangzhou, Jiangsu 225003, China
| | - Yumin He
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
- Medical Research Center, Northern Jiangsu People’s Hospital, Yangzhou, Jiangsu 225001, China
| | - Yijie Sun
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Li Ji
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Tiancheng Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Quan Shen
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Xiaochun Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yan Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Shixing Yang
- Clinical Laboratory Center, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Wen Zhang
- Clinical Laboratory Center, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Chenglin Zhou
- Clinical Laboratory Center, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China
| |
Collapse
|
43
|
Bergeron HC, Hansen MR, Tripp RA. Interferons-Implications in the Immune Response to Respiratory Viruses. Microorganisms 2023; 11:2179. [PMID: 37764023 PMCID: PMC10535750 DOI: 10.3390/microorganisms11092179] [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: 07/17/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
Interferons (IFN) are an assemblage of signaling proteins made and released by various host cells in response to stimuli, including viruses. Respiratory syncytial virus (RSV), influenza virus, and SARS-CoV-2 are major causes of respiratory disease that induce or antagonize IFN responses depending on various factors. In this review, the role and function of type I, II, and III IFN responses to respiratory virus infections are considered. In addition, the role of the viral proteins in modifying anti-viral immunity is noted, as are the specific IFN responses that underly the correlates of immunity and protection from disease.
Collapse
Affiliation(s)
| | | | - Ralph A. Tripp
- Department of Infectious Diseases, University of Georgia College of Veterinary Medicine, Athens, GA 30605, USA; (H.C.B.); (M.R.H.)
| |
Collapse
|
44
|
Staheli JP, Neal ML, Navare A, Mast FD, Aitchison JD. Predicting host-based, synthetic lethal antiviral targets from omics data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.15.553430. [PMID: 37645861 PMCID: PMC10462099 DOI: 10.1101/2023.08.15.553430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Traditional antiviral therapies often have limited effectiveness due to toxicity and development of drug resistance. Host-based antivirals, while an alternative, may lead to non-specific effects. Recent evidence shows that virus-infected cells can be selectively eliminated by targeting synthetic lethal (SL) partners of proteins disrupted by viral infection. Thus, we hypothesized that genes depleted in CRISPR KO screens of virus-infected cells may be enriched in SL partners of proteins altered by infection. To investigate this, we established a computational pipeline predicting SL drug targets of viral infections. First, we identified SARS-CoV-2-induced changes in gene products via a large compendium of omics data. Second, we identified SL partners for each altered gene product. Last, we screened CRISPR KO data for SL partners required for cell viability in infected cells. Despite differences in virus-induced alterations detected by various omics data, they share many predicted SL targets, with significant enrichment in CRISPR KO-depleted datasets. Comparing data from SARS-CoV-2 and influenza infections, we found possible broad-spectrum, host-based antiviral SL targets. This suggests that CRISPR KO data are replete with common antiviral targets due to their SL relationship with virus-altered states and that such targets can be revealed from analysis of omics datasets and SL predictions.
Collapse
Affiliation(s)
- Jeannette P. Staheli
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, 98101, USA
| | - Maxwell L. Neal
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, 98101, USA
| | - Arti Navare
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, 98101, USA
| | - Fred D. Mast
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, 98101, USA
| | - John D. Aitchison
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, 98101, USA
| |
Collapse
|
45
|
Ye C, Park JG, Chiem K, Dravid P, Allué-Guardia A, Garcia-Vilanova A, Pino Tamayo P, Shivanna V, Kapoor A, Walter MR, Kobie JJ, Plemper RK, Torrelles JB, Martinez-Sobrido L. Immunization with Recombinant Accessory Protein-Deficient SARS-CoV-2 Protects against Lethal Challenge and Viral Transmission. Microbiol Spectr 2023; 11:e0065323. [PMID: 37191507 PMCID: PMC10269623 DOI: 10.1128/spectrum.00653-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/27/2023] [Indexed: 05/17/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to a worldwide coronavirus disease 2019 (COVID-19) pandemic. Despite the high efficacy of the authorized vaccines, there may be uncertain and unknown side effects or disadvantages associated with current vaccination approaches. Live-attenuated vaccines (LAVs) have been shown to elicit robust and long-term protection by the induction of host innate and adaptive immune responses. In this study, we sought to verify an attenuation strategy by generating 3 double open reading frame (ORF)-deficient recombinant SARS-CoV-2s (rSARS-CoV-2s) simultaneously lacking two accessory ORF proteins (ORF3a/ORF6, ORF3a/ORF7a, and ORF3a/ORF7b). We report that these double ORF-deficient rSARS-CoV-2s have slower replication kinetics and reduced fitness in cultured cells compared with their parental wild-type (WT) counterpart. Importantly, these double ORF-deficient rSARS-CoV-2s showed attenuation in both K18 hACE2 transgenic mice and golden Syrian hamsters. A single intranasal dose vaccination induced high levels of neutralizing antibodies against SARS-CoV-2 and some variants of concern and activated viral component-specific T cell responses. Notably, double ORF-deficient rSARS-CoV-2s were able to protect, as determined by the inhibition of viral replication, shedding, and transmission, against challenge with SARS-CoV-2 in both K18 hACE2 mice and golden Syrian hamsters. Collectively, our results demonstrate the feasibility of implementing the double ORF-deficient strategy to develop safe, immunogenic, and protective LAVs to prevent SARS-CoV-2 infection and associated COVID-19. IMPORTANCE Live-attenuated vaccines (LAVs) are able to induce robust immune responses, including both humoral and cellular immunity, representing a very promising option to provide broad and long-term immunity. To develop LAVs for SARS-CoV-2, we engineered attenuated recombinant SARS-CoV-2 (rSARS-CoV-2) that simultaneously lacks the viral open reading frame 3a (ORF3a) in combination with either ORF6, ORF7a, or ORF7b (Δ3a/Δ6, Δ3a/Δ7a, and Δ3a/Δ7b, respectively) proteins. Among them, the rSARS-CoV-2 Δ3a/Δ7b was completely attenuated and able to provide 100% protection against an otherwise lethal challenge in K18 hACE2 transgenic mice. Moreover, the rSARS-CoV-2 Δ3a/Δ7b conferred protection against viral transmission between golden Syrian hamsters.
Collapse
Affiliation(s)
- Chengjin Ye
- Disease Intervention and Prevention, and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Jun-Gyu Park
- Disease Intervention and Prevention, and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Kevin Chiem
- Disease Intervention and Prevention, and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Piyush Dravid
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Anna Allué-Guardia
- Disease Intervention and Prevention, and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Andreu Garcia-Vilanova
- Disease Intervention and Prevention, and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Paula Pino Tamayo
- Disease Intervention and Prevention, and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Vinay Shivanna
- Disease Intervention and Prevention, and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Amit Kapoor
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Mark R. Walter
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - James J. Kobie
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Richard K. Plemper
- Center for Translational Antiviral Research, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Jordi B. Torrelles
- Disease Intervention and Prevention, and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Luis Martinez-Sobrido
- Disease Intervention and Prevention, and Population Health Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| |
Collapse
|
46
|
Rudraraju R, Gartner MJ, Neil JA, Stout ES, Chen J, Needham EJ, See M, Mackenzie-Kludas C, Yang Lee LY, Wang M, Pointer H, Karavendzas K, Abu-Bonsrah D, Drew D, Yang Sun YB, Tan JP, Sun G, Salavaty A, Charitakis N, Nim HT, Currie PD, Tham WH, Porrello E, Polo JM, Humphrey SJ, Ramialison M, Elliott DA, Subbarao K. Parallel use of human stem cell lung and heart models provide insights for SARS-CoV-2 treatment. Stem Cell Reports 2023; 18:1308-1324. [PMID: 37315523 PMCID: PMC10262339 DOI: 10.1016/j.stemcr.2023.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 06/16/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) primarily infects the respiratory tract, but pulmonary and cardiac complications occur in severe coronavirus disease 2019 (COVID-19). To elucidate molecular mechanisms in the lung and heart, we conducted paired experiments in human stem cell-derived lung alveolar type II (AT2) epithelial cell and cardiac cultures infected with SARS-CoV-2. With CRISPR-Cas9-mediated knockout of ACE2, we demonstrated that angiotensin-converting enzyme 2 (ACE2) was essential for SARS-CoV-2 infection of both cell types but that further processing in lung cells required TMPRSS2, while cardiac cells required the endosomal pathway. Host responses were significantly different; transcriptome profiling and phosphoproteomics responses depended strongly on the cell type. We identified several antiviral compounds with distinct antiviral and toxicity profiles in lung AT2 and cardiac cells, highlighting the importance of using several relevant cell types for evaluation of antiviral drugs. Our data provide new insights into rational drug combinations for effective treatment of a virus that affects multiple organ systems.
Collapse
Affiliation(s)
- Rajeev Rudraraju
- The Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Matthew J Gartner
- The Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Jessica A Neil
- The Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Elizabeth S Stout
- The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Joseph Chen
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Elise J Needham
- Charles Perkins Centre and School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Camperdown, NSW, Australia
| | - Michael See
- The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, VIC, Australia; Monash Bioinformatics Platform, Monash University, Clayton, VIC, Australia
| | - Charley Mackenzie-Kludas
- The Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Leo Yi Yang Lee
- The Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Mingyang Wang
- The Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Hayley Pointer
- The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Kathy Karavendzas
- The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Dad Abu-Bonsrah
- The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Damien Drew
- Infection and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Yu Bo Yang Sun
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Jia Ping Tan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Guizhi Sun
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Adrian Salavaty
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia; EMBL Australia, Monash University, Clayton, VIC, Australia
| | - Natalie Charitakis
- The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Pediatrics, The Royal Children's Hospital, University of Melbourne Parkville, VIC, Australia
| | - Hieu T Nim
- The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia; Department of Pediatrics, The Royal Children's Hospital, University of Melbourne Parkville, VIC, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia; EMBL Australia, Monash University, Clayton, VIC, Australia
| | - Wai-Hong Tham
- Infection and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Enzo Porrello
- The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, VIC, Australia; Melbourne Centre for Cardiovascular Genomics and Regenerative Medicine, The Royal Children's Hospital, Melbourne, VIC, Australia; Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC, Australia.
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia; Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia.
| | - Sean J Humphrey
- Charles Perkins Centre and School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Camperdown, NSW, Australia.
| | - Mirana Ramialison
- The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia; Department of Pediatrics, The Royal Children's Hospital, University of Melbourne Parkville, VIC, Australia.
| | - David A Elliott
- The Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Melbourne, VIC, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia; Department of Pediatrics, The Royal Children's Hospital, University of Melbourne Parkville, VIC, Australia.
| | - Kanta Subbarao
- The Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia; The WHO Collaborating Centre for Reference and Research on Influenza, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| |
Collapse
|
47
|
Rodriguez-Rodriguez BA, Ciabattoni GO, Duerr R, Valero-Jimenez AM, Yeung ST, Crosse KM, Schinlever AR, Bernard-Raichon L, Rodriguez Galvan J, McGrath ME, Vashee S, Xue Y, Loomis CA, Khanna KM, Cadwell K, Desvignes L, Frieman MB, Ortigoza MB, Dittmann M. A neonatal mouse model characterizes transmissibility of SARS-CoV-2 variants and reveals a role for ORF8. Nat Commun 2023; 14:3026. [PMID: 37230979 PMCID: PMC10211296 DOI: 10.1038/s41467-023-38783-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
Small animal models have been a challenge for the study of SARS-CoV-2 transmission, with most investigators using golden hamsters or ferrets. Mice have the advantages of low cost, wide availability, less regulatory and husbandry challenges, and the existence of a versatile reagent and genetic toolbox. However, adult mice do not robustly transmit SARS-CoV-2. Here we establish a model based on neonatal mice that allows for transmission of clinical SARS-CoV-2 isolates. We characterize tropism, respiratory tract replication and transmission of ancestral WA-1 compared to variants Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), Omicron BA.1 and Omicron BQ.1.1. We identify inter-variant differences in timing and magnitude of infectious particle shedding from index mice, both of which shape transmission to contact mice. Furthermore, we characterize two recombinant SARS-CoV-2 lacking either the ORF6 or ORF8 host antagonists. The removal of ORF8 shifts viral replication towards the lower respiratory tract, resulting in significantly delayed and reduced transmission in our model. Our results demonstrate the potential of our neonatal mouse model to characterize viral and host determinants of SARS-CoV-2 transmission, while revealing a role for an accessory protein in this context.
Collapse
Affiliation(s)
| | - Grace O Ciabattoni
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ralf Duerr
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Department of Medicine/Division of Infectious Diseases and Immunology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Vaccine Center, NYU Grossmann of Medicine, New York, NY, 10016, USA
| | - Ana M Valero-Jimenez
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Stephen T Yeung
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Keaton M Crosse
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Austin R Schinlever
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Lucie Bernard-Raichon
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Joaquin Rodriguez Galvan
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Marisa E McGrath
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Sanjay Vashee
- Department of Synthetic Biology and Bioenergy, J. Craig Venter Institute, Rockville, MD, 20850, USA
| | - Yong Xue
- Department of Synthetic Biology and Bioenergy, J. Craig Venter Institute, Rockville, MD, 20850, USA
| | - Cynthia A Loomis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Kamal M Khanna
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- Perlmutter Cancer Center, New York University Langone Health, New York, NY, 10016, USA
| | - Ken Cadwell
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Ludovic Desvignes
- Department of Medicine/Division of Infectious Diseases and Immunology, New York University Grossman School of Medicine, New York, NY, 10016, USA
- High Containment Laboratories - Office of Science and Research, NYU Langone Health, New York, NY, 10016, USA
| | - Matthew B Frieman
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Mila B Ortigoza
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Department of Medicine/Division of Infectious Diseases and Immunology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| | - Meike Dittmann
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
48
|
Bills CJ, Xia H, Chen JYC, Yeung J, Kalveram B, Walker D, Xie X, Shi PY. Mutations in SARS-CoV-2 variant nsp6 enhance type-I interferon antagonism. Emerg Microbes Infect 2023; 12:2209208. [PMID: 37114433 PMCID: PMC10184609 DOI: 10.1080/22221751.2023.2209208] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to evolve after its emergence. Given its importance in viral infection and vaccine development, mutations in the viral Spike gene have been studied extensively; however, the impact of mutations outside the Spike gene are poorly understood. Here, we report that a triple deletion (ΔSGF or ΔLSG) in nonstructural protein 6 (nsp6) independently acquired in Alpha and Omicron sublineages of SARS-CoV-2 augments nsp6-mediated antagonism of type-I interferon (IFN-I) signaling. Specifically, these triple deletions enhance the ability of mutant nsp6 to suppress phosphorylation of STAT1 and STAT2. A parental SARS-CoV-2 USA-WA1/2020 strain containing the nsp6 ΔSGF deletion (ΔSGF-WA1) shows reduced susceptibility to IFN-I treatment in vitro, outcompetes the parental strain in human primary airway cultures, and increases virulence in mice; however, the ΔSGF-WA1 virus is less virulent than the Alpha variant (which has the nsp6 ΔSGF deletion and additional mutations in other genes). Analyses of host responses from ΔSGF-WA1-infected mice and primary airway cultures reveal activation of pathways indicative of a cytokine storm. These results provide evidence that mutations outside the Spike protein affect virus-host interactions and may alter pathogenesis of SARS-CoV-2 variants in humans.
Collapse
Affiliation(s)
- Cody J Bills
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Hongjie Xia
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - John Yun-Chung Chen
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jason Yeung
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Birte Kalveram
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - David Walker
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
- Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
49
|
Oleinik LA, Madonov PG, Pykhtina MB. Potential of Interferon Lambda as an Inhibitor of SARS-CoV-2. Mol Biol 2023; 57:291-298. [PMID: 37128210 PMCID: PMC10131541 DOI: 10.1134/s0026893323020152] [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: 08/05/2022] [Revised: 10/06/2022] [Accepted: 10/06/2022] [Indexed: 05/03/2023]
Abstract
This study provides an overview of scientific results on the feasibility of using type III interferons against SARS-CoV-2. We have analyzed data obtained from the PubMed electronic database for the period 2020‒2022. The results of our own studies of pharmacological substances based on recombinant IFN-λ1 and its pegylated form are also presented. Completed and ongoing investigations allow us to position IFN-λ as an effective therapeutic agent against SARS-CoV-2.
Collapse
Affiliation(s)
- L. A. Oleinik
- Research Institute of Clinical and Experimental Lymрhology—Branch of Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - P. G. Madonov
- Research Institute of Clinical and Experimental Lymрhology—Branch of Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - M. B. Pykhtina
- Research Institute of Clinical and Experimental Lymрhology—Branch of Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
50
|
Godbold GD, Hewitt FC, Kappell AD, Scholz MB, Agar SL, Treangen TJ, Ternus KL, Sandbrink JB, Koblentz GD. Improved understanding of biorisk for research involving microbial modification using annotated sequences of concern. Front Bioeng Biotechnol 2023; 11:1124100. [PMID: 37180048 PMCID: PMC10167326 DOI: 10.3389/fbioe.2023.1124100] [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: 12/14/2022] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
Regulation of research on microbes that cause disease in humans has historically been focused on taxonomic lists of 'bad bugs'. However, given our increased knowledge of these pathogens through inexpensive genome sequencing, 5 decades of research in microbial pathogenesis, and the burgeoning capacity of synthetic biologists, the limitations of this approach are apparent. With heightened scientific and public attention focused on biosafety and biosecurity, and an ongoing review by US authorities of dual-use research oversight, this article proposes the incorporation of sequences of concern (SoCs) into the biorisk management regime governing genetic engineering of pathogens. SoCs enable pathogenesis in all microbes infecting hosts that are 'of concern' to human civilization. Here we review the functions of SoCs (FunSoCs) and discuss how they might bring clarity to potentially problematic research outcomes involving infectious agents. We believe that annotation of SoCs with FunSoCs has the potential to improve the likelihood that dual use research of concern is recognized by both scientists and regulators before it occurs.
Collapse
Affiliation(s)
| | | | | | | | - Stacy L. Agar
- Signature Science, LLC, Charlottesville, VA, United States
| | - Todd J. Treangen
- Department of Computer Science, Rice University, Houston, TX, United States
| | | | - Jonas B. Sandbrink
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Gregory D. Koblentz
- Schar School of Policy and Government, George Mason University, Arlington, VA, United States
| |
Collapse
|