1
|
Kou L, Wan Y, Nemoto YL, Tsujita K, Itoh T. Role of membrane proximal actin regulators in SARS-CoV-2 spike-induced cell-cell fusion. Biochem Biophys Res Commun 2025; 766:151846. [PMID: 40300332 DOI: 10.1016/j.bbrc.2025.151846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/01/2025]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein has the ability to induce multinucleated syncytia via cell-cell fusion, which is thought to be related to the pathogenesis of the coronavirus disease 2019 (COVID-19). However, the mechanism by which spike protein regulates cell fusion remains unclear. Given the close correlation between cell-cell fusion and membrane protrusions, we investigated the role of membrane-proximal actin regulators in spike-induced cell fusion. We found that while Rac-Arp2/3 dependent branched actin polymerization is required for spike-mediated cell fusion, RhoA dependent actomyosin contractility has an inhibitory effect on fusion. In addition, plasma membrane tension regulated by membrane-cortex attachment plays a negative role in spike-dependent cell fusion. Furthermore, we identified several BAR proteins, which couple membrane curvature with actin dynamics, involved in spike-induced syncytia formation. Our study suggests that these actin regulators could be promising targets for inhibiting SARS-CoV-2 spike protein-induced syncytia formation.
Collapse
Affiliation(s)
- Linting Kou
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan
| | - Yumeng Wan
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan
| | - Yuri L Nemoto
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan; Biosignal Research Center, Kobe University, Kobe, Hyogo, 657-8501, Japan
| | - Kazuya Tsujita
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan; Biosignal Research Center, Kobe University, Kobe, Hyogo, 657-8501, Japan.
| | - Toshiki Itoh
- Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Hyogo, 650-0017, Japan; Biosignal Research Center, Kobe University, Kobe, Hyogo, 657-8501, Japan.
| |
Collapse
|
2
|
Saminathan P, Mathews IT, Alimadadi A, Fung K, Kakugawa K, Joosten LA, Netea MG, Jain M, Cheng S, Hedrick CC, Sharma S. Sex differences in adenosine deaminase activity associate with disparities in SARS-CoV-2 innate immunity. iScience 2025; 28:112418. [PMID: 40343269 PMCID: PMC12059719 DOI: 10.1016/j.isci.2025.112418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/08/2025] [Accepted: 04/09/2025] [Indexed: 05/11/2025] Open
Abstract
Females demonstrate elevated type-I interferon production and a stronger antiviral immune response; however, the mechanisms underlying sex-based differences in antiviral immunity are incompletely understood. We previously reported that low adenosine deaminase (ADA) activity perturbs the methylation-based transcriptional silencing of endogenous retroviral elements (hERV), which stimulates IFN-Stimulated Genes (ISG) and primes antiviral immunity. Here we demonstrate lower ADA activity in females compared to their male counterparts, which correlated with higher hERV and ISG expression in female lungs. Sex differences in ADA2 were linked to the number and expression profiles of blood and lung-derived monocyte populations. Single-cell RNA sequencing of respiratory cells from patients with COVID-19 showed a significant female bias in hERV-ISG signatures, and implicated IL-18 as a driver of sex-specific ADA2 expression. Observations in healthy and COVID-19 cohorts indicate that higher ADA activity is associated with suppressed antiviral innate immunity in the male respiratory tract, which may drive adverse COVID-19 outcomes.
Collapse
Affiliation(s)
- Priyanka Saminathan
- Center for Sex Differences in the Immune System, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Ian T. Mathews
- Center for Sex Differences in the Immune System, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ahmad Alimadadi
- Center for Sex Differences in the Immune System, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Immunology Center of Georgia and Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Kai Fung
- Center for Sex Differences in the Immune System, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Kiyokazu Kakugawa
- Laboratory for Inflammatory Immune Metabolism, RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa 230-0045, Japan
| | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Community Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Community Center for Infectious Diseases, Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn 53127, Germany
| | - Mohit Jain
- Sapient Bioanalytics, San Diego, CA 92121, USA
| | - Susan Cheng
- Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Catherine C. Hedrick
- Immunology Center of Georgia and Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Sonia Sharma
- Center for Sex Differences in the Immune System, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Laboratory for Inflammatory Immune Metabolism, RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa 230-0045, Japan
| |
Collapse
|
3
|
Dong D, Song Y, Wu S, Wang B, Peng C, Zhang W, Kong W, Zhang Z, Song J, Hou LH, Li S. Molecular basis of Ad5-nCoV vaccine-induced immunogenicity. Structure 2025; 33:858-868.e5. [PMID: 40112804 DOI: 10.1016/j.str.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/12/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
Ad5-nCoV (Convidecia) is listed for emergency use against COVID-19 by the World Health Organization (WHO) and has been globally administered to millions of people. It utilizes human adenovirus 5 (Ad5) replication-incompetent vector to deliver the spike (S) protein gene from various SARS-CoV-2 strains. Despite promising clinical data, the molecular mechanism underlying its high immunogenicity and adverse reactions remain incompletely understood. Here, we primarily applied cryo-electron tomography (cryo-ET), fluorescence microscopy and mass spectrometry to analyze the Ad5-nCoV_Wu and Ad5-nCoV_O vaccine-induced S antigens. These antigens encode the unmodified SARS-CoV-2 Wuhan-Hu-1 S gene and the stabilized Omicron S gene, respectively. Our findings highlight the structural integrity, antigenicity, and dense distribution on cell membrane of the vaccine-induced S proteins. Ad5-nCoV_O induced S proteins exhibit improved stability and reduced syncytia formation among inoculated cells. Our work demonstrates that Ad5-nCoV is a prominent platform for antigen induction and cryo-ET can be a useful technique for vaccine characterization and development.
Collapse
Affiliation(s)
- Dongyang Dong
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yutong Song
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shipo Wu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Busen Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Cheng Peng
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Weiping Zhang
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Weizheng Kong
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zheyuan Zhang
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jingwen Song
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Li-Hua Hou
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Sai Li
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
4
|
Wu L, Yan Y, Yuan Y, Zhao Z, Qu W, Huang X, Wang H, Li P, Li X. Viral protease binds to nucleosomal DNA and cleaves nuclear cGAS that attenuates type I interferon. mBio 2025; 16:e0339524. [PMID: 39998223 PMCID: PMC11980361 DOI: 10.1128/mbio.03395-24] [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: 10/30/2024] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Nuclear cyclic GMP-AMP synthetase (cGAS) binds to nucleosome with high affinity to prevent its activation by self-DNA. Upon stimulation with double-stranded DNA, cGAS is activated and translocates from the nucleus to the cytoplasm, guided by its N-terminal domain. However, it remains unclear whether viruses can hijack cGAS translocation and regulate its activation. Here, we discovered that the protease 3C of picornavirus Seneca Valley virus (SVV) translocates from the cytoplasm to the nucleus upon viral infection and binds to nuclear DNA. Protease 3C specifically cleaves histone H2A while leaving other histone proteins unaffected. Additionally, DNA binding enhances the protease 3C's ability to cleave nuclear cGAS, leading to its retention in the nucleus. This, in turn, suppresses the induction of type I interferon (IFN-I) following poly(dA:dT) stimulation. These findings reveal a novel mechanism by which a viral protease binds nuclear DNA, cleaves nuclear cGAS and histone H2A, and thereby mislocalizes cGAS, facilitating immune evasion. IMPORTANCE Cyclic GMP-AMP synthetase (cGAS) is robustly expressed in the nucleus and tightly tethered by chromatin to prevent its activation with self-DNA. During stimulation or infection, nuclear cGAS is activated and translocates from the nucleus to the cytoplasm. However, the viral strategies specifically targeting nuclear cGAS are completely unexplored. Here, we discovered that protease 3C of Seneca Valley virus translocates from the cytoplasm to the nucleus upon viral infection, binds to nuclear DNA, and specifically cleaves H2A. Furthermore, DNA binding to 3C enhances the cleavage of nuclear cGAS within its N-terminal domain. The hindrance of cGAS translocation from the nucleus to the cytoplasm results in the suppression of IFN-I induction and leads to immune evasion. This work uncovers a unique mechanism wherein a viral protease binds to nuclear DNA and cleaves nuclear cGAS and histone H2A, leading to viral evasion of cGAS-mediated immune restriction.
Collapse
Affiliation(s)
- Lei Wu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ya Yan
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Ye Yuan
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhenchao Zhao
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Weiyu Qu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiangyu Huang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Haiwei Wang
- State Key Laboratory of Animal Disease Control, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Xin Li
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
5
|
Shen Y, Huang W, Nie J, Zhang L. Progress Update on STING Agonists as Vaccine Adjuvants. Vaccines (Basel) 2025; 13:371. [PMID: 40333245 PMCID: PMC12030840 DOI: 10.3390/vaccines13040371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/18/2025] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Low antigen immunogenicity poses a significant challenge in vaccine development, often leading to inadequate immune responses and reduced vaccine efficacy. Therefore, the discovery of potent immune-enhancing adjuvants is crucial. STING (stimulator of interferon genes) agonists are a promising class of adjuvants which have been identified in various immune cells and are activated in response to DNA fragments, triggering a broad range of type-I interferon-dependent immune responses. Integrating STING agonists with vaccine components is an ideal strategy to bolster vaccine-induced immunity to infections and cancer cells. Several STING agonists are currently under investigation in preclinical studies and clinical trials; however, some have shown limited efficacy, while others exhibit off-target effects. To ensure safety, they are typically delivered with carriers that exhibit high biocompatibility and insolubility. In this review, we present the latest research on natural and synthetic STING agonists that have been effectively used in vaccine development, and summarize their application in adjuvant preventive and therapeutic vaccines. Additionally, we discuss the safety of STING agonists as vaccine adjuvants by reviewing potential delivery strategies. Overall, incorporating STING agonists into vaccine formulations represents a significant advancement in vaccine research with the potential to significantly enhance immune responses and improve vaccine efficacy. However, ongoing research is still required to identify the most effective and safe delivery strategies for STING agonists, as well as to evaluate their long-term safety and efficacy in clinical trials.
Collapse
Affiliation(s)
- Yanru Shen
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China; (Y.S.); (W.H.); (J.N.)
| | - Weijin Huang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China; (Y.S.); (W.H.); (J.N.)
- WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing 102629, China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China
| | - Jianhui Nie
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China; (Y.S.); (W.H.); (J.N.)
- WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing 102629, China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China
| | - Li Zhang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China; (Y.S.); (W.H.); (J.N.)
- WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing 102629, China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China
| |
Collapse
|
6
|
Luksch H, Schulze F, Geißler-Lösch D, Sprott D, Höfs L, Szegö EM, Tonnus W, Winkler S, Günther C, Linkermann A, Behrendt R, Teichmann LL, Falkenburger BH, Rösen-Wolff A. Tissue inflammation induced by constitutively active STING is mediated by enhanced TNF signaling. eLife 2025; 14:e101350. [PMID: 40111902 PMCID: PMC11996172 DOI: 10.7554/elife.101350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 03/17/2025] [Indexed: 03/22/2025] Open
Abstract
Constitutive activation of STING by gain-of-function mutations triggers manifestation of the systemic autoinflammatory disease STING-associated vasculopathy with onset in infancy (SAVI). In order to investigate the role of signaling by tumor necrosis factor (TNF) in SAVI, we used genetic inactivation of TNF receptors 1 and 2 in murine SAVI, which is characterized by T cell lymphopenia, inflammatory lung disease, and neurodegeneration. Genetic inactivation of TNFR1 and TNFR2, however, rescued the loss of thymocytes, reduced interstitial lung disease, and neurodegeneration. Furthermore, genetic inactivation of TNFR1 and TNFR2 blunted transcription of cytokines, chemokines, and adhesions proteins, which result from chronic STING activation in SAVI mice. In addition, increased transendothelial migration of neutrophils was ameliorated. Taken together, our results demonstrate a pivotal role of TNFR signaling in the pathogenesis of SAVI in mice and suggest that available TNFR antagonists could ameliorate SAVI in patients.
Collapse
Affiliation(s)
- Hella Luksch
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Felix Schulze
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - David Geißler-Lösch
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - David Sprott
- Department of Physiology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Lennart Höfs
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Eva M Szegö
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine III, Faculty of Medicine and University Hospital Carl GustavDresdenGermany
| | - Stefan Winkler
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Claudia Günther
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, Faculty of Medicine and University Hospital Carl GustavDresdenGermany
| | - Rayk Behrendt
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital BonnBonnGermany
| | | | - Björn H Falkenburger
- Department of Neurology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
- Deutsches Zentrum für Neurodegenerative ErkrankungenDresdenGermany
| | - Angela Rösen-Wolff
- Department of Pediatrics, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität DresdenDresdenGermany
| |
Collapse
|
7
|
Chen Z, Behrendt R, Wild L, Schlee M, Bode C. Cytosolic nucleic acid sensing as driver of critical illness: mechanisms and advances in therapy. Signal Transduct Target Ther 2025; 10:90. [PMID: 40102400 PMCID: PMC11920230 DOI: 10.1038/s41392-025-02174-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Nucleic acids from both self- and non-self-sources act as vital danger signals that trigger immune responses. Critical illnesses such as acute respiratory distress syndrome, sepsis, trauma and ischemia lead to the aberrant cytosolic accumulation and massive release of nucleic acids that are detected by antiviral innate immune receptors in the endosome or cytosol. Activation of receptors for deoxyribonucleic acids and ribonucleic acids triggers inflammation, a major contributor to morbidity and mortality in critically ill patients. In the past decade, there has been growing recognition of the therapeutic potential of targeting nucleic acid sensing in critical care. This review summarizes current knowledge of nucleic acid sensing in acute respiratory distress syndrome, sepsis, trauma and ischemia. Given the extensive research on nucleic acid sensing in common pathological conditions like cancer, autoimmune disorders, metabolic disorders and aging, we provide a comprehensive summary of nucleic acid sensing beyond critical illness to offer insights that may inform its role in critical conditions. Additionally, we discuss potential therapeutic strategies that specifically target nucleic acid sensing. By examining nucleic acid sources, sensor activation and function, as well as the impact of regulating these pathways across various acute diseases, we highlight the driving role of nucleic acid sensing in critical illness.
Collapse
Affiliation(s)
- Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Rayk Behrendt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Lennart Wild
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
8
|
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
|
9
|
Rui Y, Shen S, Wang Y, Cheng L, Chen S, Hu Y, Cai Y, Wei W, Su J, Yu XF. HIV-1 Vpu and SARS-CoV-2 ORF3a proteins disrupt STING-mediated activation of antiviral NF-κB signaling. Sci Signal 2025; 18:eadd6593. [PMID: 39836751 DOI: 10.1126/scisignal.add6593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/12/2023] [Accepted: 12/19/2024] [Indexed: 01/30/2025]
Abstract
Activation of the stimulator of interferon genes (STING) pathway by cytosolic DNA leads to the activation of the transcription factors interferon regulatory factor 3 (IRF3) and nuclear factor κB (NF-κB). Although many viruses produce proteins that inhibit IRF3-dependent antiviral responses, some viruses produce proteins that inhibit STING-induced NF-κB activation without blocking IRF3 activation. Here, we found that STING-activated, NF-κB-dependent, and IRF3-independent innate immunity inhibited the replication of the DNA virus herpes simplex virus type 1 (HSV-1), the RNA virus coxsackievirus A16 (CV-A16), and the retrovirus HIV-1. The HIV-1 nonstructural protein Vpu bound to STING and prevented it from interacting with the upstream NF-κB pathway kinase inhibitor of NF-κB subunit β (IKKβ), thus blocking NF-κB signaling. This function of Vpu was conserved among Vpu proteins from diverse HIV-1 and simian immunodeficiency virus strains and was distinct from its action in disrupting other host antiviral pathways. Furthermore, the ORF3a protein from the coronavirus SARS-CoV-2 also promoted viral replication by interacting with STING and blocking STING-induced activity of NF-κB but not of IRF3. These findings demonstrate that diverse viral proteins have convergently evolved to selectively inhibit NF-κB-mediated innate immunity downstream of STING activation, suggesting that targeting this pathway may represent a promising antiviral strategy.
Collapse
Affiliation(s)
- Yajuan Rui
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
- Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310000, China
| | - Si Shen
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
- Department of Respiratory Disease, Thoracic Disease Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yanpu Wang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
- Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310000, China
| | - Leyi Cheng
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Shiqi Chen
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Ying Hu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Yong Cai
- School of Life Science, Jilin University, Changchun 130012, China
| | - Wei Wei
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Translational Medicine and Institute of Virology and AIDS Research, First Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Jiaming Su
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
- Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310000, China
| | - Xiao-Fang Yu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
- Cancer Center of Zhejiang University, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
10
|
Wang H, Cui Z, Sun W, Yi M, Cheng Y, Zhang Y, Du Y, Pan T, Gao R, Feng L, Zeng B, Huang G, Li Y, Wang Y, Zhang CJ, He R, Wang C. MYO1F positions cGAS on the plasma membrane to ensure full and functional signaling. Mol Cell 2025; 85:150-165.e7. [PMID: 39694035 DOI: 10.1016/j.molcel.2024.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/24/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024]
Abstract
Cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) synthase (cGAS) detects viral or endogenous DNA, activating the innate immune response to infections and autoimmune diseases. Upon binding to double-stranded DNA, cGAS synthesizes 2'3' cGMP-AMP, which triggers type I interferon production. Besides its presence in the cytosol and nucleus, cGAS is found at the plasma membrane, although its significance remains unclear. Here, we report that cGAS associates with myosin 1F (MYO1F) at the plasma membrane of human and mouse macrophages. During viral infection, phosphorylation of MYO1F by spleen-associated tyrosine kinase (SYK) facilitates the recruitment of lysine acetyltransferase 2A (KAT2A), which acetylates cGAS at lysine residues 421, 292, and 131, essential for its activation. Moreover, membrane-localized cGAS is crucial for signaling activation and type I interferon production triggered by virus-cell fusion due to Mn2+ release from organelles. Our results highlight the importance of MYO1F-mediated cGAS localization for its full activation in response to viral infection.
Collapse
Affiliation(s)
- Heping Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China; The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 611731 Chengdu, China
| | - Zhihui Cui
- Key Laboratory of Molecular Biophysics of the Ministry of Education, National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China; The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 611731 Chengdu, China
| | - Wanwei Sun
- Key Laboratory of Molecular Biophysics of the Ministry of Education, National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China; The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 611731 Chengdu, China
| | - Ming Yi
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 611731 Chengdu, China; Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, 611731 Chengdu, China; Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China
| | - Yuheng Cheng
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yunpeng Zhang
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yanyun Du
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 611731 Chengdu, China; Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, 611731 Chengdu, China; Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China
| | - Ting Pan
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China
| | - Ru Gao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, 430074 Wuhan, China; The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 611731 Chengdu, China
| | - Lingyun Feng
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 611731 Chengdu, China; Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, 611731 Chengdu, China; Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China
| | - Bo Zeng
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 611731 Chengdu, China; Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, 611731 Chengdu, China; Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China
| | - Guoling Huang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China
| | - Yangyang Li
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 611731 Chengdu, China; Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, 611731 Chengdu, China; Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China
| | - Yuan Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China
| | - Cun-Jin Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China; Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072 Chengdu, China
| | - Ruirui He
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 611731 Chengdu, China; Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, 611731 Chengdu, China; Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China.
| | - Chenhui Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 611731 Chengdu, China; Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, 611731 Chengdu, China; Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, China.
| |
Collapse
|
11
|
Zhao X, Wei T, Hou Y, Wu Y, Zhou H, Meng J, Wang Q, Liu Y. ATM/ATR-Mediated DNA Damage Response Facilitates SARS-CoV-2 Spike Protein-Induced Syncytium Formation. J Med Virol 2025; 97:e70137. [PMID: 39740081 DOI: 10.1002/jmv.70137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 01/02/2025]
Abstract
Multinucleated cells are present in lung tissues of patients infected by SARS-CoV-2. Although the spike protein can cause the fusion of infected cells and ACE2-expressing cells to form syncytia and induce damage, how host cell responses to this damage and the role of DNA damage response (DDR) signals in cell fusion are still unclear. Therefore, we investigated the effect of SARS-CoV-2 spike protein on the fusion of homologous and heterologous cells expressing ACE2 in vitro models, focusing on the protein levels of ATR and ATM, the major kinases responding to DNA damage, and their substrates CHK1 and CHK2. We found that both homologous and heterologous cell fusion activated the ATR-CHK1 and ATM-CHK2 signaling axis and induced the aggregation of γH2AX, 53BP1 and RAD51 in syncytia. In addition, siRNA or inhibitors of ATM and ATR suppressed syncytia formation by decreasing the level of S protein. These results showed the important role of DDR in stabilizing the S protein and in favoring its induction of cell fusion and syncytium formation, suggesting that the virus exploits the host DDR to facilitate its spread among infected cells.
Collapse
Affiliation(s)
- Xiaotong Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Tingting Wei
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yujia Hou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yanjin Wu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Haitao Zhou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiahui Meng
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Qin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
12
|
Fan H, Tian M, Liu S, Ye C, Li Z, Wu K, Zhu C. Strategies Used by SARS-CoV-2 to Evade the Innate Immune System in an Evolutionary Perspective. Pathogens 2024; 13:1117. [PMID: 39770376 PMCID: PMC11677916 DOI: 10.3390/pathogens13121117] [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: 11/24/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/05/2025] Open
Abstract
By the end of 2019, the COVID-19 pandemic, resulting from the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), had diffused widely across the globe, with 770 million infected individuals and over 7 million deaths reported. In addition to its high infectivity and pathogenicity and its rapid mutation rate, the unique capacity of SARS-CoV-2 to circumvent the immune system has also contributed to the widespread nature of this pandemic. SARS-CoV-2 elicits the onset of innate immune system activation and initiates antiviral responses once it has infected the host. While battling the host's immune responses, SARS-CoV-2 has established many countermeasures to evade attack and clearance. As the exploration of SARS-CoV-2 continues, substantial evidence has revealed that the 29 proteins synthesized by the SARS-CoV-2 genome are integral to the viral infection process. They not only facilitate viral replication and transmission, but also assist SARS-CoV-2 in escaping the host's immune defenses, positioning them as promising therapeutic targets that have attracted considerable attention in recent studies. This review summarizes the manner in which SARS-CoV-2 interfaces with the innate immune system, with a particular focus on the continuous evolution of SARS-CoV-2 and the implications of mutations.
Collapse
Affiliation(s)
- Hong Fan
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China; (H.F.); (C.Y.); (Z.L.)
| | - Mingfu Tian
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (M.T.); (S.L.); (K.W.)
| | - Siyu Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (M.T.); (S.L.); (K.W.)
| | - Chenglin Ye
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China; (H.F.); (C.Y.); (Z.L.)
| | - Zhiqiang Li
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China; (H.F.); (C.Y.); (Z.L.)
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; (M.T.); (S.L.); (K.W.)
| | - Chengliang Zhu
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China; (H.F.); (C.Y.); (Z.L.)
| |
Collapse
|
13
|
An Y, Wang C, Wang Z, Kong F, Liu H, Jiang M, Liu T, Zhang S, Du K, Yin L, Jiao P, Li Y, Fan B, Zhou C, Wang M, Sun H, Lei J, Zhao S, Gong Y. Tight junction protein LSR is a host defense factor against SARS-CoV-2 infection in the small intestine. EMBO J 2024; 43:6124-6151. [PMID: 39443717 PMCID: PMC11612383 DOI: 10.1038/s44318-024-00281-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 09/29/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024] Open
Abstract
The identification of host factors with antiviral potential is important for developing effective prevention and therapeutic strategies against SARS-CoV-2 infection. Here, by using immortalized cell lines, intestinal organoids, ex vivo intestinal tissues and humanized ACE2 mouse model as proof-of-principle systems, we have identified lipolysis-stimulated lipoprotein receptor (LSR) as a crucial host defense factor against SARS-CoV-2 infection in the small intestine. Loss of endogenous LSR enhances ACE2-dependent infection by SARS-CoV-2 Spike (S) protein-pseudotyped virus and authentic SARS-CoV-2 virus, and exogenous administration of LSR protects against viral infection. Mechanistically, LSR interacts with ACE2 both in cis and in trans, preventing its binding to S protein, and thus inhibiting viral entry and S protein-mediated cell-cell fusion. Finally, a small LSR-derived peptide blocks S protein binding to the ACE2 receptor in vitro. These results identify both a previously unknown function for LSR in antiviral host defense against SARS-CoV-2, with potential implications for peptide-based pan-variant therapeutic interventions.
Collapse
Affiliation(s)
- Yanan An
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Laboratory of Tight Junction, Binzhou Medical University, Yantai, Shandong, China
| | - Chao Wang
- Department of Urology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ziqi Wang
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Laboratory of Tight Junction, Binzhou Medical University, Yantai, Shandong, China
| | - Feng Kong
- Shandong Provincial Engineering Laboratory of Urologic Tissue Reconstruction, Jinan, Shandong, China
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hao Liu
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Laboratory of Tight Junction, Binzhou Medical University, Yantai, Shandong, China
| | - Min Jiang
- Department of Pharmacology, Binzhou Medical University, Yantai, Shandong, China
| | - Ti Liu
- Shandong Provincial Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Shu Zhang
- Shandong Provincial Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Kaige Du
- Shandong Provincial Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Liang Yin
- Shandong Provincial Engineering Laboratory of Urologic Tissue Reconstruction, Jinan, Shandong, China
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Peng Jiao
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Urology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ying Li
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Laboratory of Tight Junction, Binzhou Medical University, Yantai, Shandong, China
| | - Baozhen Fan
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Chengjun Zhou
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Mingxia Wang
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Laboratory of Tight Junction, Binzhou Medical University, Yantai, Shandong, China
| | - Hui Sun
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China
- Laboratory of Tight Junction, Binzhou Medical University, Yantai, Shandong, China
| | - Jie Lei
- Shandong Provincial Center for Disease Control and Prevention, Jinan, Shandong, China.
| | - Shengtian Zhao
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
- Shandong Provincial Engineering Laboratory of Urologic Tissue Reconstruction, Jinan, Shandong, China.
- Department of Urology, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Yongfeng Gong
- Department of Physiology, Binzhou Medical University, Yantai, Shandong, China.
- Shandong Engineering Research Center of Molecular Medicine for Renal Diseases, Yantai, Shandong, China.
- Laboratory of Tight Junction, Binzhou Medical University, Yantai, Shandong, China.
| |
Collapse
|
14
|
Lee JH, Sergi C, Kast RE, Kanwar BA, Bourbeau J, Oh S, Sohn MG, Lee CJ, Coleman MD. Aggravating mechanisms from COVID-19. Virol J 2024; 21:228. [PMID: 39334442 PMCID: PMC11430051 DOI: 10.1186/s12985-024-02506-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces immune-mediated diseases. The pathophysiology of COVID-19 uses the following three mechanisms: (1) inflammasome activation mechanism; (2) cGAS-STING signaling mechanism; and (3) SAMHD1 tetramerization mechanism, which leads to IFN-I production. Interactions between the host and virus govern induction, resulting in multiorgan impacts. The NLRP3 with cGAS-STING constitutes the primary immune response. The expression of SARS-CoV-2 ORF3a, NSP6, NSP7, and NSP8 blocks innate immune activation and facilitates virus replication by targeting the RIG-I/MDA5, TRIF, and cGAS-STING signaling. SAMHD1 has a target motif for CDK1 to protect virion assembly, threonine 592 to modulate a catalytically active tetramer, and antiviral IFN responses to block retroviral infection. Plastic and allosteric nucleic acid binding of SAMHD1 modulates the antiretroviral activity of SAMHD1. Therefore, inflammasome activation, cGAS-STING signaling, and SAMHD1 tetramerization explain acute kidney injury, hepatic, cardiac, neurological, and gastrointestinal injury of COVID-19. It might be necessary to effectively block the pathological courses of diverse diseases.
Collapse
Affiliation(s)
- Jong Hoon Lee
- Science and Research Center, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Geriatrics, Gyeonggi Medical Center Pocheon Hospital, 1648 Pocheon-ro Sin-eup-dong, Pocheon-si, Gyeonggi-do, 11142, Republic of Korea.
| | - Consolato Sergi
- Division of Anatomical Pathology, Children's Hospital of Eastern Ontario (CHEO), University of Ottawa, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Richard E Kast
- IIAIGC Study Center, 11 Arlington Ct, Burlington, 05408 VT, USA
| | - Badar A Kanwar
- Haider Associates, 1999 Forest Ridge Dr, Bedford, TX, 76021, USA
| | - Jean Bourbeau
- Respiratory Epidemiology and Clinical Research Unit, McGill University Health Centre, Montréal, QC, Canada
| | - Sangsuk Oh
- Department of Food Engineering, Food Safety Laboratory, Memory Unit, Ewha Womans University, Seoul, 03670, Korea
| | - Mun-Gi Sohn
- Department of Food Science, KyungHee University College of Life Science, Seoul, 17104, Republic of Korea
| | - Chul Joong Lee
- Department of Anesthesiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Michael D Coleman
- College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
15
|
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
|
16
|
Zhou X, Wang J, Yu L, Qiao G, Qin D, Yuen-Kwan Law B, Ren F, Wu J, Wu A. Mitophagy and cGAS-STING crosstalk in neuroinflammation. Acta Pharm Sin B 2024; 14:3327-3361. [PMID: 39220869 PMCID: PMC11365416 DOI: 10.1016/j.apsb.2024.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 09/04/2024] Open
Abstract
Mitophagy, essential for mitochondrial health, selectively degrades damaged mitochondria. It is intricately linked to the cGAS-STING pathway, which is crucial for innate immunity. This pathway responds to mitochondrial DNA and is associated with cellular stress response. Our review explores the molecular details and regulatory mechanisms of mitophagy and the cGAS-STING pathway. We critically evaluate the literature demonstrating how dysfunctional mitophagy leads to neuroinflammatory conditions, primarily through the accumulation of damaged mitochondria, which activates the cGAS-STING pathway. This activation prompts the production of pro-inflammatory cytokines, exacerbating neuroinflammation. This review emphasizes the interaction between mitophagy and the cGAS-STING pathways. Effective mitophagy may suppress the cGAS-STING pathway, offering protection against neuroinflammation. Conversely, impaired mitophagy may activate the cGAS-STING pathway, leading to chronic neuroinflammation. Additionally, we explored how this interaction influences neurodegenerative disorders, suggesting a common mechanism underlying these diseases. In conclusion, there is a need for additional targeted research to unravel the complexities of mitophagy-cGAS-STING interactions and their role in neurodegeneration. This review highlights potential therapies targeting these pathways, potentially leading to new treatments for neuroinflammatory and neurodegenerative conditions. This synthesis enhances our understanding of the cellular and molecular foundations of neuroinflammation and opens new therapeutic avenues for neurodegenerative disease research.
Collapse
Affiliation(s)
- Xiaogang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Jing Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Gan Qiao
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Dalian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China
| | - Fang Ren
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, China
| | - Jianming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Anguo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
17
|
Li H, Wan L, Liu M, Ma E, Huang L, Yang Y, Li Q, Fang Y, Li J, Han B, Zhang C, Sun L, Hou X, Li H, Sun M, Qian S, Duan X, Zhao R, Yang X, Chen Y, Wu S, Zhang X, Zhang Y, Cheng G, Chen G, Gao Q, Xu J, Hou L, Wei C, Zhong H. SARS-CoV-2 spike-induced syncytia are senescent and contribute to exacerbated heart failure. PLoS Pathog 2024; 20:e1012291. [PMID: 39102426 PMCID: PMC11326701 DOI: 10.1371/journal.ppat.1012291] [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: 10/18/2023] [Revised: 08/15/2024] [Accepted: 05/27/2024] [Indexed: 08/07/2024] Open
Abstract
SARS-CoV-2 spike protein (SARS-2-S) induced cell-cell fusion in uninfected cells may occur in long COVID-19 syndrome, as circulating SARS-2-S or extracellular vesicles containing SARS-2-S (S-EVs) were found to be prevalent in post-acute sequelae of COVID-19 (PASC) for up to 12 months after diagnosis. Although isolated recombinant SARS-2-S protein has been shown to increase the SASP in senescent ACE2-expressing cells, the direct linkage of SARS-2-S syncytia with senescence in the absence of virus infection and the degree to which SARS-2-S syncytia affect pathology in the setting of cardiac dysfunction are unknown. Here, we found that the senescent outcome of SARS-2-S induced syncytia exacerbated heart failure progression. We first demonstrated that syncytium formation in cells expressing SARS-2-S delivered by DNA plasmid or LNP-mRNA exhibits a senescence-like phenotype. Extracellular vesicles containing SARS-2-S (S-EVs) also confer a potent ability to form senescent syncytia without de novo synthesis of SARS-2-S. However, it is important to note that currently approved COVID-19 mRNA vaccines do not induce syncytium formation or cellular senescence. Mechanistically, SARS-2-S syncytia provoke the formation of functional MAVS aggregates, which regulate the senescence fate of SARS-2-S syncytia by TNFα. We further demonstrate that senescent SARS-2-S syncytia exhibit shrinked morphology, leading to the activation of WNK1 and impaired cardiac metabolism. In pre-existing heart failure mice, the WNK1 inhibitor WNK463, anti-syncytial drug niclosamide, and senolytic dasatinib protect the heart from exacerbated heart failure triggered by SARS-2-S. Our findings thus suggest a potential mechanism for COVID-19-mediated cardiac pathology and recommend the application of WNK1 inhibitor for therapy especially in individuals with post-acute sequelae of COVID-19.
Collapse
Affiliation(s)
- Huilong Li
- Beijing Institute of Biotechnology, Beijing, China
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Luming Wan
- Beijing Institute of Biotechnology, Beijing, China
| | - Muyi Liu
- Beijing Institute of Biotechnology, Beijing, China
| | - Enhao Ma
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Linfei Huang
- Beijing Institute of Biotechnology, Beijing, China
| | - Yilong Yang
- Beijing Institute of Biotechnology, Beijing, China
| | - Qihong Li
- The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yi Fang
- The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jingfei Li
- Beijing Institute of Biotechnology, Beijing, China
| | - Bingqing Han
- Beijing Institute of Biotechnology, Beijing, China
| | - Chang Zhang
- Beijing Institute of Biotechnology, Beijing, China
| | - Lijuan Sun
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Xufeng Hou
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Haiyang Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mingyu Sun
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Sichong Qian
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xuejing Duan
- Department of Pathology, Fuwai Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, China
| | - Ruzhou Zhao
- Beijing Institute of Biotechnology, Beijing, China
| | - Xiaopan Yang
- Beijing Institute of Biotechnology, Beijing, China
| | - Yi Chen
- Beijing Institute of Biotechnology, Beijing, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Beijing, China
| | - Xuhui Zhang
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | | | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Gengye Chen
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Qi Gao
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Junjie Xu
- Beijing Institute of Biotechnology, Beijing, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Beijing, China
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Congwen Wei
- Beijing Institute of Biotechnology, Beijing, China
| | - Hui Zhong
- Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
18
|
Lee JH, Sergi C, Kast RE, Kanwar BA, Bourbeau J, Oh S, Sohn MG, Lee CJ, Coleman MD. Basic implications on three pathways associated with SARS-CoV-2. Biomed J 2024:100766. [PMID: 39004185 DOI: 10.1016/j.bj.2024.100766] [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: 05/21/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) interacts between the host and virus and govern induction, resulting in multiorgan impacts. Its pathophysiology involves the followings: 1) the angiotensin-converting enzyme (ACE2) and Toll-like receptor (TLR) pathways: 2) the neuropilin (NRP) pathway: 3) the spike protein pathway. Therefore, it is necessary to block the pathological course with modulating innate lymphoid cells against diverse corona variants in the future.
Collapse
Affiliation(s)
- Jong Hoon Lee
- Science and Research Center, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Consolato Sergi
- Division of Anatomical Pathology, Children's Hospital of Eastern Ontario (CHEO), University of Ottawa, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Richard E Kast
- IIAIGC Study Center, 11 Arlington Ct, Burlington, 05408, VT, USA
| | - Badar A Kanwar
- Haider Associates, 1999 Forest Ridge Dr, Bedford, TX, 76021, USA
| | - Jean Bourbeau
- Respiratory Epidemiology and Clinical Research Unit, McGill University Health Centre, Montréal, QC, Canada
| | - Sangsuk Oh
- Department of Food Engineering, Food Safety Laboratory, Memory Unit, Ewha Womans University, Seoul, 03670, Republic of Korea
| | - Mun-Gi Sohn
- Department of Food Science, KyungHee University College of Life Science, Seoul, 17104, Republic of Korea
| | - Chul Joong Lee
- Department of Anesthesiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Michael D Coleman
- College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
19
|
Su L, Yu T, Zhang C, Huo P, Zhao Z. A prediction model for secondary invasive fungal infection among severe SARS-CoV-2 positive patients in ICU. Front Cell Infect Microbiol 2024; 14:1382720. [PMID: 39040601 PMCID: PMC11260608 DOI: 10.3389/fcimb.2024.1382720] [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: 04/04/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Background The global COVID-19 pandemic has resulted in over seven million deaths, and IFI can further complicate the clinical course of COVID-19. Coinfection of COVID-19 and IFI (secondary IFI) pose significant threats not only to healthcare systems but also to patient lives. After the control measures for COVID-19 were lifted in China, we observed a substantial number of ICU patients developing COVID-19-associated IFI. This creates an urgent need for predictive assessment of COVID-19 patients in the ICU environment for early detection of suspected fungal infection cases. Methods This study is a single-center, retrospective research endeavor. We conducted a case-control study on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) positive patients. The cases consisted of patients who developed any secondary IFI during their ICU stay at Jilin University China-Japan Union Hospital in Changchun, Jilin Province, China, from December 1st, 2022, to August 31st, 2023. The control group consisted of SARS-CoV-2 positive patients without secondary IFI. Descriptive and comparative analyses were performed, and a logistic regression prediction model for secondary IFI in COVID-19 patients was established. Additionally, we observed an increased incidence of COVID-19-associated pulmonary aspergillosis (CAPA) during this pandemic. Therefore, we conducted a univariate subgroup analysis on top of IFI, using non-CAPA patients as the control subgroup. Results From multivariate analysis, the prediction model identified 6 factors that are significantly associated with IFI, including the use of broad-spectrum antibiotics for more than 2 weeks (aOR=4.14, 95% CI 2.03-8.67), fever (aOR=2.3, 95%CI 1.16-4.55), elevated log IL-6 levels (aOR=1.22, 95% CI 1.04-1.43) and prone position ventilation (aOR=2.38, 95%CI 1.15-4.97) as independent risk factors for COVID-19 secondary IFI. High BMI (BMI ≥ 28 kg/m2) (aOR=0.85, 95% CI 0.75-0.94) and the use of COVID-19 immunoglobulin (aOR=0.45, 95% CI 0.2-0.97) were identified as independent protective factors against COVID-19 secondary IFI. The Receiver Operating Curve (ROC) area under the curve (AUC) of this model was 0.81, indicating good classification. Conclusion We recommend paying special attention for the occurrence of secondary IFI in COVID-19 patients with low BMI (BMI < 28 kg/m2), elevated log IL-6 levels and fever. Additionally, during the treatment of COVID-19 patients, we emphasize the importance of minimizing the duration of broad-spectrum antibiotic use and highlight the potential of immunoglobulin application in reducing the incidence of IFI.
Collapse
Affiliation(s)
- Leilei Su
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Tong Yu
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, United States
| | - Chunmei Zhang
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Pengfei Huo
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhongyan Zhao
- Department of Critical Care Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Schäfer TE, Knol LI, Haas FV, Hartley A, Pernickel SCS, Jády A, Finkbeiner MSC, Achberger J, Arelaki S, Modic Ž, Schröer K, Zhang W, Schmidt B, Schuster P, Haferkamp S, Doerner J, Gebauer F, Ackermann M, Kvasnicka HM, Kulkarni A, Bots STF, Kemp V, Hawinkels LJAC, Poetsch AR, Hoeben RC, Ehrhardt A, Marchini A, Ungerechts G, Ball CR, Engeland CE. Biomarker screen for efficacy of oncolytic virotherapy in patient-derived pancreatic cancer cultures. EBioMedicine 2024; 105:105219. [PMID: 38941955 PMCID: PMC11260584 DOI: 10.1016/j.ebiom.2024.105219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/30/2024] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a tumour entity with unmet medical need. To assess the therapeutic potential of oncolytic virotherapy (OVT) against PDAC, different oncolytic viruses (OVs) are currently investigated in clinical trials. However, systematic comparisons of these different OVs in terms of efficacy against PDAC and biomarkers predicting therapeutic response are lacking. METHODS We screened fourteen patient-derived PDAC cultures which reflect the intra- and intertumoural heterogeneity of PDAC for their sensitivity to five clinically relevant OVs, namely serotype 5 adenovirus Ad5-hTERT, herpes virus T-VEC, measles vaccine strain MV-NIS, reovirus jin-3, and protoparvovirus H-1PV. Live cell analysis, quantification of viral genome/gene expression, cell viability as well as cytotoxicity assays and titration of viral progeny were conducted. Transcriptome profiling was employed to identify potential predictive biomarkers for response to OV treatment. FINDINGS Patient-derived PDAC cultures showed individual response patterns to OV treatment. Twelve of fourteen cultures were responsive to at least one OV, with no single OV proving superior or inferior across all cultures. Known host factors for distinct viruses were retrieved as potential biomarkers. Compared to the classical molecular subtype, the quasi-mesenchymal or basal-like subtype of PDAC was found to be more sensitive to H-1PV, jin-3, and T-VEC. Generally, expression of viral entry receptors did not correlate with sensitivity to OV treatment, with one exception: Expression of Galectin-1 (LGALS1), a factor involved in H-1PV entry, positively correlated with H-1PV induced cell killing. Rather, cellular pathways controlling immunological, metabolic and proliferative signaling appeared to determine outcome. For instance, high baseline expression of interferon-stimulated genes (ISGs) correlated with relative resistance to oncolytic measles virus, whereas low cyclic GMP-AMP synthase (cGAS) expression was associated with exceptional response. Combination treatment of MV-NIS with a cGAS inhibitor improved tumour cell killing in several PDAC cultures and cells overexpressing cGAS were found to be less sensitive to MV oncolysis. INTERPRETATION Considering the heterogeneity of PDAC and the complexity of biological therapies such as OVs, no single biomarker can explain the spectrum of response patterns. For selection of a particular OV, PDAC molecular subtype, ISG expression as well as activation of distinct signaling and metabolic pathways should be considered. Combination therapies can overcome resistance in specific constellations. Overall, oncolytic virotherapy is a viable treatment option for PDAC, which warrants further development. This study highlights the need for personalised treatment in OVT. By providing all primary data, this study provides a rich source and guidance for ongoing developments. FUNDING German National Science Foundation (Deutsche Forschungsgemeinschaft, DFG), German Cancer Aid (Deutsche Krebshilfe), German National Academic Scholarship Foundation (Studienstiftung des deutschen Volkes), Survival with Pancreatic Cancer Foundation.
Collapse
Affiliation(s)
- Theresa E Schäfer
- Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany; Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Lisanne I Knol
- Department for Translational Medical Oncology, National Center for Tumor Diseases Dresden (NCT/UCC), A Partnership Between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Ferdinand V Haas
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Witten, Germany
| | - Anna Hartley
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center (DKFZ), Heidelberg, Germany; DNA Vector Laboratory, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sophie C S Pernickel
- Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany; Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Attila Jády
- Department for Translational Medical Oncology, National Center for Tumor Diseases Dresden (NCT/UCC), A Partnership Between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Maximiliane S C Finkbeiner
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Witten, Germany
| | - Johannes Achberger
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Witten, Germany; Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stella Arelaki
- German Cancer Research Center (DKFZ) Heidelberg, Translational Functional Cancer Genomics, Germany
| | - Živa Modic
- Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katrin Schröer
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Witten, Germany
| | - Wenli Zhang
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Witten, Germany
| | - Barbara Schmidt
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany; Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Philipp Schuster
- Institute of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - Johannes Doerner
- Department of Surgery, Helios University Hospital Wuppertal, Wuppertal, Germany
| | - Florian Gebauer
- Department of Surgery, Helios University Hospital Wuppertal, Wuppertal, Germany
| | - Maximilian Ackermann
- Institute of Pathology and Molecular Pathology, Helios University Clinic Wuppertal, Witten/Herdecke University, Witten, Germany; Institute of Pathology, RWTH University Clinics University Aachen, Aachen, Germany
| | - Hans-Michael Kvasnicka
- Institute of Pathology and Molecular Pathology, Helios University Clinic Wuppertal, Witten/Herdecke University, Witten, Germany
| | - Amit Kulkarni
- Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, Luxembourg
| | - Selas T F Bots
- Virus and Cell Biology Lab, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Vera Kemp
- Virus and Cell Biology Lab, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Lukas J A C Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anna R Poetsch
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rob C Hoeben
- Virus and Cell Biology Lab, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anja Ehrhardt
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Witten, Germany
| | - Antonio Marchini
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center (DKFZ), Heidelberg, Germany; Laboratory of Oncolytic Virus Immuno-Therapeutics, Luxembourg Institute of Health, Luxembourg
| | - Guy Ungerechts
- Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Medical Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Claudia R Ball
- Department for Translational Medical Oncology, National Center for Tumor Diseases Dresden (NCT/UCC), A Partnership Between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Germany; Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; German Cancer Research Center (DKFZ) Heidelberg, Translational Functional Cancer Genomics, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, Germany; Faculty of Biology, TUD Dresden University of Technology, Germany
| | - Christine E Engeland
- Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany; Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), Witten/Herdecke University, Witten, Germany; Experimental Hematology and Immunotherapy, Department of Hematology, Hemostaseology, Cellular Therapy and Infectious Diseases, Faculty of Medicine and Leipzig University Hospital, Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany.
| |
Collapse
|
21
|
Nian Z, Mao Y, Xu Z, Deng M, Xu Y, Xu H, Chen R, Xu Y, Huang N, Mao F, Xu C, Wang Y, Niu M, Chen A, Xue X, Zhang H, Guo G. Multi-omics analysis uncovered systemic lupus erythematosus and COVID-19 crosstalk. Mol Med 2024; 30:81. [PMID: 38862942 PMCID: PMC11167821 DOI: 10.1186/s10020-024-00851-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Studies have highlighted a possible crosstalk between the pathogeneses of COVID-19 and systemic lupus erythematosus (SLE); however, the interactive mechanisms remain unclear. We aimed to elucidate the impact of COVID-19 on SLE using clinical information and the underlying mechanisms of both diseases. METHODS RNA-seq datasets were used to identify shared hub gene signatures between COVID-19 and SLE, while genome-wide association study datasets were used to delineate the interaction mechanisms of the key signaling pathways. Finally, single-cell RNA-seq datasets were used to determine the primary target cells expressing the shared hub genes and key signaling pathways. RESULTS COVID-19 may affect patients with SLE through hematologic involvement and exacerbated inflammatory responses. We identified 14 shared hub genes between COVID-19 and SLE that were significantly associated with interferon (IFN)-I/II. We also screened and obtained four core transcription factors related to these hub genes, confirming the regulatory role of the IFN-I/II-mediated Janus kinase/signal transducers and activators of transcription (JAK-STAT) signaling pathway on these hub genes. Further, SLE and COVID-19 can interact via IFN-I/II and IFN-I/II receptors, promoting the levels of monokines, including interleukin (IL)-6/10, tumor necrosis factor-α, and IFN-γ, and elevating the incidence rate and risk of cytokine release syndrome. Therefore, in SLE and COVID-19, both hub genes and core TFs are enriched within monocytes/macrophages. CONCLUSIONS The interaction between SLE and COVID-19 promotes the activation of the IFN-I/II-triggered JAK-STAT signaling pathway in monocytes/macrophages. These findings provide a new direction and rationale for diagnosing and treating patients with SLE-COVID-19 comorbidity.
Collapse
Affiliation(s)
- Zekai Nian
- Second Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Yicheng Mao
- Ophthalmology College, Wenzhou Medical University, Wenzhou, China
| | - Zexia Xu
- Department of Nephrology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ming Deng
- Public Health and Management College, Wenzhou Medical University, Wenzhou, China
| | - Yixi Xu
- School of Public Administration, Hangzhou Normal University, Hangzhou, China
| | - Hanlu Xu
- Ophthalmology College, Wenzhou Medical University, Wenzhou, China
| | - Ruoyao Chen
- Second Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Yiliu Xu
- Research Center of Fluid Machinery Engineering and Technology, Jiangsu University, Zhenjiang, China
| | - Nan Huang
- Public Health and Management College, Wenzhou Medical University, Wenzhou, China
| | - Feiyang Mao
- Second Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Chenyu Xu
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yulin Wang
- Public Health and Management College, Wenzhou Medical University, Wenzhou, China
| | - Mengyuan Niu
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Aqiong Chen
- Department of Rheumatology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Xiangyang Xue
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Huidi Zhang
- Department of Nephrology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Gangqiang Guo
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
22
|
Rezabakhsh A, Sadaie MR, Ala A, Roosta Y, Habtemariam S, Sahebnasagh A, Khezri MR. STING agonists as promising vaccine adjuvants to boost immunogenicity against SARS-related coronavirus derived infection: possible role of autophagy. Cell Commun Signal 2024; 22:305. [PMID: 38831299 PMCID: PMC11145937 DOI: 10.1186/s12964-024-01680-0] [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: 01/23/2024] [Accepted: 05/26/2024] [Indexed: 06/05/2024] Open
Abstract
As a major component of innate immunity and a positive regulator of interferons, the Stimulator of interferon gene (STING) has an immunotherapy potential to govern a variety of infectious diseases. Despite the recent advances regarding vaccines against COVID-19, nontoxic novel adjuvants with the potential to enhance vaccine efficacy are urgently desired. In this connection, it has been well-documented that STING agonists are applied to combat COVID-19. This approach is of major significance for boosting immune responses most likely through an autophagy-dependent manner in susceptible individuals against infection induced by severe acute respiratory syndrome Coronavirus (SARS‑CoV‑2). Given that STING agonists exert substantial immunomodulatory impacts under a wide array of pathologic conditions, these agents could be considered novel adjuvants for enhancing immunogenicity against the SARS-related coronavirus. Here, we intend to discuss the recent advances in STING agonists' recruitment to boost innate immune responses upon vaccination against SARS-related coronavirus infections. In light of the primordial role of autophagy modulation, the potential of being an antiviral vaccine adjuvant was also explored.
Collapse
Affiliation(s)
- Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - M Reza Sadaie
- NovoMed Consulting, Biomedical Sciences, Germantown, Maryland, USA
| | - Alireza Ala
- Emergency and Trauma Care Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Roosta
- Hematology, Immune Cell Therapy, and Stem Cells Transplantation Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Solomon Habtemariam
- Pharmacognosy Research and Herbal Analysis Services UK, University of Greenwich, Kent, UK
| | - Adeleh Sahebnasagh
- Clinical Research Center, Department of Internal Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mohammad Rafi Khezri
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, 5715799313, Iran.
| |
Collapse
|
23
|
Malireddi RKS, Sharma BR, Kanneganti TD. Innate Immunity in Protection and Pathogenesis During Coronavirus Infections and COVID-19. Annu Rev Immunol 2024; 42:615-645. [PMID: 38941608 PMCID: PMC11373870 DOI: 10.1146/annurev-immunol-083122-043545] [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: 06/30/2024]
Abstract
The COVID-19 pandemic was caused by the recently emerged β-coronavirus SARS-CoV-2. SARS-CoV-2 has had a catastrophic impact, resulting in nearly 7 million fatalities worldwide to date. The innate immune system is the first line of defense against infections, including the detection and response to SARS-CoV-2. Here, we discuss the innate immune mechanisms that sense coronaviruses, with a focus on SARS-CoV-2 infection and how these protective responses can become detrimental in severe cases of COVID-19, contributing to cytokine storm, inflammation, long-COVID, and other complications. We also highlight the complex cross talk among cytokines and the cellular components of the innate immune system, which can aid in viral clearance but also contribute to inflammatory cell death, cytokine storm, and organ damage in severe COVID-19 pathogenesis. Furthermore, we discuss how SARS-CoV-2 evades key protective innate immune mechanisms to enhance its virulence and pathogenicity, as well as how innate immunity can be therapeutically targeted as part of the vaccination and treatment strategy. Overall, we highlight how a comprehensive understanding of innate immune mechanisms has been crucial in the fight against SARS-CoV-2 infections and the development of novel host-directed immunotherapeutic strategies for various diseases.
Collapse
Affiliation(s)
- R K Subbarao Malireddi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Bhesh Raj Sharma
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | | |
Collapse
|
24
|
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
|
25
|
Xie F, Zhu Q. The regulation of cGAS-STING signaling by RNA virus-derived components. Virol J 2024; 21:101. [PMID: 38693578 PMCID: PMC11064393 DOI: 10.1186/s12985-024-02359-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/04/2024] [Indexed: 05/03/2024] Open
Abstract
The Cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) serves as a key innate immune signaling axis involved in the regulation of various human diseases. It has been found that cGAS-STING pathway can recognize a variety of cytosolic double-stranded DNA (dsDNA), contributing to cause a robust type I interferon response thereby affecting the occurrence and progression of viral infection. Accumulating evidence indicates RNA virus-derived components play an important role in regulating cGAS-STING signaling, either as protective or pathogenic factors in the pathogenesis of diseases. Thus, a comprehensive understanding of the function of RNA virus-derived components in regulating cGAS-STING signaling will provide insights into developing novel therapies. Here, we review the existing literature on cGAS-STING pathway regulated by RNA virus-derived components to propose insights into pharmacologic strategies targeting the cGAS-STING pathway.
Collapse
Affiliation(s)
- Feiting Xie
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, China.
| | - Qiugang Zhu
- Department of Laboratory Medicine, Shangyu People's Hospital of Shaoxing, Shaoxing, China
| |
Collapse
|
26
|
Purwono PB, Vacharathit V, Manopwisedjaroen S, Ludowyke N, Suksatu A, Thitithanyanont A. Infection kinetics, syncytia formation, and inflammatory biomarkers as predictive indicators for the pathogenicity of SARS-CoV-2 Variants of Concern in Calu-3 cells. PLoS One 2024; 19:e0301330. [PMID: 38568894 PMCID: PMC10990222 DOI: 10.1371/journal.pone.0301330] [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: 07/02/2023] [Accepted: 03/13/2024] [Indexed: 04/05/2024] Open
Abstract
The ongoing COVID-19 pandemic has led to the emergence of new SARS-CoV-2 variants as a result of continued host-virus interaction and viral genome mutations. These variants have been associated with varying levels of transmissibility and disease severity. We investigated the phenotypic profiles of six SARS-CoV-2 variants (WT, D614G, Alpha, Beta, Delta, and Omicron) in Calu-3 cells, a human lung epithelial cell line. In our model demonstrated that all variants, except for Omicron, had higher efficiency in virus entry compared to the wild-type. The Delta variant had the greatest phenotypic advantage in terms of early infection kinetics and marked syncytia formation, which could facilitate cell-to-cell spreading, while the Omicron variant displayed slower replication and fewer syncytia formation. We also identified the Delta variant as the strongest inducer of inflammatory biomarkers, including pro-inflammatory cytokines/chemokines (IP-10/CXCL10, TNF-α, and IL-6), anti-inflammatory cytokine (IL-1RA), and growth factors (FGF-2 and VEGF-A), while these inflammatory mediators were not significantly elevated with Omicron infection. These findings are consistent with the observations that there was a generally more pronounced inflammatory response and angiogenesis activity within the lungs of COVID-19 patients as well as more severe symptoms and higher mortality rate during the Delta wave, as compared to less severe symptoms and lower mortality observed during the current Omicron wave in Thailand. Our findings suggest that early infectivity kinetics, enhanced syncytia formation, and specific inflammatory mediator production may serve as predictive indicators for the virulence potential of future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Priyo Budi Purwono
- Faculty of Science, Department of Microbiology, Mahidol University, Bangkok, Thailand
- Faculty of Medicine, Department of Microbiology, Universitas Airlangga, Surabaya, Indonesia
| | - Vimvara Vacharathit
- Faculty of Science, Department of Microbiology, Mahidol University, Bangkok, Thailand
- Faculty of Science, Systems Biology of Diseases Research Unit, Mahidol University, Bangkok, Thailand
| | | | - Natali Ludowyke
- Faculty of Science, Department of Microbiology, Mahidol University, Bangkok, Thailand
| | - Ampa Suksatu
- Faculty of Science, Department of Microbiology, Mahidol University, Bangkok, Thailand
| | - Arunee Thitithanyanont
- Faculty of Science, Department of Microbiology, Mahidol University, Bangkok, Thailand
- Faculty of Science, Department of Microbiology, Pornchai Matangkasombut Center for Microbial Genomics, Mahidol University, Bangkok, Thailand
| |
Collapse
|
27
|
Gewaid H, Bowie AG. Regulation of type I and type III interferon induction in response to pathogen sensing. Curr Opin Immunol 2024; 87:102424. [PMID: 38761566 DOI: 10.1016/j.coi.2024.102424] [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: 03/23/2023] [Revised: 02/19/2024] [Accepted: 05/06/2024] [Indexed: 05/20/2024]
Abstract
Type I and III interferons (IFN-I and IFN-III) have a central role in the early antimicrobial response against invading pathogens. Induction of IFN-Is and IFN-IIIs arises due to the sensing by pattern recognition receptors of pathogen-associated molecular patterns (from micro-organisms) or of damage-associated molecular patterns (DAMPs; produced by host cells). Here, we review recent developments on how IFN-I and IFN-III expression is stimulated by different pathogens and how the signalling pathways leading to IFN induction are tightly regulated. We also summarise the growing knowledge of the sensing pathways that lead to IFN-I and IFN-III induction in response to severe acute respiratory syndrome coronavirus 2.
Collapse
Affiliation(s)
- Hossam Gewaid
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
28
|
Li Q, Wu P, Du Q, Hanif U, Hu H, Li K. cGAS-STING, an important signaling pathway in diseases and their therapy. MedComm (Beijing) 2024; 5:e511. [PMID: 38525112 PMCID: PMC10960729 DOI: 10.1002/mco2.511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/15/2024] [Accepted: 02/21/2024] [Indexed: 03/26/2024] Open
Abstract
Since cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway was discovered in 2013, great progress has been made to elucidate the origin, function, and regulating mechanism of cGAS-STING signaling pathway in the past decade. Meanwhile, the triggering and transduction mechanisms have been continuously illuminated. cGAS-STING plays a key role in human diseases, particularly DNA-triggered inflammatory diseases, making it a potentially effective therapeutic target for inflammation-related diseases. Here, we aim to summarize the ancient origin of the cGAS-STING defense mechanism, as well as the triggers, transduction, and regulating mechanisms of the cGAS-STING. We will also focus on the important roles of cGAS-STING signal under pathological conditions, such as infections, cancers, autoimmune diseases, neurological diseases, and visceral inflammations, and review the progress in drug development targeting cGAS-STING signaling pathway. The main directions and potential obstacles in the regulating mechanism research and therapeutic drug development of the cGAS-STING signaling pathway for inflammatory diseases and cancers will be discussed. These research advancements expand our understanding of cGAS-STING, provide a theoretical basis for further exploration of the roles of cGAS-STING in diseases, and open up new strategies for targeting cGAS-STING as a promising therapeutic intervention in multiple diseases.
Collapse
Affiliation(s)
- Qijie Li
- Sichuan province Medical and Engineering Interdisciplinary Research Center of Nursing & Materials/Nursing Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduSichuanChina
| | - Ping Wu
- Department of Occupational DiseasesThe Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital)ChengduSichuanChina
| | - Qiujing Du
- Sichuan province Medical and Engineering Interdisciplinary Research Center of Nursing & Materials/Nursing Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduSichuanChina
| | - Ullah Hanif
- Sichuan province Medical and Engineering Interdisciplinary Research Center of Nursing & Materials/Nursing Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduSichuanChina
| | - Hongbo Hu
- Center for Immunology and HematologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ka Li
- Sichuan province Medical and Engineering Interdisciplinary Research Center of Nursing & Materials/Nursing Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduSichuanChina
| |
Collapse
|
29
|
Eltayeb A, Al-Sarraj F, Alharbi M, Albiheyri R, Mattar EH, Abu Zeid IM, Bouback TA, Bamagoos A, Uversky VN, Rubio-Casillas A, Redwan EM. Intrinsic factors behind long COVID: IV. Hypothetical roles of the SARS-CoV-2 nucleocapsid protein and its liquid-liquid phase separation. J Cell Biochem 2024; 125:e30530. [PMID: 38349116 DOI: 10.1002/jcb.30530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/10/2024] [Accepted: 01/24/2024] [Indexed: 03/12/2024]
Abstract
When the SARS-CoV-2 virus infects humans, it leads to a condition called COVID-19 that has a wide spectrum of clinical manifestations, from no symptoms to acute respiratory distress syndrome. The virus initiates damage by attaching to the ACE-2 protein on the surface of endothelial cells that line the blood vessels and using these cells as hosts for replication. Reactive oxygen species levels are increased during viral replication, which leads to oxidative stress. About three-fifths (~60%) of the people who get infected with the virus eradicate it from their body after 28 days and recover their normal activity. However, a large fraction (~40%) of the people who are infected with the virus suffer from various symptoms (anosmia and/or ageusia, fatigue, cough, myalgia, cognitive impairment, insomnia, dyspnea, and tachycardia) beyond 12 weeks and are diagnosed with a syndrome called long COVID. Long-term clinical studies in a group of people who contracted SARS-CoV-2 have been contrasted with a noninfected matched group of people. A subset of infected people can be distinguished by a set of cytokine markers to have persistent, low-grade inflammation and often self-report two or more bothersome symptoms. No medication can alleviate their symptoms efficiently. Coronavirus nucleocapsid proteins have been investigated extensively as potential drug targets due to their key roles in virus replication, among which is their ability to bind their respective genomic RNAs for incorporation into emerging virions. This review highlights basic studies of the nucleocapsid protein and its ability to undergo liquid-liquid phase separation. We hypothesize that this ability of the nucleocapsid protein for phase separation may contribute to long COVID. This hypothesis unlocks new investigation angles and could potentially open novel avenues for a better understanding of long COVID and treating this condition.
Collapse
Affiliation(s)
- Ahmed Eltayeb
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Faisal Al-Sarraj
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mona Alharbi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Raed Albiheyri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ehab H Mattar
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Isam M Abu Zeid
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Thamer A Bouback
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Atif Bamagoos
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Moscow Region, Russia
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan, Jalisco, Mexico
- Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco, Mexico
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
30
|
Heil M. Self-DNA driven inflammation in COVID-19 and after mRNA-based vaccination: lessons for non-COVID-19 pathologies. Front Immunol 2024; 14:1259879. [PMID: 38439942 PMCID: PMC10910434 DOI: 10.3389/fimmu.2023.1259879] [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: 07/17/2023] [Accepted: 12/26/2023] [Indexed: 03/06/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic triggered an unprecedented concentration of economic and research efforts to generate knowledge at unequalled speed on deregulated interferon type I signalling and nuclear factor kappa light chain enhancer in B-cells (NF-κB)-driven interleukin (IL)-1β, IL-6, IL-18 secretion causing cytokine storms. The translation of the knowledge on how the resulting systemic inflammation can lead to life-threatening complications into novel treatments and vaccine technologies is underway. Nevertheless, previously existing knowledge on the role of cytoplasmatic or circulating self-DNA as a pro-inflammatory damage-associated molecular pattern (DAMP) was largely ignored. Pathologies reported 'de novo' for patients infected with Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV)-2 to be outcomes of self-DNA-driven inflammation in fact had been linked earlier to self-DNA in different contexts, e.g., the infection with Human Immunodeficiency Virus (HIV)-1, sterile inflammation, and autoimmune diseases. I highlight particularly how synergies with other DAMPs can render immunogenic properties to normally non-immunogenic extracellular self-DNA, and I discuss the shared features of the gp41 unit of the HIV-1 envelope protein and the SARS-CoV 2 Spike protein that enable HIV-1 and SARS-CoV-2 to interact with cell or nuclear membranes, trigger syncytia formation, inflict damage to their host's DNA, and trigger inflammation - likely for their own benefit. These similarities motivate speculations that similar mechanisms to those driven by gp41 can explain how inflammatory self-DNA contributes to some of most frequent adverse events after vaccination with the BNT162b2 mRNA (Pfizer/BioNTech) or the mRNA-1273 (Moderna) vaccine, i.e., myocarditis, herpes zoster, rheumatoid arthritis, autoimmune nephritis or hepatitis, new-onset systemic lupus erythematosus, and flare-ups of psoriasis or lupus. The hope is to motivate a wider application of the lessons learned from the experiences with COVID-19 and the new mRNA vaccines to combat future non-COVID-19 diseases.
Collapse
Affiliation(s)
- Martin Heil
- Departamento de Ingeniería Genética, Laboratorio de Ecología de Plantas, Centro de Investigación y de Estudios Avanzados (CINVESTAV)-Unidad Irapuato, Irapuato, Mexico
| |
Collapse
|
31
|
Zhang K, Huang Q, Li X, Zhao Z, Hong C, Sun Z, Deng B, Li C, Zhang J, Wang S. The cGAS-STING pathway in viral infections: a promising link between inflammation, oxidative stress and autophagy. Front Immunol 2024; 15:1352479. [PMID: 38426093 PMCID: PMC10902852 DOI: 10.3389/fimmu.2024.1352479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
The host defence responses play vital roles in viral infection and are regulated by complex interactive networks. The host immune system recognizes viral pathogens through the interaction of pattern-recognition receptors (PRRs) with pathogen-associated molecular patterns (PAMPs). As a PRR mainly in the cytoplasm, cyclic GMP-AMP synthase (cGAS) senses and binds virus DNA and subsequently activates stimulator of interferon genes (STING) to trigger a series of intracellular signalling cascades to defend against invading pathogenic microorganisms. Integrated omic and functional analyses identify the cGAS-STING pathway regulating various host cellular responses and controlling viral infections. Aside from its most common function in regulating inflammation and type I interferon, a growing body of evidence suggests that the cGAS-STING signalling axis is closely associated with a series of cellular responses, such as oxidative stress, autophagy, and endoplasmic reticulum stress, which have major impacts on physiological homeostasis. Interestingly, these host cellular responses play dual roles in the regulation of the cGAS-STING signalling axis and the clearance of viruses. Here, we outline recent insights into cGAS-STING in regulating type I interferon, inflammation, oxidative stress, autophagy and endoplasmic reticulum stress and discuss their interactions with viral infections. A detailed understanding of the cGAS-STING-mediated potential antiviral effects contributes to revealing the pathogenesis of certain viruses and sheds light on effective solutions for antiviral therapy.
Collapse
Affiliation(s)
- Kunli Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Qiuyan Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Xinming Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Ziqiao Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Chun Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Zeyi Sun
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Bo Deng
- Division of Nephrology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunling Li
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
| | - Jianfeng Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| | - Sutian Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| |
Collapse
|
32
|
Sievers BL, Cheng MTK, Csiba K, Meng B, Gupta RK. SARS-CoV-2 and innate immunity: the good, the bad, and the "goldilocks". Cell Mol Immunol 2024; 21:171-183. [PMID: 37985854 PMCID: PMC10805730 DOI: 10.1038/s41423-023-01104-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/01/2023] [Indexed: 11/22/2023] Open
Abstract
An ancient conflict between hosts and pathogens has driven the innate and adaptive arms of immunity. Knowledge about this interplay can not only help us identify biological mechanisms but also reveal pathogen vulnerabilities that can be leveraged therapeutically. The humoral response to SARS-CoV-2 infection has been the focus of intense research, and the role of the innate immune system has received significantly less attention. Here, we review current knowledge of the innate immune response to SARS-CoV-2 infection and the various means SARS-CoV-2 employs to evade innate defense systems. We also consider the role of innate immunity in SARS-CoV-2 vaccines and in the phenomenon of long COVID.
Collapse
Affiliation(s)
| | - Mark T K Cheng
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kata Csiba
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Bo Meng
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Ravindra K Gupta
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
33
|
Abstract
Most enveloped viruses encode viral fusion proteins to penetrate host cell by membrane fusion. Interestingly, many enveloped viruses can also use viral fusion proteins to induce cell-cell fusion, both in vitro and in vivo, leading to the formation of syncytia or multinucleated giant cells (MGCs). In addition, some non-enveloped viruses encode specialized viral proteins that induce cell-cell fusion to facilitate viral spread. Overall, viruses that can induce cell-cell fusion are nearly ubiquitous in mammals. Virus cell-to-cell spread by inducing cell-cell fusion may overcome entry and post-entry blocks in target cells and allow evasion of neutralizing antibodies. However, molecular mechanisms of virus-induced cell-cell fusion remain largely unknown. Here, I summarize the current understanding of virus-induced cell fusion and syncytia formation.
Collapse
Affiliation(s)
- Maorong Xie
- Division of Infection and Immunity, UCL, London, UK.
| |
Collapse
|
34
|
Dobrovolny HM. Mathematical Modeling of Virus-Mediated Syncytia Formation: Past Successes and Future Directions. Results Probl Cell Differ 2024; 71:345-370. [PMID: 37996686 DOI: 10.1007/978-3-031-37936-9_17] [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: 11/25/2023]
Abstract
Many viruses have the ability to cause cells to fuse into large multi-nucleated cells, known as syncytia. While the existence of syncytia has long been known and its importance in helping spread viral infection within a host has been understood, few mathematical models have incorporated syncytia formation or examined its role in viral dynamics. This review examines mathematical models that have incorporated virus-mediated cell fusion and the insights they have provided on how syncytia can change the time course of an infection. While the modeling efforts are limited, they show promise in helping us understand the consequences of syncytia formation if future modeling efforts can be coupled with appropriate experimental efforts to help validate the models.
Collapse
Affiliation(s)
- Hana M Dobrovolny
- Department of Physics & Astronomy, Texas Christian University, Fort Worth, TX, USA.
| |
Collapse
|
35
|
Lee JD, Menasche BL, Mavrikaki M, Uyemura MM, Hong SM, Kozlova N, Wei J, Alfajaro MM, Filler RB, Müller A, Saxena T, Posey RR, Cheung P, Muranen T, Heng YJ, Paulo JA, Wilen CB, Slack FJ. Differences in syncytia formation by SARS-CoV-2 variants modify host chromatin accessibility and cellular senescence via TP53. Cell Rep 2023; 42:113478. [PMID: 37991919 PMCID: PMC10785701 DOI: 10.1016/j.celrep.2023.113478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/13/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) remains a significant public health threat due to the ability of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants to evade the immune system and cause breakthrough infections. Although pathogenic coronaviruses such as SARS-CoV-2 and Middle East respiratory syndrome (MERS)-CoV lead to severe respiratory infections, how these viruses affect the chromatin proteomic composition upon infection remains largely uncharacterized. Here, we use our recently developed integrative DNA And Protein Tagging methodology to identify changes in host chromatin accessibility states and chromatin proteomic composition upon infection with pathogenic coronaviruses. SARS-CoV-2 infection induces TP53 stabilization on chromatin, which contributes to its host cytopathic effect. We mapped this TP53 stabilization to the SARS-CoV-2 spike and its propensity to form syncytia, a consequence of cell-cell fusion. Differences in SARS-CoV-2 spike variant-induced syncytia formation modify chromatin accessibility, cellular senescence, and inflammatory cytokine release via TP53. Our findings suggest that differences in syncytia formation alter senescence-associated inflammation, which varies among SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Jonathan D Lee
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Bridget L Menasche
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Maria Mavrikaki
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Madison M Uyemura
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Su Min Hong
- Department of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Nina Kozlova
- Department of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jin Wei
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Mia M Alfajaro
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Renata B Filler
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Arne Müller
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Tanvi Saxena
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Ryan R Posey
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Priscilla Cheung
- Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Taru Muranen
- Department of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Yujing J Heng
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Craig B Wilen
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Frank J Slack
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Department of Genetics, Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
36
|
Liu Y, Zhou Y, Xu W, Li J, Wang S, Shen X, Wen X, Liu L. Aptamer-based kinetically controlled DNA reactions coupled with metal-organic framework nanoprobes for sensitive detection of SARS-CoV-2 spike protein. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:6583-6589. [PMID: 38014562 DOI: 10.1039/d3ay01585h] [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
Since the outbreak in 2019, COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become the deadliest infectious disease worldwide for people of all ages, from children to older adults. As a main structural protein of SARS-CoV-2, spike protein is reported to play a key role in the entry of the virus into host cells and is considered as an effective antigenic marker for COVID-19 diagnosis. Herein, we develop a new aptamer-based fluorescence method for SARS-CoV-2 spike protein detection based on using kinetically controlled DNA reactions and metal-organic framework nanoprobes. Specifically, the binding of SARS-CoV-2 spike protein to its aptamer is designed to precisely control the kinetics of a DNA displacement reaction, leading to the release of free signaling probes. By reasonable integration of magnetic enrichment and exonuclease-fuelled recycling, the released probes efficiently disrupt the interaction within metal-organic framework nanoprobes, thereby generating a remarkable fluorescent response. Experimental results show that the method not only exhibits a wide linear range and a low detection limit of 7.8 fg mL-1 for SARS-CoV-2 spike protein detection but also demonstrates desirable specificity and utility in complex samples. Therefore, the method may provide a valuable tool for the detection of SARS-CoV-2 spike protein, and has bright prospects in the rapid diagnosis of COVID-19, which is of great significance for guiding rational treatment during a pandemic of respiratory infectious diseases and reducing the occurrence of severe disease in children.
Collapse
Affiliation(s)
- Yan Liu
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu 610021, Sichuan, China.
| | - Yuanlin Zhou
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu 610021, Sichuan, China.
| | - Wanting Xu
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu 610021, Sichuan, China.
| | - Jiarong Li
- College of Clinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Shuning Wang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xiaojia Shen
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu 610021, Sichuan, China.
| | - Xiaobin Wen
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu 610021, Sichuan, China.
| | - Li Liu
- Department of Pediatrics, Chengdu Second People's Hospital, Chengdu 610021, Sichuan, China.
| |
Collapse
|
37
|
Sadik J, Sharma M, Deshmukh CV, Maqhbool SB, Thekiya AH, Kamble PP. Assessment of Genotoxic Biomarker in Tongue and Buccal Mucosal Epithelial Cells of COVID-19 Patients: An Observational Study. Cureus 2023; 15:e48706. [PMID: 38094540 PMCID: PMC10716604 DOI: 10.7759/cureus.48706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2023] [Indexed: 04/27/2025] Open
Abstract
INTRODUCTION Angiotensin-converting enzyme 2 (ACE2) is the main host cell receptor for coronavirus disease 2019 (COVID-19) and is highly expressed in the tongue and buccal mucosa. Therefore, the present study was conducted to investigate genotoxic changes in epithelial cells of the buccal and tongue mucosa following COVID-19 infection. MATERIALS AND METHODS This study included 40 patients aged 25-40 years, divided into two groups: Group 1 (control group) included 20 healthy individuals with no prior history of COVID-19 infection subdivided into Group 1a (buccal mucosa), and Group 1b (tongue mucosa); Group 2 (case group) included 20 patients with a history of mild to moderate COVID-19 infection subdivided into Group 2a (buccal mucosa) and Group 2b (tongue mucosa). Genotoxic biomarkers, such as the number of micronuclei, pyknosis, karyolysis, and karyorrhexis, were assessed in epithelial cells from the buccal mucosa and the ventral surface of the tongue. Analysis of variance was used for intragroup comparisons, followed by post-hoc analysis using Tukey's test. RESULTS The mean age of the patients was 27.4±6.52 years. Statistically significant differences were observed between cases and controls in the number of micronuclei, pyknosis, karyolysis, and karyorrhexis in the epithelial cells of the buccal and tongue mucosa (p = 0.05). CONCLUSION SARS-CoV-2 has pronounced genotoxic effects on the epithelium of the ventral surface of the tongue in comparison to the buccal mucosa Therefore, patients with COVID-19 should be monitored regularly to develop future carcinomas, particularly those with habits of smoking, alcohol consumption, and tobacco usage.
Collapse
Affiliation(s)
- Jafer Sadik
- Department of Dentistry, Government Hospital, Periyur, IND
| | - Manish Sharma
- Department of Oral Pathology, Jawahar Medical Foundation (JMF) Annasaheb Chudaman Patil Memorial (ACPM) Dental College, Dhule, IND
| | - Chetan V Deshmukh
- Department of Public Health Dentistry, Nair Hospital Dental College, Mumbai, IND
| | | | - Altaf H Thekiya
- Department of Orthodontics and Dentofacial Orthopedics, Diamond Dental Care, Nanded, IND
| | - Priyanka P Kamble
- Department of Oral Pathology, Jawahar Medical Foundation (JMF) Annasaheb Chudaman Patil Memorial (ACPM) Dental College, Dhule, IND
| |
Collapse
|
38
|
Ali H, Naseem A, Siddiqui ZI. SARS-CoV-2 Syncytium under the Radar: Molecular Insights of the Spike-Induced Syncytia and Potential Strategies to Limit SARS-CoV-2 Replication. J Clin Med 2023; 12:6079. [PMID: 37763019 PMCID: PMC10531702 DOI: 10.3390/jcm12186079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
SARS-CoV-2 infection induces non-physiological syncytia when its spike fusogenic protein on the surface of the host cells interacts with the ACE2 receptor on adjacent cells. Spike-induced syncytia are beneficial for virus replication, transmission, and immune evasion, and contribute to the progression of COVID-19. In this review, we highlight the properties of viral fusion proteins, mainly the SARS-CoV-2 spike, and the involvement of the host factors in the fusion process. We also highlight the possible use of anti-fusogenic factors as an antiviral for the development of therapeutics against newly emerging SARS-CoV-2 variants and how the fusogenic property of the spike could be exploited for biomedical applications.
Collapse
Affiliation(s)
- Hashim Ali
- Department of Pathology, University of Cambridge, Addenbrookes Hospital, Cambridge CB2 0QQ, UK
| | - Asma Naseem
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
| | - Zaheenul Islam Siddiqui
- Diabetes and Obesity Research Center, NYU Grossman Long Island School of Medicine, New York, NY 11501, USA
| |
Collapse
|
39
|
Lee JD, Menasche BL, Mavrikaki M, Uyemura MM, Hong SM, Kozlova N, Wei J, Alfajaro MM, Filler RB, Müller A, Saxena T, Posey RR, Cheung P, Muranen T, Heng YJ, Paulo JA, Wilen CB, Slack FJ. Differences in syncytia formation by SARS-CoV-2 variants modify host chromatin accessibility and cellular senescence via TP53. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.31.555625. [PMID: 37693555 PMCID: PMC10491142 DOI: 10.1101/2023.08.31.555625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
COVID-19 remains a significant public health threat due to the ability of SARS-CoV-2 variants to evade the immune system and cause breakthrough infections. Although pathogenic coronaviruses such as SARS-CoV-2 and MERS-CoV lead to severe respiratory infections, how these viruses affect the chromatin proteomic composition upon infection remains largely uncharacterized. Here we used our recently developed integrative DNA And Protein Tagging (iDAPT) methodology to identify changes in host chromatin accessibility states and chromatin proteomic composition upon infection with pathogenic coronaviruses. SARS-CoV-2 infection induces TP53 stabilization on chromatin, which contributes to its host cytopathic effect. We mapped this TP53 stabilization to the SARS-CoV-2 spike and its propensity to form syncytia, a consequence of cell-cell fusion. Differences in SARS-CoV-2 spike variant-induced syncytia formation modify chromatin accessibility, cellular senescence, and inflammatory cytokine release via TP53. Our findings suggest that differences in syncytia formation alter senescence-associated inflammation, which varies among SARS-CoV-2 variants.
Collapse
|
40
|
Liu X, Wang Y, Song T, Zheng Y, Zhang X, Li J, Li L, Augusto G, Sun F. Nonstructural protein VP2 of chicken anemia virus triggers IFN-β expression via host cGAS. Vet Microbiol 2023; 284:109842. [PMID: 37562113 DOI: 10.1016/j.vetmic.2023.109842] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/25/2023] [Accepted: 07/30/2023] [Indexed: 08/12/2023]
Abstract
Chicken anemia virus (CAV) constitutes an important economic threat for the poultry industry. Advancing the understanding of the pathogenic process of CAV infection, we had previously demonstrated that CAV VP1 has the ability to inhibit expression of IFN-β via cGAS-STING signalling pathway. Here to go further to reveal this regulatory role of viral phosphatase VP2, we have performed protein-protein interaction assays with cGAS adaptors, as well as IFN-β induction screenings. Contrary to VP1, VP2 of CAV stimulates the expression of IFN-β, a regulatory effect more closely associated with cGAS (in the context of the cGAS-STING axis) than with STING, TBK1 or IRF7. The results reported here offer new insights about the molecular mechanisms that varied viral proteins act in a timely manner on the host during CAV infection.
Collapse
Affiliation(s)
- Xuelan Liu
- Anhui Province Key Lab of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; International Immunology Center, Anhui Agricultural University, Hefei 230036, China
| | - Yuan Wang
- Anhui Province Key Lab of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Tao Song
- Anhui Province Key Lab of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; International Immunology Center, Anhui Agricultural University, Hefei 230036, China
| | - Yuting Zheng
- Anhui Province Key Lab of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; International Immunology Center, Anhui Agricultural University, Hefei 230036, China
| | - Xiaowang Zhang
- Anhui Province Key Lab of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; International Immunology Center, Anhui Agricultural University, Hefei 230036, China
| | - Jinnian Li
- Anhui Province Key Lab of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Lin Li
- Anhui Province Key Lab of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Gilles Augusto
- The Jenner Institute, University of Oxford, OX3 7DQ Oxford, United Kingdom
| | - Feifei Sun
- Anhui Province Key Lab of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|
41
|
Di Primio C, Quaranta P, Mignanelli M, Siano G, Bimbati M, Scarlatti A, Piazza CR, Spezia PG, Perrera P, Basolo F, Poma AM, Costa M, Pistello M, Cattaneo A. Severe acute respiratory syndrome coronavirus 2 infection leads to Tau pathological signature in neurons. PNAS NEXUS 2023; 2:pgad282. [PMID: 37731949 PMCID: PMC10508204 DOI: 10.1093/pnasnexus/pgad282] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/07/2023] [Accepted: 08/22/2023] [Indexed: 09/22/2023]
Abstract
COVID-19 has represented an issue for global health since its outbreak in March 2020. It is now evident that the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection results in a wide range of long-term neurological symptoms and is worryingly associated with the aggravation of Alzheimer's disease. Little is known about the molecular basis of these manifestations. Here, several strain variants were used to infect SH-SY5Y neuroblastoma cells and K18-hACE C57BL/6J mice. The Tau phosphorylation profile and aggregation propensity upon infection were investigated on cellular extracts, subcellular fractions, and brain tissue. The viral proteins spike, nucleocapsid, and membrane were overexpressed in SH-SY5Y cells, and the direct interaction and effect on Tau phosphorylation were checked using immunoblot experiments. Upon infection, Tau is phosphorylated at several pathological epitopes associated with Alzheimer's disease and other tauopathies. Moreover, this event increases Tau's propensity to form insoluble aggregates and alters its subcellular localization. Our data support the hypothesis that SARS-CoV-2 infection in the central nervous system triggers downstream effects altering Tau function, eventually leading to the impairment of neuronal function.
Collapse
Affiliation(s)
- Cristina Di Primio
- Institute of Neuroscience, Italian National Research Council (CNR), Pisa 56124, Italy
| | - Paola Quaranta
- Institute of Neuroscience, Italian National Research Council (CNR), Pisa 56124, Italy
- Retrovirus Center, Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56100, Italy
| | - Marianna Mignanelli
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore di Pisa, Pisa 56126, Italy
| | - Giacomo Siano
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore di Pisa, Pisa 56126, Italy
| | - Matteo Bimbati
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore di Pisa, Pisa 56126, Italy
- Department of Biotechnology, University of Verona, Verona 37134, Italy
| | - Arianna Scarlatti
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore di Pisa, Pisa 56126, Italy
| | - Carmen Rita Piazza
- Retrovirus Center, Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56100, Italy
- Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Piero Giorgio Spezia
- Retrovirus Center, Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56100, Italy
| | - Paola Perrera
- Retrovirus Center, Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56100, Italy
| | - Fulvio Basolo
- Department of Surgical, Medical and Molecular Pathology, University Hospital of Pisa, Pisa 56124, Italy
| | - Anello Marcello Poma
- Department of Surgical, Medical and Molecular Pathology, University Hospital of Pisa, Pisa 56124, Italy
| | - Mario Costa
- Institute of Neuroscience, Italian National Research Council (CNR), Pisa 56124, Italy
| | - Mauro Pistello
- Retrovirus Center, Virology Section, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa 56100, Italy
- Virology Unit, Pisa University Hospital, Pisa 56100, Italy
| | - Antonino Cattaneo
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore di Pisa, Pisa 56126, Italy
| |
Collapse
|
42
|
Kim IJ, Lee YH, Khalid MM, Chen IP, Zhang Y, Ott M, Verdin E. SARS-CoV-2 protein ORF8 limits expression levels of Spike antigen and facilitates immune evasion of infected host cells. J Biol Chem 2023; 299:104955. [PMID: 37354973 PMCID: PMC10289268 DOI: 10.1016/j.jbc.2023.104955] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023] Open
Abstract
Recovery from COVID-19 depends on the ability of the host to effectively neutralize virions and infected cells, a process largely driven by antibody-mediated immunity. However, with the newly emerging variants that evade Spike-targeting antibodies, re-infections and breakthrough infections are increasingly common. A full characterization of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mechanisms counteracting antibody-mediated immunity is therefore needed. Here, we report that ORF8 is a virally encoded SARS-CoV-2 factor that controls cellular Spike antigen levels. We show that ORF8 limits the availability of mature Spike by inhibiting host protein synthesis and retaining Spike at the endoplasmic reticulum, reducing cell-surface Spike levels and recognition by anti-SARS-CoV-2 antibodies. In conditions of limited Spike availability, we found ORF8 restricts Spike incorporation during viral assembly, reducing Spike levels in virions. Cell entry of these virions then leaves fewer Spike molecules at the cell surface, limiting antibody recognition of infected cells. Based on these findings, we propose that SARS-CoV-2 variants may adopt an ORF8-dependent strategy that facilitates immune evasion of infected cells for extended viral production.
Collapse
Affiliation(s)
- Ik-Jung Kim
- Buck Institute for Research on Aging, Novato, California, United States.
| | - Yong-Ho Lee
- Buck Institute for Research on Aging, Novato, California, United States; Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Mir M Khalid
- Gladstone Institutes, San Francisco, California, United States; Department of Medicine, University of California, San Francisco, San Francisco, California, United States
| | - Irene P Chen
- Gladstone Institutes, San Francisco, California, United States; Department of Medicine, University of California, San Francisco, San Francisco, California, United States
| | - Yini Zhang
- Buck Institute for Research on Aging, Novato, California, United States
| | - Melanie Ott
- Gladstone Institutes, San Francisco, California, United States; Department of Medicine, University of California, San Francisco, San Francisco, California, United States; Chan Zuckerberg Biohub, San Francisco, California, United States
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, California, United States.
| |
Collapse
|
43
|
Lee YJ, Seok SH, Lee NY, Choi HJ, Lee YW, Chang HJ, Hwang JY, On DI, Noh HA, Lee SB, Kwon HK, Yun JW, Shin JS, Seo JY, Nam KT, Lee H, Lee HY, Park JW, Seong JK. Murine Coronavirus Disease 2019 Lethality Is Characterized by Lymphoid Depletion Associated with Suppressed Antigen-Presenting Cell Functionality. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:866-882. [PMID: 37024046 PMCID: PMC10073095 DOI: 10.1016/j.ajpath.2023.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/28/2023] [Accepted: 03/16/2023] [Indexed: 04/08/2023]
Abstract
The disease severity of coronavirus disease 2019 (COVID-19) varies considerably from asymptomatic to serious, with fatal complications associated with dysregulation of innate and adaptive immunity. Lymphoid depletion in lymphoid tissues and lymphocytopenia have both been associated with poor disease outcomes in patients with COVID-19, but the mechanisms involved remain elusive. In this study, human angiotensin-converting enzyme 2 (hACE2) transgenic mouse models susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection were used to investigate the characteristics and determinants of lethality associated with the lymphoid depletion observed in SARS-CoV-2 infection. The lethality of Wuhan SARS-CoV-2 infection in K18-hACE2 mice was characterized by severe lymphoid depletion and apoptosis in lymphoid tissues related to fatal neuroinvasion. The lymphoid depletion was associated with a decreased number of antigen-presenting cells (APCs) and their suppressed functionality below basal levels. Lymphoid depletion with reduced APC function was a specific feature observed in SARS-CoV-2 infection but not in influenza A infection and had the greatest prognostic value for disease severity in murine COVID-19. Comparison of transgenic mouse models resistant and susceptible to SARS-CoV-2 infection revealed that suppressed APC function could be determined by the hACE2 expression pattern and interferon-related signaling. Thus, we demonstrated that lymphoid depletion associated with suppressed APC function characterizes the lethality of COVID-19 mouse models. Our data also suggest a potential therapeutic approach to prevent the severe progression of COVID-19 by enhancing APC functionality.
Collapse
Affiliation(s)
- Yu Jin Lee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, Republic of Korea
| | - Sang Hyeok Seok
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, Republic of Korea
| | - Na Yun Lee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, Republic of Korea
| | - Hee Jin Choi
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, Republic of Korea
| | - Yoon Woo Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Hee Jung Chang
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Ji-Yeon Hwang
- Preclinical Research Center, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Da In On
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, Republic of Korea
| | - Hyun Ah Noh
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, Republic of Korea
| | - Su-Bin Lee
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jun-Won Yun
- College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Jeon-Soo Shin
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Biomedical Science Institute, and the Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jun-Young Seo
- Severance Biomedical Science Institute, and the Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ki Taek Nam
- Severance Biomedical Science Institute, and the Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ho Lee
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Ho Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon, Republic of Korea.
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, Republic of Korea; Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK 21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
44
|
Maison DP, Deng Y, Gerschenson M. SARS-CoV-2 and the host-immune response. Front Immunol 2023; 14:1195871. [PMID: 37404823 PMCID: PMC10315470 DOI: 10.3389/fimmu.2023.1195871] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/05/2023] [Indexed: 07/06/2023] Open
Abstract
The SARS-CoV-2 pandemic and the COVID-19 disease have affected everyone globally, leading to one of recorded history's most significant research surges. As our knowledge evolves, our approaches to the virus and treatments must also evolve. The evaluation of future research approaches to SARS-CoV-2 will necessitate reviewing the host immune response and viral antagonism of that response. This review provides an overview of the current knowledge on SARS-CoV-2 by summarizing the virus and human response. The focuses are on the viral genome, replication cycle, host immune activation, response, signaling, and antagonism. To effectively fight the pandemic, efforts must focus on the current state of research to help develop treatments and prepare for future outbreaks.
Collapse
Affiliation(s)
- David P. Maison
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| |
Collapse
|
45
|
Gao D, Hao JP, Li BY, Zheng CC, Miao BB, Zhang L, Li YL, Li L, Li XJ, Zhang L. Tetrahydroxy stilbene glycoside ameliorates neuroinflammation for Alzheimer's disease via cGAS-STING. Eur J Pharmacol 2023:175809. [PMID: 37328043 DOI: 10.1016/j.ejphar.2023.175809] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 05/10/2023] [Accepted: 05/25/2023] [Indexed: 06/18/2023]
Abstract
Alzheimer's disease (AD), also known as senile dementia, is the most common degenerative disease of the central nervous system. Neuroinflammation is currently believed to be a crucial factor in the progression of AD, while its exact mechanism remains unclear. In this study, we demonstrated that AD transgenic mice exhibited cognitive deficits accompanied by the elevated serum and brain inflammation. Treating with a natural active ingredient tetrahydroxy stilbene glucoside (TSG) from the Chinese herb Polygonum multiflorum that has been well known for its unique anti-aging effect, learning-memory ability of AD mice was distinctly improved. Meanwhile, it was observed that the expressions of serum inflammatory cytokines and the activation of microglia in cerebral cortex and hippocampus were suppressed after TSG treatment, which was probably attributable to the decrease of cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) triggered immune response and NLRP3 inflammasome activation. Furthermore, cell culture experiments employing LPS combined with IFN-γ induced microglia activation showed that TSG reversed the polarization status of M1-type microglia to restore the quiescence, and cGAS-STING elevation was observed in the activated microglia and normalized by TSG incubation. In addition, TSG suppressed the production of inflammatory cytokines such as IL-1β, IL-6, TNF-α, IFN-α and IFN-β, as well as the expression of IFN regulatory proteins such as IFIT1 and IRF7 in the LPS/IFN-γ-stimulated inflammatory response in BV2 cell. Finally, it was also verified that TSG are, in part, through a cGAS-STING dependent pathway and triggered NLRP3 inflammasome activation to inhibit neuroinflammation through interfering with cGAS-STING inhibitors. Taken together, our findings highlight the health benefits of TSG and its potential application in preventing cognitive disorders by inhibiting neuroinflammation through cGAS-STING signaling pathway in AD.
Collapse
Affiliation(s)
- Dan Gao
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Jin-Ping Hao
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Bo-Ya Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Ceng-Ceng Zheng
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Bei-Bei Miao
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Li Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Ya-Li Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Lin Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Xing-Jie Li
- Senior Department of Infectious Diseases, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nervous System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China.
| |
Collapse
|
46
|
Coderch C, Arranz-Herrero J, Nistal-Villan E, de Pascual-Teresa B, Rius-Rocabert S. The Many Ways to Deal with STING. Int J Mol Sci 2023; 24:ijms24109032. [PMID: 37240378 DOI: 10.3390/ijms24109032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The stimulator of interferon genes (STING) is an adaptor protein involved in the activation of IFN-β and many other genes associated with the immune response activation in vertebrates. STING induction has gained attention from different angles such as the potential to trigger an early immune response against different signs of infection and cell damage, or to be used as an adjuvant in cancer immune treatments. Pharmacological control of aberrant STING activation can be used to mitigate the pathology of some autoimmune diseases. The STING structure has a well-defined ligand binding site that can harbor natural ligands such as specific purine cyclic di-nucleotides (CDN). In addition to a canonical stimulation by CDNs, other non-canonical stimuli have also been described, whose exact mechanism has not been well defined. Understanding the molecular insights underlying the activation of STING is important to realize the different angles that need to be considered when designing new STING-binding molecules as therapeutic drugs since STING acts as a versatile platform for immune modulators. This review analyzes the different determinants of STING regulation from the structural, molecular, and cell biology points of view.
Collapse
Affiliation(s)
- Claire Coderch
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - Javier Arranz-Herrero
- Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Departamento CC, Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - Estanislao Nistal-Villan
- Departamento CC, Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - Beatriz de Pascual-Teresa
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - Sergio Rius-Rocabert
- Departamento CC, Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| |
Collapse
|
47
|
Cai S, Zhang C, Zhuang Z, Zhang S, Ma L, Yang S, Zhou T, Wang Z, Xie W, Jin S, Zhao J, Guan X, Wu J, Cui J, Wu Y. Phase-separated nucleocapsid protein of SARS-CoV-2 suppresses cGAS-DNA recognition by disrupting cGAS-G3BP1 complex. Signal Transduct Target Ther 2023; 8:170. [PMID: 37100798 PMCID: PMC10131525 DOI: 10.1038/s41392-023-01420-9] [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: 09/23/2022] [Revised: 03/06/2023] [Accepted: 03/20/2023] [Indexed: 04/28/2023] Open
Abstract
Currently, the incidence and fatality rate of SARS-CoV-2 remain continually high worldwide. COVID-19 patients infected with SARS-CoV-2 exhibited decreased type I interferon (IFN-I) signal, along with limited activation of antiviral immune responses as well as enhanced viral infectivity. Dramatic progresses have been made in revealing the multiple strategies employed by SARS-CoV-2 in impairing canonical RNA sensing pathways. However, it remains to be determined about the SARS-CoV-2 antagonism of cGAS-mediated activation of IFN responses during infection. In the current study, we figure out that SARS-CoV-2 infection leads to the accumulation of released mitochondria DNA (mtDNA), which in turn triggers cGAS to activate IFN-I signaling. As countermeasures, SARS-CoV-2 nucleocapsid (N) protein restricts the DNA recognition capacity of cGAS to impair cGAS-induced IFN-I signaling. Mechanically, N protein disrupts the assembly of cGAS with its co-factor G3BP1 by undergoing DNA-induced liquid-liquid phase separation (LLPS), subsequently impairs the double-strand DNA (dsDNA) detection ability of cGAS. Taken together, our findings unravel a novel antagonistic strategy by which SARS-CoV-2 reduces DNA-triggered IFN-I pathway through interfering with cGAS-DNA phase separation.
Collapse
Affiliation(s)
- Sihui Cai
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, The First Affiliated Hospital of Sun Yat-sen University, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chenqiu Zhang
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, The First Affiliated Hospital of Sun Yat-sen University, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhen Zhuang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shengnan Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling Ma
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuai Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tao Zhou
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zheyu Wang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weihong Xie
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shouheng Jin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiangdong Guan
- Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Jianfeng Wu
- Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Jun Cui
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, The First Affiliated Hospital of Sun Yat-sen University, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Yaoxing Wu
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, The First Affiliated Hospital of Sun Yat-sen University, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Department of Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
48
|
Zhang J, Yang W, Roy S, Liu H, Roberts R, Wang L, Shi L, Ma W. Tight junction protein occludin is an internalization factor for SARS-CoV-2 infection and mediates virus cell-to-cell transmission. Proc Natl Acad Sci U S A 2023; 120:e2218623120. [PMID: 37068248 PMCID: PMC10151465 DOI: 10.1073/pnas.2218623120] [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: 10/31/2022] [Accepted: 03/13/2023] [Indexed: 04/19/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spreads efficiently by spike-mediated, direct cell-to-cell transmission. However, the underlying mechanism is poorly understood. Herein, we demonstrate that the tight junction protein occludin (OCLN) is critical to this process. SARS-CoV-2 infection alters OCLN distribution and expression and causes syncytium formation that leads to viral spread. OCLN knockdown fails to alter SARS-CoV-2 binding but significantly lowers internalization, syncytium formation, and transmission. OCLN overexpression also has no effect on virus binding but enhances virus internalization, cell-to-cell transmission, and replication. OCLN directly interacts with the SARS-CoV-2 spike, and the endosomal entry pathway is involved in OCLN-mediated cell-to-cell fusion rather than in the cell surface entry pathway. All SARS-CoV-2 strains tested (prototypic, alpha, beta, gamma, delta, kappa, and omicron) are dependent on OCLN for cell-to-cell transmission, although the extent of syncytium formation differs between strains. We conclude that SARS-CoV-2 utilizes OCLN as an internalization factor for cell-to-cell transmission.
Collapse
Affiliation(s)
- Jialin Zhang
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| | - Wenyu Yang
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| | - Sawrab Roy
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| | - Heidi Liu
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| | - R. Michael Roberts
- Division of Animal Sciences, College of Agriculture, Food, & Natural Resources, University of Missouri, Columbia, MO65211
- Christopher S Bond Life Sciences Center, University of Missouri, Columbia, MO65211
| | - Liping Wang
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| | - Lei Shi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| | - Wenjun Ma
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO65211
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO65211
| |
Collapse
|
49
|
He X, Sun Y, Lu J, Naz F, Ma S, Liu J. Cytoplasmic DNAs: Sources, sensing, and roles in the development of lung inflammatory diseases and cancer. Front Immunol 2023; 14:1117760. [PMID: 37122745 PMCID: PMC10130589 DOI: 10.3389/fimmu.2023.1117760] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Cytoplasmic DNA is emerging as a pivotal contributor to the pathogenesis of inflammatory diseases and cancer, such as COVID-19 and lung carcinoma. However, the complexity of various cytoplasmic DNA-related pathways and their crosstalk remains challenging to distinguish their specific roles in many distinct inflammatory diseases, especially for the underlying mechanisms. Here, we reviewed the latest findings on cytoplasmic DNA and its signaling pathways in inflammatory lung conditions and lung cancer progression. We found that sustained activation of cytoplasmic DNA sensing pathways contributes to the development of common lung diseases, which may result from external factors or mutations of key genes in the organism. We further discussed the interplays between cytoplasmic DNA and anti-inflammatory or anti-tumor effects for potential immunotherapy. In sum, this review aids in understanding the roles of cytoplasmic DNAs and exploring more therapeutic strategies.
Collapse
Affiliation(s)
- Xintong He
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Ye Sun
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jianzhang Lu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Faiza Naz
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Shenglin Ma
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Jian Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
- Cancer Center, Zhejiang University, Hangzhou, China
- Biomedical and Heath Translational Research Center of Zhejiang Province, Haining, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
50
|
Amurri L, Horvat B, Iampietro M. Interplay between RNA viruses and cGAS/STING axis in innate immunity. Front Cell Infect Microbiol 2023; 13:1172739. [PMID: 37077526 PMCID: PMC10106766 DOI: 10.3389/fcimb.2023.1172739] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023] Open
Abstract
While the function of cGAS/STING signalling axis in the innate immune response to DNA viruses is well deciphered, increasing evidence demonstrates its significant contribution in the control of RNA virus infections. After the first evidence of cGAS/STING antagonism by flaviviruses, STING activation has been detected following infection by various enveloped RNA viruses. It has been discovered that numerous viral families have implemented advanced strategies to antagonize STING pathway through their evolutionary path. This review summarizes the characterized cGAS/STING escape strategies to date, together with the proposed mechanisms of STING signalling activation perpetrated by RNA viruses and discusses possible therapeutic approaches. Further studies regarding the interaction between RNA viruses and cGAS/STING-mediated immunity could lead to major discoveries important for the understanding of immunopathogenesis and for the treatment of RNA viral infections.
Collapse
|