1
|
Xin X, Wu D, Zhao P, Li Y, Qin H, Dai J, Zhou Y, Lyu Y, Yang Y, Zhu Y, Shi H, Yang L, Yin L. Catch-to-Amplify Nanoparticles with Bacteria Surface for Sequential Mucosal Immune Activation for Acute Myeloid Leukemia Therapy. ACS NANO 2025; 19:14661-14679. [PMID: 40202129 DOI: 10.1021/acsnano.4c08515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Mucosal-mediated immune deficiency is associated with immune evasion and poor clinical outcomes in acute myeloid leukemia (AML). Here, we describe the elicitation of mucosal and systemic immune response by oral delivery of MDP-modified PEG-lipid (MDP-PEG-DSPE) and polylactic acid-polyhistidine (PLA-PHis) copolymer constructed nanosystem (mPOD) into Peyer's patches. To protect against gastrointestinal degradation, enteric-soluble capsules are utilized for encapsulating mPOD to promote penetration across intestinal mucus and engender robust Peyer's patch targeting initiated by MDP-PEG-DSPE. Compared with intravenous and intramuscular administration, the oral delivery of MDP-PEG-DSPE and 5'-triphosphate-modified RNA (ppp-RNA) into gut-associated lymphoid tissues reinforces dendritic cell maturation and migration, amplifies mucosal immune response, and boosts the production of secretory immunoglobulin A via retinoic acid-inducible gene I/nucleotide-binding oligomerization domain 2 (RIG-I/NOD2) signaling activation. In the AML murine model, the provoked mucosal immunity positively regulates the systemic cytotoxic immune reactions, which, in turn, eradicate disseminated malignant leukemic cells and provide defense against leukemia attacks.
Collapse
MESH Headings
- Animals
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/pathology
- Nanoparticles/chemistry
- Mice
- Immunity, Mucosal/drug effects
- Humans
- Mice, Inbred C57BL
- Polyethylene Glycols/chemistry
Collapse
Affiliation(s)
- Xiaofei Xin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China
| | - Di Wu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Pengbo Zhao
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yuanyuan Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Huanyu Qin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jinyu Dai
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yong Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yifu Lyu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Yang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Ying Zhu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hang Shi
- Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Lei Yang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China
| | - Lifang Yin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
2
|
De Rosa C, Morgillo F, Amato L, Iommelli F, De Rosa V, Tirino V, Papaccio F, Tuccillo C, Di Guida G, D’Angiolella DM, Di Liello A, Zappavigna S, Caraglia M, Gambardella A, Nardone V, Ramkumar K, Wang Q, Wang J, De Vita F, Ciardiello D, Martinelli E, Troiani T, Napolitano S, Martini G, Servetto A, Byers LA, Ciardiello F, Della Corte CM. DNA-PK inhibition sustains the antitumor innate immune response in small cell lung cancer. iScience 2025; 28:111943. [PMID: 40034862 PMCID: PMC11875153 DOI: 10.1016/j.isci.2025.111943] [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: 08/20/2024] [Revised: 11/20/2024] [Accepted: 01/29/2025] [Indexed: 03/05/2025] Open
Abstract
Small cell lung cancer (SCLC) is a highly aggressive form of lung cancer with limited treatment options. Patients often respond well to initial chemo-immunotherapy but relapse quickly, necessitating new strategies to enhance immune responsiveness. Recent research explores combining DNA-damaging therapies with immunotherapy to activate the STING pathway and improve the antitumor immune response. The addition of DNA Damage Repair (DDR) inhibitors, such as DNA-PKcs inhibitors, after chemotherapy has shown promise in activating innate immune sensors and enhancing CD8+ T cell and NK cell pathways in SCLC models. This approach could potentially reshape the tumor microenvironment and sustain an antitumor immune response, offering a maintenance strategy for SCLC treatment.
Collapse
Affiliation(s)
- Caterina De Rosa
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Floriana Morgillo
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Luisa Amato
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Francesca Iommelli
- Institute of Biostructures and Bioimaging, National Research Council, 80145 Naples, Italy
| | - Viviana De Rosa
- Institute of Biostructures and Bioimaging, National Research Council, 80145 Naples, Italy
| | - Virginia Tirino
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy
| | - Federica Papaccio
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, University of Salerno, 84084 Baronissi, Italy
| | - Concetta Tuccillo
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Gaetano Di Guida
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | | | - Alessandra Di Liello
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Silvia Zappavigna
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
- Laboratory of Precision and Molecular Oncology, Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Antonio Gambardella
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Valerio Nardone
- Radiology and Radiotherapy, Department of Precision Medicine, University of Campania ‘Luigi Vanvitelli’, 80138 Naples, Italy
| | - Kavya Ramkumar
- Department of Thoracic Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston TX 77030, USA
| | - Qi Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston TX 77030, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston TX 77030, USA
| | - Ferdinando De Vita
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Davide Ciardiello
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology (IEO), IRCCS, 20141 Milan, Italy
| | - Erika Martinelli
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Teresa Troiani
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Stefania Napolitano
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Giulia Martini
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Alberto Servetto
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples 80131, Italy
| | - Lauren Averett Byers
- Department of Thoracic Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston TX 77030, USA
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80131 Naples, Italy
| | | |
Collapse
|
3
|
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
|
4
|
Ghosh A, Britto J, Chandran B, Roy A. IFI16 recruits HDAC1 and HDAC2 to deacetylate the Kaposi's sarcoma-associated herpesvirus (KSHV) latency-associated nuclear antigen (LANA), facilitating latency. J Virol 2025; 99:e0154924. [PMID: 39927772 PMCID: PMC11915870 DOI: 10.1128/jvi.01549-24] [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: 09/03/2024] [Accepted: 01/17/2025] [Indexed: 02/11/2025] Open
Abstract
IFI16 (interferon-γ-inducible protein 16) is an innate-immune DNA sensor that detects viral dsDNA in the nucleus. It also functions as an antiviral restriction factor, playing a crucial role in regulating the latency/lytic balance of several herpesviruses, including Kaposi's sarcoma-associated herpesvirus (KSHV). We previously demonstrated that IFI16 achieves this by regulating the deposition of H3K9me3 marks on the KSHV genome. Here, we explored whether IFI16 impacts the KSHV latency/lytic balance through additional mechanisms. Our analysis of the IFI16 interactome revealed that IFI16 binds to the class-I HDACs, HDAC1 and HDAC2, and recruits them to the KSHV major latency protein, latency-associated nuclear antigen (LANA). Previous reports have suggested that LANA undergoes lysine acetylation through unknown mechanisms, which results in the loss of its ability to bind to the KSHV transactivator protein (RTA) promoter. However, how the LANA acetylation-deacetylation cycle is orchestrated and what effect this has on KSHV gene expression remains unknown. Here, we demonstrate that LANA, by default, undergoes post-translational acetylation, and during latency, IFI16 interacts with this acetylated LANA and recruits HDAC1/2 to it. This keeps LANA in a deacetylated form, competent in binding and repressing lytic promoters. However, during lytic reactivation, IFI16 is degraded via the proteasomal pathway, leading to the accumulation of acetylated LANA, which cannot bind to the RTA promoter. This results in the de-repression of the RTA and, subsequently, other lytic promoters, driving reactivation. These findings shed new light on the role of IFI16 in KSHV latency and suggest that KSHV utilizes the cellular IFI16-HDAC1/2 interaction to facilitate its latency. IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic γ-herpesviruses etiologically associated with several human malignancies, including Kaposi's sarcoma, primary effusion B-cell lymphoma, and multicentric Castleman's disease. Understanding the molecular mechanisms governing the establishment and maintenance of latency in γ-herpesviruses is crucial because latency plays a pivotal role in oncogenesis and disease manifestation post-infection. Here, we have elucidated a new mechanism by which IFI16, a previously discovered antiviral restriction factor, is hijacked by KSHV to recruit class-I HDACs on latency-associated nuclear antigen (LANA), resulting in the latter's deacetylation. The acetylation status of LANA is critical for KSHV latency because it governs LANA's binding to the KSHV replication and transcription activator (RTA) promoter, an immediate-early gene crucial for lytic reactivation. Depletion of IFI16 results in the accumulation of acetylated LANA, which is incapable of maintaining latency. These newly discovered interactions between IFI16 and LANA and between IFI16 and HDAC1/2 enhance our understanding of KSHV latency regulations.
Collapse
Affiliation(s)
- Anandita Ghosh
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Jeffrey Britto
- Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Bala Chandran
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Arunava Roy
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
5
|
Zhao W, Pang S, Zhang J, Yao Z, Song Y, Sun Y. AFB1 exposure promotes SIV replication and lung damage via RIG-I- and p38-mediated RETREG1/FAM134B-dependent endoplasmic reticulum autophagy. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117970. [PMID: 40009944 DOI: 10.1016/j.ecoenv.2025.117970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 02/16/2025] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
Aflatoxin B1 (AFB1) contamination is common worldwide and highly harmful to humans and animals. Our previous studies suggested that AFB1 exposure promotes the replication of H1N1 swine influenza virus (SIV). However, its mechanism is not clear. Here, TCID50, qRT-PCR, and WB assays were used to detect SIV replication, after which proteomic detection was used to screen key proteins and pathways. Thirty piglets were subsequently randomly divided into 6 groups. The low-pathogenicity SIV was inoculated to establish a piglet model of SIV infection. Different doses of AFB1 were administered daily to SIV-infected piglets for 14 d. The in vitro results revealed that 0.02-0.04 μg/mL AFB1 markedly promoted SIV replication. Proteomic analysis revealed that reticulophagy regulator 1 (RETREG1/FAM134B) and p38 signaling were markedly upregulated, whereas RIG-I signaling was significantly downregulated. The above results were confirmed by qRT-PCR and WB assays. Transmission electron microscopy was used to further prove that AFB1 promoted endoplasmic reticulum autophagy (ER-phagy) in SIV-infected PAMs. RIG-I activator and p38 inhibitor reversed the upregulation of RETREG1 and AFB1-promoted SIV replication, and RETREG1 inhibitor reversed the AFB1-promoted SIV replication. In vivo experiments confirmed that AFB1 upregulated RETREG1 and p38, downregulated RIG-I, and promoted SIV replication and lung damage. Taken together, our results reveal that AFB1 promotes SIV replication and lung damage via RIG-I- and p38-mediated RETREG1/FAM134B-dependent ER-phagy and suggest the therapeutic potential of RETREG1-, RIG-I-, and p38-related drugs for influenza. Our findings also provide insights into why the occurrence of other infectious diseases is increasing.
Collapse
Affiliation(s)
- Wenshuo Zhao
- College of Animal Science and Medicine, Shenyang Agricultural University, Shenyang 110866, China; Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang 110866, China; Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, P.R. China
| | - Siyao Pang
- College of Animal Science and Medicine, Shenyang Agricultural University, Shenyang 110866, China; Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang 110866, China; Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, P.R. China
| | - Jinlong Zhang
- College of Animal Science and Medicine, Shenyang Agricultural University, Shenyang 110866, China; Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang 110866, China; Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, P.R. China
| | - Zhaoran Yao
- College of Animal Science and Medicine, Shenyang Agricultural University, Shenyang 110866, China; Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang 110866, China; Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, P.R. China
| | - Yuqi Song
- College of Animal Science and Medicine, Shenyang Agricultural University, Shenyang 110866, China; Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang 110866, China; Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, P.R. China
| | - Yuhang Sun
- College of Animal Science and Medicine, Shenyang Agricultural University, Shenyang 110866, China; Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang 110866, China; Key Laboratory of Ruminant Infectious Disease Prevention and Control (East), Ministry of Agriculture and Rural Affairs, P.R. China.
| |
Collapse
|
6
|
Kulsuptrakul J, Emerman M, Mitchell PS. CARD8 inflammasome activation during HIV-1 cell-to-cell transmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.21.608981. [PMID: 39229127 PMCID: PMC11370340 DOI: 10.1101/2024.08.21.608981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Our previous work demonstrated that CARD8 detects HIV-1 infection by sensing the enzymatic activity of the HIV protease, resulting in CARD8-dependent inflammasome activation (Kulsuptrakul et al., 2023). CARD8 harbors a motif in its N-terminus that functions as a HIV protease substrate mimic, permitting innate immune recognition of HIV-1 protease activity, which when cleaved by HIV protease triggers CARD8 inflammasome activation. Here, we sought to understand CARD8 responses in the context of HIV-1 cell-to-cell transmission via a viral synapse. We observed that cell-to-cell transmission of HIV-1 between infected T cells and primary human monocyte-derived macrophages induces CARD8 inflammasome activation in a manner that is dependent on viral protease activity and largely independent of the NLRP3 inflammasome. Additionally, to further evaluate the viral determinants of CARD8 sensing, we tested a panel of HIV protease inhibitor resistant clones to establish how variation in HIV protease affects CARD8 activation. We identified mutant HIV-1 proteases that differentially cleave and activate CARD8 compared to wildtype HIV-1, thus indicating that natural variation in HIV protease affects not only the cleavage of the viral Gag-Pol polyprotein but also likely impacts innate sensing and inflammation.
Collapse
Affiliation(s)
- Jessie Kulsuptrakul
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA
- Divisions of Human Biology and Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Michael Emerman
- Divisions of Human Biology and Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Patrick S. Mitchell
- Department of Microbiology, University of Washington, Seattle, WA 98109, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98109, USA
| |
Collapse
|
7
|
Lv X, Zheng T, Lei X, Ren L, Zhao S, Wang J, Zhuo Z, Wang J. RTP4 restricts influenza A virus infection by targeting the viral NS1 protein. Virology 2025; 603:110397. [PMID: 39798334 DOI: 10.1016/j.virol.2025.110397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/22/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
The influenza A virus evades the host innate immune response to establish infection by inhibiting RIG-I activation through its nonstructural protein 1 (NS1). Here, we reported that receptor-transporting protein 4 (RTP4), an interferon-stimulated gene (ISG), targets NS1 to inhibit influenza A virus infection. Depletion of RTP4 significantly increased influenza A virus multiplication, while NS1-deficient viruses were unaffected. Mechanistically, RTP4 interacts with NS1 in an RNA-dependent manner and sequesters it from the TRIM25-RIG-I complex, thereby restoring TRIM25-mediated RIG-I K63-linked ubiquitination and subsequent activation of IRF3. Antiviral activity of RTP4 requires the evolutionarily conserved CXXC motifs and an H149 residue in the zinc finger domain, mutations of which disrupted RTP4-NS1 interaction and abrogated the ability of RTP4 to rescue RIG-I-mediated signaling. Collectively, our findings provided insights into the mechanism by which an ISG restricts influenza A virus replication by reactivating host antiviral signaling.
Collapse
Affiliation(s)
- Xueying Lv
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Beijing, China; State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China; Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, China
| | - Tian Zheng
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobo Lei
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Beijing, China
| | - Lili Ren
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Beijing, China
| | - Shiyi Zhao
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Jingfeng Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Zhou Zhuo
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
| | - Jianwei Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences, Beijing, China; National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
8
|
Luo J, Lu Y, Dai E, Yin N, Wang T, Qian H, Jiang Q, Cao X, Wang C, Zeng Y. The PB1 protein of H9N2 influenza A virus inhibits antiviral innate immunity by targeting MAVS for TRIM25-mediated autophagic degradation. Poult Sci 2025; 104:104639. [PMID: 39647358 PMCID: PMC11666973 DOI: 10.1016/j.psj.2024.104639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
The proteins encoded by Influenza A virus (IAV) evade the innate immune system through diverse strategies to facilitate their replication. However, the regulatory mechanisms remain not fully understood. In this study, we identified that the H9N2 PB1 protein suppressed the activities of the IFN-β, ISRE, and NF-κB promoters. Furthermore, H9N2 PB1 inhibited the phosphorylation of IRF3, IκBα, and TBK1 and the secretion of IFN-β. The results demonstrated H9N2 PB1 as a negative regulator of the RIG-I signaling pathway. Subsequent investigations revealed a specific interaction between H9N2 PB1 and MAVS, which disturbed the stability of MAVS. Notably, we discovered that H9N2 PB1 exploited the function of TRIM25, leading to the autophagic degradation of MAVS through K48-linked polyubiquitination. In conclusion, we uncovered a negative regulatory axis consisting of H9N2 PB1-TRIM25-MAVS-IFN-I. These findings provide valuable insights into the molecular interactions involved in the regulation of the host's innate immune antiviral response by IAV.
Collapse
Affiliation(s)
- Jiawei Luo
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China
| | - Yiyuan Lu
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, PR China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, PR China
| | - Enqi Dai
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China
| | - Nianchun Yin
- Suining Municipal Agricultural and Rural Bureau of Sichuan Province, Suining, 629000, PR China
| | - Ting Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China
| | - Hongxi Qian
- TECHLEX Food Co. Ltd, Mianyang, 621000, PR China
| | - Qingrong Jiang
- Sichuan Sundaily Farm Ecological Food Co. Ltd, Mianyang, 621000, PR China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, PR China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, PR China.
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, PR China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, PR China.
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, Changchun, 130118, PR China; Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, 130118, PR China; Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, PR China.
| |
Collapse
|
9
|
Wang C, Li L, Zhai X, Chang H, Liu H. Evasion of the Antiviral Innate Immunity by PRV. Int J Mol Sci 2024; 25:13140. [PMID: 39684850 DOI: 10.3390/ijms252313140] [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: 11/01/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Pseudorabies virus (PRV) establishes persistent latent infections by effectively evading the host's antiviral innate immune response. PRV has developed sophisticated strategies to bypass immune surveillance through coevolution with its host. Currently, no effective vaccine exists to prevent or treat infections caused by emerging PRV variants, and the interactions between PRV and the host's innate immune defenses remain incompletely understood. Nevertheless, ongoing research is uncovering insights that may lead to novel treatments and preventive approaches for herpesvirus-related diseases. This review summarizes recent advances in understanding how PRV disrupts key adaptors in immune signaling pathways to evade antiviral immunity. Additionally, we explored the intrinsic cellular defenses that play crucial roles in combating viral invasion. A deeper understanding of the immune evasion strategies of PRV could inform the development of new therapeutic targets and vaccines.
Collapse
Affiliation(s)
- Chenlong Wang
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450046, China
| | - Longxi Li
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450046, China
| | - Xinyu Zhai
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450046, China
| | - Hongtao Chang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Huimin Liu
- College of Life Sciences, Henan Agricultural University, Zhengzhou 450046, China
| |
Collapse
|
10
|
Liu J, Chen K, Wu W, Pang Z, Zhu D, Yan X, Wang B, Qiu J, Fang Z. GRP78 exerts antiviral function against influenza A virus infection by activating the IFN/JAK-STAT signaling. Virology 2024; 600:110249. [PMID: 39303344 DOI: 10.1016/j.virol.2024.110249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Influenza is an acute viral respiratory infection that causes mild to severe illness in humans and animals. Current studies show that glucose-regulated protein 78 (GRP78) can exert crucial functions during viral infection; however, the mechanism by which GRP78 regulates influenza A virus (IAV) infection remains unclear. In the present study, we found that IAV infection increased GRP78 expression. Overexpression of GRP78 significantly inhibited IAV replication, as indicated by reduced viral mRNA levels, protein levels, and viral titers. Mechanistically, Type I interferon (IFN) response signaling is upregulated during IAV infection by GRP78. Further study showed that GRP78 interacts with tyrosine kinase 2 (TYK2) and enhances its phosphorylation, thereby activating downstream STAT1/2 and antiviral IFN-stimulated gene (ISG) expression. Collectively, these results demonstrate an important mechanism by which GRP78 exerts in innate antiviral effect in IAV infection. This mechanism could be used as a therapeutic target for anti-influenza treatment.
Collapse
Affiliation(s)
- Jiaxin Liu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Kanghong Chen
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Wenjiao Wu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Zefen Pang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China
| | - Dandong Zhu
- School of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Xiukui Yan
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Bangqi Wang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| | - Jianxiang Qiu
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| | - Zhixin Fang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| |
Collapse
|
11
|
Feng R, Li D, Yan Z, Li X, Xie J. EMCV VP2 degrades IFI16 through Caspase-dependent apoptosis to evade IFI16-STING pathway. Virol J 2024; 21:296. [PMID: 39551733 PMCID: PMC11571899 DOI: 10.1186/s12985-024-02568-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 11/03/2024] [Indexed: 11/19/2024] Open
Abstract
Interferon (IFN)-γ inducible protein 16 (IFI16), a key DNA sensor, triggers downstream STING-dependent type I interferon (IFN-I) production and antiviral immunity. However, how the IFI16-STING signaling pathway is regulated by EMCV infection is still not well elucidated. In this study, we investigated the interaction between IFI16 and EMCV. Results indicated EMCV infection suppressed IFI16 expression in A549 cells. This study reveals that IFI16 plays an active role in combating EMCV. Screening viral proteins in conjunction with IFI16, we found that the EMCV VP2 protein hinders the antiviral response mediated by IFI16 by causing degradation of the IFI16 protein via the caspase-dependent apoptosis pathway. Our study communicates the antiviral role of the IFI16-STING pathway during EMCV infection. Importantly, this study unveils the novel mechanism by which VP2 counteracts the innate immune signaling activated by foreign DNA.
Collapse
Affiliation(s)
- Ruofei Feng
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, China
| | - Dianyu Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life Science and Engineering, Northwest Minzu University, No. 1 Xibeixincun, Lanzhou, 730030, China
| | - Zhenfang Yan
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, China
- College of Life Science and Engineering, Northwest Minzu University, No. 1 Xibeixincun, Lanzhou, 730030, China
| | - Xiangrong Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, China.
- Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, China.
| | - Jingying Xie
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, China.
- College of Life Science and Engineering, Northwest Minzu University, No. 1 Xibeixincun, Lanzhou, 730030, China.
| |
Collapse
|
12
|
Sosnovtseva AO, Demidova NA, Klimova RR, Kovalev MA, Kushch AA, Starodubova ES, Latanova AA, Karpov DS. Control of HSV-1 Infection: Directions for the Development of CRISPR/Cas-Based Therapeutics and Diagnostics. Int J Mol Sci 2024; 25:12346. [PMID: 39596412 PMCID: PMC11595115 DOI: 10.3390/ijms252212346] [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: 10/05/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
It is estimated that nearly all individuals have been infected with herpesviruses, with herpes simplex virus type 1 (HSV-1) representing the most prevalent virus. In most cases, HSV-1 causes non-life-threatening skin damage in adults. However, in patients with compromised immune systems, it can cause serious diseases, including death. The situation is further complicated by the emergence of strains that are resistant to both traditional and novel antiviral drugs. It is, therefore, imperative that new methods of combating HSV-1 and other herpesviruses be developed without delay. CRISPR/Cas systems may prove an effective means of controlling herpesvirus infections. This review presents the current understanding of the underlying molecular mechanisms of HSV-1 infection and discusses four potential applications of CRISPR/Cas systems in the fight against HSV-1 infections. These include the search for viral and cellular genes that may serve as effective targets, the optimization of anti-HSV-1 activity of CRISPR/Cas systems in vivo, the development of CRISPR/Cas-based HSV-1 diagnostics, and the validation of HSV-1 drug resistance mutations.
Collapse
Affiliation(s)
- Anastasiia O. Sosnovtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia; (A.O.S.); (M.A.K.); (E.S.S.); (A.A.L.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia
| | - Natalia A. Demidova
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Gamaleya Str., 18, 123098 Moscow, Russia; (N.A.D.); (R.R.K.); (A.A.K.)
| | - Regina R. Klimova
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Gamaleya Str., 18, 123098 Moscow, Russia; (N.A.D.); (R.R.K.); (A.A.K.)
| | - Maxim A. Kovalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia; (A.O.S.); (M.A.K.); (E.S.S.); (A.A.L.)
| | - Alla A. Kushch
- N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Gamaleya Str., 18, 123098 Moscow, Russia; (N.A.D.); (R.R.K.); (A.A.K.)
| | - Elizaveta S. Starodubova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia; (A.O.S.); (M.A.K.); (E.S.S.); (A.A.L.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia
| | - Anastasia A. Latanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia; (A.O.S.); (M.A.K.); (E.S.S.); (A.A.L.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia
| | - Dmitry S. Karpov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia; (A.O.S.); (M.A.K.); (E.S.S.); (A.A.L.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov Str., 32, 119991 Moscow, Russia
| |
Collapse
|
13
|
Li J, Hao Y, Wu L, Liang H, Ni L, Wang F, Wang S, Duan Y, Xu Q, Xiao J, Yang D, Gao G, Ding Y, Gao C, Xiao J, Zhao H. Exploration of common pathogenesis and candidate hub genes between HIV and monkeypox co-infection using bioinformatics and machine learning. Sci Rep 2024; 14:26701. [PMID: 39496786 PMCID: PMC11535269 DOI: 10.1038/s41598-024-78540-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/31/2024] [Indexed: 11/06/2024] Open
Abstract
This study explored the pathogenesis of human immunodeficiency virus (HIV) and monkeypox co-infection, identifying candidate hub genes and potential drugs using bioinformatics and machine learning. Datasets for HIV (GSE 37250) and monkeypox (GSE 24125) were obtained from the GEO database. Common differentially expressed genes (DEGs) in co-infection were identified by intersecting DEGs from monkeypox datasets with genes from key HIV modules screened using Weighted Gene Co-Expression Network Analysis (WGCNA). After gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis and construction of protein-protein interaction (PPI) network, candidate hub genes were further screened based on machine learning algorithms. Transcriptional factors (TFs) and miRNA-candidate hub gene networks were constructed to understand regulatory mechanisms and protein-drug interactions to identify potential therapeutic drugs. Seven candidate hub genes-MX2, ADAR, POLR2H, RPL5, IFI16, IFIT2, and RPS5-were identified. TFs and miRNAs associated with these hub genes, playing a key role in regulating viral infection and inflammation due to the activation of antiviral innate immunity, were also identified through network analysis. Potential therapeutic drugs were screened based on these hub genes: AZT, a nucleotide reverse transcriptase inhibitor, suppressed viral replication in HIV and monkeypox co-infection, while mefloquine inhibited inflammation due to the activation of antiviral innate immunity. In conclusion, the study identified candidate hub genes, their transcriptional regulation, signaling pathways, and small-molecule drugs in HIV and monkeypox co-infection, contributing to understanding the pathogenesis of HIV and monkeypox co-infection and informing precise therapeutic strategies.
Collapse
Affiliation(s)
- Jialu Li
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Yiwei Hao
- Division of Medical Record and Statistics, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Liang Wu
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Hongyuan Liang
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Liang Ni
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Fang Wang
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Sa Wang
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Yujiao Duan
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Qiuhua Xu
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Jinjing Xiao
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, China
| | - Di Yang
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Guiju Gao
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Yi Ding
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Chengyu Gao
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China
| | - Jiang Xiao
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China.
| | - Hongxin Zhao
- Clinical Center of HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Jingshun East Street, Chaoyang District, Beijing, 100015, China.
| |
Collapse
|
14
|
Kobayashi Y, Bustos MA, Hayashi Y, Yu Q, Hoon D. Interferon-induced factor 16 is essential in metastatic melanoma to maintain STING levels and the immune responses upon IFN-γ response pathway activation. J Immunother Cancer 2024; 12:e009590. [PMID: 39424359 PMCID: PMC11492949 DOI: 10.1136/jitc-2024-009590] [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] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitor (ICIs)-based therapies are the standard of care treatment for patients with metastatic melanoma (MM). The stimulator of interferon genes (STING) signaling pathway is critical in controlling immune responses to ICIs. Interferon (IFN)-γ-inducible protein 16 (IFI16) is a cytosolic DNA sensor that activates the STING signaling pathway. The link between IFI16 and STING signaling pathway on IFN-γ stimulation and the connection to ICIs response remains not completely understood. METHODS Deconvolution analyses were performed using the TCGA-SKCM, GSE91061, and PRJEB23709 public RNA sequencing (RNA-seq) data sets that contained RNA-seq for patients with MM. Functional assays combined with cytokine arrays were performed using MM cell lines to validate in silico data. Multiplex immunofluorescence was performed on untreated or pretreatment tumor samples from patients with MM. RESULTS Deconvolution analysis showed that high-IFI16 levels in melanoma cells were associated with a good prognosis in patients with MM and positively correlated with M1-macrophage infiltration. Functional assays using MM cell lines demonstrated that IFI16 is a key molecule to sense cytosolic DNA and activate STING and nuclear factor kappa B (NF-κB) signaling pathways, independent of cyclic GMP-AMP synthase or absent in melanoma 2, on IFN-γ stimulation. IFI16 knockdown significantly decreased CXCL10 and ICAM1 secretion. EZH2 inhibitor reversed the repressive epigenetic control on IFI16 to promote STING and NF-κB signaling pathways on IFN-γ stimulation. Increased IFI16, ICAM1, and CXCL10 levels in tumor samples from patients with MM were positively correlated with M1-macrophage infiltration and a significantly better response to ICIs. CONCLUSIONS This study identifies IFI16 as a key sensor during IFN-γ stimulation associated with ICI response, and it proposes the epigenetic EZH2 inhibitor as an alternative treatment strategy to overcome ICI resistance in patients with MM.
Collapse
Affiliation(s)
- Yuta Kobayashi
- Dept. of Translational Molecular Medicine, Saint John's Cancer Institute, Santa Monica, California, USA
| | - Matias A Bustos
- Dept. of Translational Molecular Medicine, Saint John's Cancer Institute, Santa Monica, California, USA
| | - Yoshinori Hayashi
- Dept. of Translational Molecular Medicine, Saint John's Cancer Institute, Santa Monica, California, USA
| | - Qiang Yu
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore
| | - Dave Hoon
- Dept. of Translational Molecular Medicine, Saint John's Cancer Institute, Santa Monica, California, USA
| |
Collapse
|
15
|
Walch P, Broz P. Viral-bacterial co-infections screen in vitro reveals molecular processes affecting pathogen proliferation and host cell viability. Nat Commun 2024; 15:8595. [PMID: 39366977 PMCID: PMC11452664 DOI: 10.1038/s41467-024-52905-2] [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: 03/14/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
The broadening of accessible methodologies has enabled mechanistic insights into single-pathogen infections, yet the molecular mechanisms underlying co-infections remain largely elusive, despite their clinical frequency and relevance, generally exacerbating symptom severity and fatality. Here, we describe an unbiased in vitro screening of pairwise co-infections in a murine macrophage model, quantifying pathogen proliferation and host cell death in parallel over time. The screen revealed that the majority of interactions are antagonistic for both metrics, highlighting general patterns depending on the pathogen virulence strategy. We subsequently decipher two distinct molecular interaction points: Firstly, murine Adenovirus 3 modifies ASC-dependent inflammasome responses in murine macrophages, altering host cell death and cytokine production, thereby impacting secondary Salmonella infection. Secondly, murine Adenovirus 2 infection triggers upregulation of Mprip, a crucial mediator of phagocytosis, which in turn causes increased Yersinia uptake, specifically in virus pre-infected bone-marrow-derived macrophages. This work therefore encompasses both a first-of-its-kind systematic assessment of host-pathogen-pathogen interactions, and mechanistic insight into molecular mediators during co-infection.
Collapse
Affiliation(s)
- Philipp Walch
- University of Lausanne, Department of Immunobiology, Chemin des Boveresses 155, CH-1066, Epalinges, Switzerland
| | - Petr Broz
- University of Lausanne, Department of Immunobiology, Chemin des Boveresses 155, CH-1066, Epalinges, Switzerland.
| |
Collapse
|
16
|
Deng Y, Pang X, Chen L, Peng W, Huang X, Huang P, Zhao S, Li Z, Cai X, Huang Q, Zeng J, Feng Y, Chen B. IFI-16 inhibition attenuates myocardial remodeling following myocardial infarction. iScience 2024; 27:110568. [PMID: 39188985 PMCID: PMC11345598 DOI: 10.1016/j.isci.2024.110568] [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: 02/26/2024] [Revised: 05/16/2024] [Accepted: 07/19/2024] [Indexed: 08/28/2024] Open
Abstract
Myocardial remodeling (MR) following myocardial infarction (MI) contributes to heart failure. Inflammation is a key determinant in cardiac remodeling, with potential prognostic improvements by inhibiting inflammatory factors. Pattern recognition receptors, including interferon gamma-inducible protein-16 (IFI-16), play significant roles in this process, yet its specific involvement remains underexplored. This study investigates IFI-16's role in initiating inflammation via the inflammasome and its direct interaction with galectin-3 protein post-MI. Elevated IFI-16 levels were observed in human and rat myocytes and a mouse MI model under hypoxic, nutrient-deprived conditions, correlating with increased inflammation-associated proteins. Suppression of IFI-16/IFI-204 using short hairpin RNA (shRNA) lentivirus or adeno-associated virus decreased inflammatory factor activation, thereby mitigating remodeling and enhancing cardiac function post-MI. Co-immunoprecipitation (coIP) and double-fluorescence staining confirmed IFI-16's ability to interact directly with galectin-3. These findings underscore IFI-16's critical role as a pro-inflammatory factor in post-MI MR, suggesting its inhibition as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Yi Deng
- The Second Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- The Postdoctoral Research Station, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Emergency Department of Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
| | - Xiuqing Pang
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Guangzhou 510630, China
| | - Li Chen
- The Second Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Emergency Department of Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
| | - Weihang Peng
- The Second Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
| | - Xiaoyan Huang
- The Second Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Emergency Department of Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
| | - Peiying Huang
- The Second Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Emergency Department of Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
| | - Shuai Zhao
- The Second Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Emergency Department of Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
| | - Zhishang Li
- The Second Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Emergency Department of Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
| | - Xingui Cai
- The Second Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Emergency Department of Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
| | - Qiuping Huang
- The Second Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Emergency Department of Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
| | - Jing Zeng
- The Second Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Emergency Department of Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
| | - Yuchao Feng
- The Second Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Emergency Department of Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
| | - Bojun Chen
- The Second Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Guangdong Provincial Key Laboratory of Research on Emergency in Traditional Chinese Medicine, Clinical Research Team of Prevention and Treatment of Cardiac Emergencies with Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
- Emergency Department of Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou City, Guangdong Province 511400, China
| |
Collapse
|
17
|
An W, Lakhina S, Leong J, Rawat K, Husain M. Host Innate Antiviral Response to Influenza A Virus Infection: From Viral Sensing to Antagonism and Escape. Pathogens 2024; 13:561. [PMID: 39057788 PMCID: PMC11280125 DOI: 10.3390/pathogens13070561] [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: 05/31/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Influenza virus possesses an RNA genome of single-stranded, negative-sensed, and segmented configuration. Influenza virus causes an acute respiratory disease, commonly known as the "flu" in humans. In some individuals, flu can lead to pneumonia and acute respiratory distress syndrome. Influenza A virus (IAV) is the most significant because it causes recurring seasonal epidemics, occasional pandemics, and zoonotic outbreaks in human populations, globally. The host innate immune response to IAV infection plays a critical role in sensing, preventing, and clearing the infection as well as in flu disease pathology. Host cells sense IAV infection through multiple receptors and mechanisms, which culminate in the induction of a concerted innate antiviral response and the creation of an antiviral state, which inhibits and clears the infection from host cells. However, IAV antagonizes and escapes many steps of the innate antiviral response by different mechanisms. Herein, we review those host and viral mechanisms. This review covers most aspects of the host innate immune response, i.e., (1) the sensing of incoming virus particles, (2) the activation of downstream innate antiviral signaling pathways, (3) the expression of interferon-stimulated genes, (4) and viral antagonism and escape.
Collapse
Affiliation(s)
| | | | | | | | - Matloob Husain
- Department of Microbiology and Immunology, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand; (W.A.); (S.L.); (J.L.); (K.R.)
| |
Collapse
|
18
|
Melepat B, Li T, Vinkler M. Natural selection directing molecular evolution in vertebrate viral sensors. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 154:105147. [PMID: 38325501 DOI: 10.1016/j.dci.2024.105147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 12/30/2023] [Accepted: 02/03/2024] [Indexed: 02/09/2024]
Abstract
Diseases caused by pathogens contribute to molecular adaptations in host immunity. Variety of viral pathogens challenging animal immunity can drive positive selection diversifying receptors recognising the infections. However, whether distinct virus sensing systems differ across animals in their evolutionary modes remains unclear. Our review provides a comparative overview of natural selection shaping molecular evolution in vertebrate viral-binding pattern recognition receptors (PRRs). Despite prevailing negative selection arising from the functional constraints, multiple lines of evidence now suggest diversifying selection in the Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I-like receptors (RLRs) and oligoadenylate synthetases (OASs). In several cases, location of the positively selected sites in the ligand-binding regions suggests effects on viral detection although experimental support is lacking. Unfortunately, in most other PRR families including the AIM2-like receptor family, C-type lectin receptors (CLRs), and cyclic GMP-AMP synthetase studies characterising their molecular evolution are rare, preventing comparative insight. We indicate shared characteristics of the viral sensor evolution and highlight priorities for future research.
Collapse
Affiliation(s)
- Balraj Melepat
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, 128 43, Prague, EU, Czech Republic
| | - Tao Li
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, 128 43, Prague, EU, Czech Republic
| | - Michal Vinkler
- Charles University, Faculty of Science, Department of Zoology, Viničná 7, 128 43, Prague, EU, Czech Republic.
| |
Collapse
|
19
|
Aouadi W, Najburg V, Legendre R, Varet H, Kergoat L, Tangy F, Larrous F, Komarova AV, Bourhy H. Comparative analysis of rabies pathogenic and vaccine strains detection by RIG-I-like receptors. Microbes Infect 2024; 26:105321. [PMID: 38461968 DOI: 10.1016/j.micinf.2024.105321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/25/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
Rabies virus (RABV) is a lethal neurotropic virus that causes 60,000 human deaths every year globally. RABV infection is characterized by the suppression of the interferon (IFN)-mediated antiviral response. However, molecular mechanisms leading to RABV sensing by RIG-I-like receptors (RLR) that initiates IFN signaling currently remain elusive. Here, we showed that RABV RNAs are primarily recognized by the RIG-I RLR, resulting in an IFN response in the infected cells, but this response varied according to the type of RABV used. Pathogenic RABV strain RNAs, Tha, were poorly detected in the cytosol by RIG-I and therefore caused a weak antiviral response. However, we revealed a strong IFN activity triggered by the attenuated RABV vaccine strain RNAs, SAD, mediated by RIG-I. We characterized two major 5' copy-back defective interfering (5'cb DI) genomes generated during SAD replication. Furthermore, we identified an interaction between 5'cb DI genomes, and RIG-I correlated with a high stimulation of the type I IFN signaling. This study indicates that wild-type RABV RNAs poorly activate the RIG-I pathway, while the presence of 5'cb DIs in the live-attenuated vaccine strain serves as an intrinsic adjuvant that strengthens its efficiency by enhancing RIG-I detection thus strongly stimulates the IFN response.
Collapse
Affiliation(s)
- Wahiba Aouadi
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, 75015 Paris, France
| | - Valérie Najburg
- Institut Pasteur, Université Paris Cité, Vaccines-innovation Laboratory, 75015 Paris, France
| | - Rachel Legendre
- Institut Pasteur, Université Paris Cité, Hub Bioinformatics, and Biostatistics, 75015 Paris, France
| | - Hugo Varet
- Institut Pasteur, Université Paris Cité, Hub Bioinformatics, and Biostatistics, 75015 Paris, France
| | - Lauriane Kergoat
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, 75015 Paris, France
| | - Frédéric Tangy
- Institut Pasteur, Université Paris Cité, Vaccines-innovation Laboratory, 75015 Paris, France
| | - Florence Larrous
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, 75015 Paris, France
| | - Anastassia V Komarova
- Institut Pasteur, Université Paris Cité, Interactomics, RNA and Immunity Laboratory, 75015 Paris, France.
| | - Hervé Bourhy
- Institut Pasteur, Université Paris Cité, Lyssavirus Epidemiology and Neuropathology Unit, 75015 Paris, France.
| |
Collapse
|
20
|
Chang X, Wang B, Zhao Y, Deng B, Liu P, Wang Y. The role of IFI16 in regulating PANoptosis and implication in heart diseases. Cell Death Discov 2024; 10:204. [PMID: 38693141 PMCID: PMC11063201 DOI: 10.1038/s41420-024-01978-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024] Open
Abstract
Interferon Gamma Inducible Protein 16 (IFI16) belongs to the HIN-200 protein family and is pivotal in immunological responses. Serving as a DNA sensor, IFI16 identifies viral and aberrant DNA, triggering immune and inflammatory responses. It is implicated in diverse cellular death mechanisms, such as pyroptosis, apoptosis, and necroptosis. Notably, these processes are integral to the emergent concept of PANoptosis, which encompasses cellular demise and inflammatory pathways. Current research implies a significant regulatory role for IFI16 in PANoptosis, particularly regarding cardiac pathologies. This review delves into the complex interplay between IFI16 and PANoptosis in heart diseases, including atherosclerosis, myocardial infarction, heart failure, and diabetic cardiomyopathy. It synthesizes evidence of IFI16's impact on PANoptosis, with the intention of providing novel insights for therapeutic strategies targeting heart diseases.
Collapse
Affiliation(s)
- Xindi Chang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Bei Wang
- Department of Emergency, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Yingli Zhao
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Bing Deng
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China
| | - Ping Liu
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China.
| | - Yiru Wang
- Department of Cardiology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wan-Ping Road, Shanghai, China.
| |
Collapse
|
21
|
Jia KW, Yao RQ, Fan YW, Zhang DJ, Zhou Y, Wang MJ, Zhang LY, Dong Y, Li ZX, Wang SY, Wang M, Li YH, Zhang LX, Lei T, Gui LC, Lu S, Yang YY, Wang SX, Yu YZ, Yao YM, Hou J. Interferon-α stimulates DExH-box helicase 58 to prevent hepatocyte ferroptosis. Mil Med Res 2024; 11:22. [PMID: 38622688 PMCID: PMC11017495 DOI: 10.1186/s40779-024-00524-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 03/19/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Liver ischemia/reperfusion (I/R) injury is usually caused by hepatic inflow occlusion during liver surgery, and is frequently observed during war wounds and trauma. Hepatocyte ferroptosis plays a critical role in liver I/R injury, however, it remains unclear whether this process is controlled or regulated by members of the DEAD/DExH-box helicase (DDX/DHX) family. METHODS The expression of DDX/DHX family members during liver I/R injury was screened using transcriptome analysis. Hepatocyte-specific Dhx58 knockout mice were constructed, and a partial liver I/R operation was performed. Single-cell RNA sequencing (scRNA-seq) in the liver post I/R suggested enhanced ferroptosis by Dhx58hep-/-. The mRNAs and proteins associated with DExH-box helicase 58 (DHX58) were screened using RNA immunoprecipitation-sequencing (RIP-seq) and IP-mass spectrometry (IP-MS). RESULTS Excessive production of reactive oxygen species (ROS) decreased the expression of the IFN-stimulated gene Dhx58 in hepatocytes and promoted hepatic ferroptosis, while treatment using IFN-α increased DHX58 expression and prevented ferroptosis during liver I/R injury. Mechanistically, DHX58 with RNA-binding activity constitutively associates with the mRNA of glutathione peroxidase 4 (GPX4), a central ferroptosis suppressor, and recruits the m6A reader YT521-B homology domain containing 2 (YTHDC2) to promote the translation of Gpx4 mRNA in an m6A-dependent manner, thus enhancing GPX4 protein levels and preventing hepatic ferroptosis. CONCLUSIONS This study provides mechanistic evidence that IFN-α stimulates DHX58 to promote the translation of m6A-modified Gpx4 mRNA, suggesting the potential clinical application of IFN-α in the prevention of hepatic ferroptosis during liver I/R injury.
Collapse
Affiliation(s)
- Kai-Wei Jia
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Ren-Qi Yao
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, 100853, China
| | - Yi-Wen Fan
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Ding-Ji Zhang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Ye Zhou
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Min-Jun Wang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Li-Yuan Zhang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Yue Dong
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Zhi-Xuan Li
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Su-Yuan Wang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Mu Wang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Yun-Hui Li
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Lu-Xin Zhang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Ting Lei
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Liang-Chen Gui
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Shan Lu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Ying-Yun Yang
- Center for Immunotherapy, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Si-Xian Wang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Yi-Zhi Yu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, 100853, China.
| | - Jin Hou
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
22
|
Bottardi S, Layne T, Ramòn AC, Quansah N, Wurtele H, Affar EB, Milot E. MNDA, a PYHIN factor involved in transcriptional regulation and apoptosis control in leukocytes. Front Immunol 2024; 15:1395035. [PMID: 38680493 PMCID: PMC11045911 DOI: 10.3389/fimmu.2024.1395035] [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: 03/02/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Abstract
Inflammation control is critical during the innate immune response. Such response is triggered by the detection of molecules originating from pathogens or damaged host cells by pattern-recognition receptors (PRRs). PRRs subsequently initiate intra-cellular signalling through different pathways, resulting in i) the production of inflammatory cytokines, including type I interferon (IFN), and ii) the initiation of a cascade of events that promote both immediate host responses as well as adaptive immune responses. All human PYRIN and HIN-200 domains (PYHIN) protein family members were initially proposed to be PRRs, although this view has been challenged by reports that revealed their impact on other cellular mechanisms. Of relevance here, the human PYHIN factor myeloid nuclear differentiation antigen (MNDA) has recently been shown to directly control the transcription of genes encoding factors that regulate programmed cell death and inflammation. While MNDA is mainly found in the nucleus of leukocytes of both myeloid (neutrophils and monocytes) and lymphoid (B-cell) origin, its subcellular localization has been shown to be modulated in response to genotoxic agents that induce apoptosis and by bacterial constituents, mediators of inflammation. Prior studies have noted the importance of MNDA as a marker for certain forms of lymphoma, and as a clinical prognostic factor for hematopoietic diseases characterized by defective regulation of apoptosis. Abnormal expression of MNDA has also been associated with altered levels of cytokines and other inflammatory mediators. Refining our comprehension of the regulatory mechanisms governing the expression of MNDA and other PYHIN proteins, as well as enhancing our definition of their molecular functions, could significantly influence the management and treatment strategies of numerous human diseases. Here, we review the current state of knowledge regarding PYHIN proteins and their role in innate and adaptive immune responses. Emphasis will be placed on the regulation, function, and relevance of MNDA expression in the control of gene transcription and RNA stability during cell death and inflammation.
Collapse
Affiliation(s)
- Stefania Bottardi
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
| | - Taylorjade Layne
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
| | - Ailyn C. Ramòn
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Norreen Quansah
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Hugo Wurtele
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - El Bachir Affar
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Eric Milot
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de l’Est-de-l’Île de Montreal, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
23
|
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
|
24
|
Shi X, Wei M, Feng Y, Yang Y, Zhang X, Chen H, Xing Y, Wang K, Wang W, Wang L, Wang A, Zhang G. IFI16 Positively Regulates RIG-I-Mediated Type I Interferon Production in a STING-Independent Manner. DNA Cell Biol 2024; 43:197-205. [PMID: 38466944 DOI: 10.1089/dna.2023.0362] [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: 03/13/2024] Open
Abstract
Previous studies have shown that interferon gene-stimulating protein (STING) is essential for IFN-γ-inducible protein 16 (IFI16) as the DNA sensor and RNA sensor to induce transcription of type I interferon (IFN-I) and is essential for IFI16 to synergize with DNA sensor GMP-AMP (cGAMP) synthase (cGAS) in induction of IFN-I transcription. While other and our previous studies have shown that IFI16 enhanced retinoic acid-inducible gene I (RIG-I)-, which was an RNA sensor, and mitochondrial antiviral signaling (MAVS)-, which was the adaptor protein of RIG-I, induced production of IFN-I, so we wonder whether IFI16 regulates the signal pathway of RNA-RIG-I-MAVS-IFN-I in a STING-dependent manner. We used HEK 293T cells, which did not express endogenous STING and were unable to mount an innate immune response upon DNA transfection and found that IFI16 could enhance RIG-I- and MAVS-mediated induction of IFN-I in a STING-independent way. Furthermore, we found that upregulation of the expression of NF-kappa-B essential modulator (NEMO) by IFI16 was not the mechanism that IFI16 regulated the induction of IFN-I. In conclusion, we found that IFI16 regulated the signal pathway of RNA-RIG-I-MAVS-IFN-I in a STING-independent manner.
Collapse
Affiliation(s)
- Xibao Shi
- College of Life Sciences, Henan Normal University, Xinxiang, China
| | - Menglu Wei
- College of Life Sciences, Henan Normal University, Xinxiang, China
| | - Yuwen Feng
- College of Life Sciences, Henan Normal University, Xinxiang, China
| | - Yuanhao Yang
- College of Life Sciences, Henan Normal University, Xinxiang, China
| | - Xiaozhuan Zhang
- College of Life Sciences, Henan Normal University, Xinxiang, China
| | - Hao Chen
- College of Life Sciences, Henan Normal University, Xinxiang, China
| | - Yuqi Xing
- College of Life Sciences, Henan Normal University, Xinxiang, China
| | - Keqi Wang
- College of Life Sciences, Henan Normal University, Xinxiang, China
| | - Wensheng Wang
- College of Life Sciences, Henan Normal University, Xinxiang, China
| | - Li Wang
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Aiping Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Gaiping Zhang
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
- School of Advanced Agricultural Sciences, Peking University, Beijing, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou, China
| |
Collapse
|
25
|
Chengcheng Z, Qingqing Z, Xiaomiao H, Wei L, Xiaorong Z, Yantao W. IFI16 plays a critical role in avian reovirus induced cellular immunosuppression and suppresses virus replication. Poult Sci 2024; 103:103506. [PMID: 38335672 PMCID: PMC10869280 DOI: 10.1016/j.psj.2024.103506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/20/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Avian reovirus (ARV), which commonly induces viral arthritis or tenosynovitis and immunosuppression in chickens, is associated with the nonstructural protein p17 that plays a crucial role in viral replication and regulates cellular signaling pathways through its interaction with cellular proteins. In our previous study, we identified the host protein IFN-γ-inducible protein-16 (IFI16) as an interacting partner of ARV p17 through yeast two-hybrid screening. In the current study, we further confirmed the interaction between IFI16 and p17 protein using coimmunoprecipitation, glutathione S-transferase (GST)-pulldown assay, and laser confocal microscopy techniques. Additionally, we found that the amino acid of p1761-119 is responsible for mediating the interaction with the HINa and HINb domains of IFI16. Interestingly, we observed a significant increase in IFI16 expression upon ARV infection or p17 protein exposure. Moreover, the replication of ARV was found to be largely influenced by the quantity of IFI16 protein. Overexpression of IFI16 led to a significant decrease in ARV replication, while knockdown of the IFI16 expression led to the contrary result. Additionally, our findings demonstrate that IFI16 plays a crucial role in the induction of inflammatory cytokines IFN-β and IL-1β during ARV infection as confirmed by qRT-PCR and ELISA analyses. In conclusion, our study provides novel insights into the functional role of p17 protein and the pathogenic mechanism underlying ARV infection, particularly its association with inflammatory response. Furthermore, it offers new perspectives for identifying potential therapeutic targets against ARV infection.
Collapse
Affiliation(s)
- Zhang Chengcheng
- College of Veterinary Medicine, Yangzhou University; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu, 225009, China
| | - Zhang Qingqing
- College of Veterinary Medicine, Yangzhou University; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu, 225009, China
| | - Hu Xiaomiao
- Yangzhou Vocational University, Yangzhou 225009, China
| | - Li Wei
- Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences/Livestock and Poultry Epidemic Diseases Research Center of Anhui Province/Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Hefei, Anhui 230031, China
| | - Zhang Xiaorong
- College of Veterinary Medicine, Yangzhou University; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu, 225009, China
| | - Wu Yantao
- College of Veterinary Medicine, Yangzhou University; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, Jiangsu, 225009, China.
| |
Collapse
|
26
|
Wang M, Deng H, Chen Y, Wang Y, Zhang Y, Liu C, Zhang M, Li T, Dang S, Li Y. Expression and clinical significance of pattern recognition receptor-associated genes in hand, foot and mouth disease. ASIAN PAC J TROP MED 2024; 17:173-183. [DOI: 10.4103/apjtm.apjtm_876_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/20/2024] [Indexed: 11/27/2024] Open
Abstract
Objective:
To explore which pattern recognition receptors (PRRs) play a key role in the development of hand, foot, and mouth disease (HFMD) by analyzing PRR-associated genes.
Methods:
We conducted a comparative analysis of PRR-associated gene expression in human peripheral blood mononuclear cells (PBMCs) infected with enterovirus 71 (EV-A71) which were derived from patients with HFMD of different severities and at different stages. A total of 30 PRR-associated genes were identified as significantly upregulated both over time and across different EV-A71 isolates. Subsequently, ELISA was employed to quantify the expression of the six most prominent genes among these 30 identified genes, specifically, BST2, IRF7, 1FI16, TRIM21, MX1, and DDX58.
Results:
Compared with those at the recovery stage, the expression levels of BST2 (P=0.027), IFI16 (P=0.016), MX1 (P=0.046) and DDX58 (P=0.008) in the acute stage of infection were significantly upregulated, while no significant difference in the expression levels of IRF7 (P=0.495) and TRIM21 (P=0.071) was found between different stages of the disease. The expression levels of BST2, IRF7, IFI16 and MX1 were significantly higher in children infected with single pathogen than those infected with mixed pathogens, and BST2, IRF7, IFI16 and MX1 expression levels were significantly lower in coxsackie B virus (COXB) positive patients than the negative patients. Expression levels of one or more of BST2, IRF7, IFI16, TRIM21, MX1 and DDX58 genes were correlated with PCT levels, various white blood cell counts, and serum antibody levels that reflect disease course of HFMD. Aspartate aminotransferase was correlated with BST2, MX1 and DDX58 expression levels.
Conclusions:
PRR-associated genes likely initiate the immune response in patients at the acute stage of HFMD.
Collapse
Affiliation(s)
- Muqi Wang
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Huiling Deng
- Department of Pediatrics, Xi'an Central Hospital, Xi'an 710004, China
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an 710003, China
| | - Yuan Chen
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an 710003, China
| | - Yikai Wang
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yufeng Zhang
- Department of Infectious Diseases, Xi'an Children's Hospital, Xi'an 710003, China
| | - Chenrui Liu
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Meng Zhang
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Ting Li
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Shuangsuo Dang
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Yaping Li
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
27
|
Yang R, Pan M, Guo J, Huang Y, Zhang QC, Deng T, Wang J. Mapping of the influenza A virus genome RNA structure and interactions reveals essential elements of viral replication. Cell Rep 2024; 43:113833. [PMID: 38416642 DOI: 10.1016/j.celrep.2024.113833] [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: 05/31/2023] [Revised: 12/04/2023] [Accepted: 02/02/2024] [Indexed: 03/01/2024] Open
Abstract
Influenza A virus (IAV) represents a constant public health threat. The single-stranded, segmented RNA genome of IAV is replicated in host cell nuclei as a series of 8 ribonucleoprotein complexes (vRNPs) with RNA structures known to exert essential function to support viral replication. Here, we investigate RNA secondary structures and RNA interactions networks of the IAV genome and construct an in vivo structure model for each of the 8 IAV genome segments. Our analyses reveal an overall in vivo and in virio resemblance of the IAV genome conformation but also wide disparities among long-range and intersegment interactions. Moreover, we identify a long-range RNA interaction that exerts an essential role in genome packaging. Disrupting this structure displays reduced infectivity, attenuating virus pathogenicity in mice. Our findings characterize the in vivo RNA structural landscape of the IAV genome and reveal viral RNA structures that can be targeted to develop antiviral interventions.
Collapse
Affiliation(s)
- Rui Yang
- The State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Minglei Pan
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jiamei Guo
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yong Huang
- The State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qiangfeng Cliff Zhang
- The State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Tao Deng
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Jianwei Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
28
|
Su R, Li X, Xiao J, Xu J, Tian J, Liu T, Hu Y. UiO-66 nanoparticles combat influenza A virus in mice by activating the RIG-I-like receptor signaling pathway. J Nanobiotechnology 2024; 22:99. [PMID: 38461229 PMCID: PMC10925002 DOI: 10.1186/s12951-024-02358-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 02/20/2024] [Indexed: 03/11/2024] Open
Abstract
The Influenza A virus (IAV) is a zoonotic pathogen that infects humans and various animal species. Infection with IAV can cause fever, anorexia, and dyspnea and is often accompanied by pneumonia characterized by an excessive release of cytokines (i.e., cytokine storm). Nanodrug delivery systems and nanoparticles are a novel approach to address IAV infections. Herein, UiO-66 nanoparticles (NPs) are synthesized using a high-temperature melting reaction. The in vitro and in vivo optimal concentrations of UiO-66 NPs for antiviral activity are 200 μg mL-1 and 60 mg kg-1, respectively. Transcriptome analysis revealed that UiO-66 NPs can activate the RIG-I-like receptor signaling pathway, thereby enhancing the downstream type I interferon antiviral effect. These NPs suppress inflammation-related pathways, including the FOXO, HIF, and AMPK signaling pathways. The inhibitory effect of UiO-66 NPs on the adsorption and entry of IAV into A549 cells is significant. This study presents novel findings that demonstrate the effective inhibition of IAV adsorption and entry into cells via UiO-66 NPs and highlights their ability to activate the cellular RIG-I-like receptor signaling pathway, thereby exerting an anti-IAV effect in vitro or in mice. These results provide valuable insights into the mechanism of action of UiO-66 NPs against IAV and substantial data for advancing innovative antiviral nanomedicine.
Collapse
Affiliation(s)
- Ruijing Su
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Xinsen Li
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Jin Xiao
- Key Laboratory of Veterinary Bioproduction and Chemical Medicine of the Ministry of Agriculture, Zhongmu Institutes of China Animal Husbandry Industry Co., Ltd, Beijing, People's Republic of China
| | - Jiawei Xu
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Jijing Tian
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China
| | - Tianlong Liu
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China.
| | - Yanxin Hu
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193, China.
| |
Collapse
|
29
|
He W, Mu X, Wu X, Liu Y, Deng J, Liu Y, Han F, Nie X. The cGAS-STING pathway: a therapeutic target in diabetes and its complications. BURNS & TRAUMA 2024; 12:tkad050. [PMID: 38312740 PMCID: PMC10838060 DOI: 10.1093/burnst/tkad050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/22/2023] [Accepted: 10/09/2023] [Indexed: 02/06/2024]
Abstract
Diabetic wound healing (DWH) represents a major complication of diabetes where inflammation is a key impediment to proper healing. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway has emerged as a central mediator of inflammatory responses to cell stress and damage. However, the contribution of cGAS-STING activation to impaired healing in DWH remains understudied. In this review, we examine the evidence that cGAS-STING-driven inflammation is a critical factor underlying defective DWH. We summarize studies revealing upregulation of the cGAS-STING pathway in diabetic wounds and discuss how this exacerbates inflammation and senescence and disrupts cellular metabolism to block healing. Partial pharmaceutical inhibition of cGAS-STING has shown promise in damping inflammation and improving DWH in preclinical models. We highlight key knowledge gaps regarding cGAS-STING in DWH, including its relationships with endoplasmic reticulum stress and metal-ion signaling. Elucidating these mechanisms may unveil new therapeutic targets within the cGAS-STING pathway to improve healing outcomes in DWH. This review synthesizes current understanding of how cGAS-STING activation contributes to DWH pathology and proposes future research directions to exploit modulation of this pathway for therapeutic benefit.
Collapse
Affiliation(s)
- Wenjie He
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| | - Xingrui Mu
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| | - Xingqian Wu
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| | - Ye Liu
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| | - Junyu Deng
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| | - Yiqiu Liu
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| | - Felicity Han
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xuqiang Nie
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- College of Pharmacy, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, No. 6 Xuefu West Road, Xinpu New District, Zunyi 563006, China
| |
Collapse
|
30
|
Silva RCMC, Gomes FM. Evolution of the Major Components of Innate Immunity in Animals. J Mol Evol 2024; 92:3-20. [PMID: 38281163 DOI: 10.1007/s00239-024-10155-2] [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: 10/28/2023] [Accepted: 01/11/2024] [Indexed: 01/30/2024]
Abstract
Innate immunity is present in all animals. In this review, we explore the main conserved mechanisms of recognition and innate immune responses among animals. In this sense, we discuss the receptors, critical for binding to pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs); the downstream signaling proteins; and transcription factors that govern immune responses. We also highlight conserved inflammatory mediators that are induced after the recognition of DAMPs and PAMPs. At last, we discuss the mechanisms that are involved in the regulation and/or generation of reactive oxygen species (ROS), influencing immune responses, like heme-oxygenases (HOs).
Collapse
Affiliation(s)
- Rafael Cardoso Maciel Costa Silva
- Laboratory of Immunoreceptors and Signaling, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Fábio Mendonça Gomes
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
31
|
Husain M. Influenza Virus Host Restriction Factors: The ISGs and Non-ISGs. Pathogens 2024; 13:127. [PMID: 38392865 PMCID: PMC10893265 DOI: 10.3390/pathogens13020127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/18/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Influenza virus has been one of the most prevalent and researched viruses globally. Consequently, there is ample information available about influenza virus lifecycle and pathogenesis. However, there is plenty yet to be known about the determinants of influenza virus pathogenesis and disease severity. Influenza virus exploits host factors to promote each step of its lifecycle. In turn, the host deploys antiviral or restriction factors that inhibit or restrict the influenza virus lifecycle at each of those steps. Two broad categories of host restriction factors can exist in virus-infected cells: (1) encoded by the interferon-stimulated genes (ISGs) and (2) encoded by the constitutively expressed genes that are not stimulated by interferons (non-ISGs). There are hundreds of ISGs known, and many, e.g., Mx, IFITMs, and TRIMs, have been characterized to restrict influenza virus infection at different stages of its lifecycle by (1) blocking viral entry or progeny release, (2) sequestering or degrading viral components and interfering with viral synthesis and assembly, or (3) bolstering host innate defenses. Also, many non-ISGs, e.g., cyclophilins, ncRNAs, and HDACs, have been identified and characterized to restrict influenza virus infection at different lifecycle stages by similar mechanisms. This review provides an overview of those ISGs and non-ISGs and how the influenza virus escapes the restriction imposed by them and aims to improve our understanding of the host restriction mechanisms of the influenza virus.
Collapse
Affiliation(s)
- Matloob Husain
- Department of Microbiology and Immunology, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand
| |
Collapse
|
32
|
Roy A, Ghosh A. Epigenetic Restriction Factors (eRFs) in Virus Infection. Viruses 2024; 16:183. [PMID: 38399958 PMCID: PMC10892949 DOI: 10.3390/v16020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
The ongoing arms race between viruses and their hosts is constantly evolving. One of the ways in which cells defend themselves against invading viruses is by using restriction factors (RFs), which are cell-intrinsic antiviral mechanisms that block viral replication and transcription. Recent research has identified a specific group of RFs that belong to the cellular epigenetic machinery and are able to restrict the gene expression of certain viruses. These RFs can be referred to as epigenetic restriction factors or eRFs. In this review, eRFs have been classified into two categories. The first category includes eRFs that target viral chromatin. So far, the identified eRFs in this category include the PML-NBs, the KRAB/KAP1 complex, IFI16, and the HUSH complex. The second category includes eRFs that target viral RNA or, more specifically, the viral epitranscriptome. These epitranscriptomic eRFs have been further classified into two types: those that edit RNA bases-adenosine deaminase acting on RNA (ADAR) and pseudouridine synthases (PUS), and those that covalently modify viral RNA-the N6-methyladenosine (m6A) writers, readers, and erasers. We delve into the molecular machinery of eRFs, their role in limiting various viruses, and the mechanisms by which viruses have evolved to counteract them. We also examine the crosstalk between different eRFs, including the common effectors that connect them. Finally, we explore the potential for new discoveries in the realm of epigenetic networks that restrict viral gene expression, as well as the future research directions in this area.
Collapse
Affiliation(s)
- Arunava Roy
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA;
| | | |
Collapse
|
33
|
Yao J, Sterling K, Wang Z, Zhang Y, Song W. The role of inflammasomes in human diseases and their potential as therapeutic targets. Signal Transduct Target Ther 2024; 9:10. [PMID: 38177104 PMCID: PMC10766654 DOI: 10.1038/s41392-023-01687-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 09/18/2023] [Accepted: 10/13/2023] [Indexed: 01/06/2024] Open
Abstract
Inflammasomes are large protein complexes that play a major role in sensing inflammatory signals and triggering the innate immune response. Each inflammasome complex has three major components: an upstream sensor molecule that is connected to a downstream effector protein such as caspase-1 through the adapter protein ASC. Inflammasome formation typically occurs in response to infectious agents or cellular damage. The active inflammasome then triggers caspase-1 activation, followed by the secretion of pro-inflammatory cytokines and pyroptotic cell death. Aberrant inflammasome activation and activity contribute to the development of diabetes, cancer, and several cardiovascular and neurodegenerative disorders. As a result, recent research has increasingly focused on investigating the mechanisms that regulate inflammasome assembly and activation, as well as the potential of targeting inflammasomes to treat various diseases. Multiple clinical trials are currently underway to evaluate the therapeutic potential of several distinct inflammasome-targeting therapies. Therefore, understanding how different inflammasomes contribute to disease pathology may have significant implications for developing novel therapeutic strategies. In this article, we provide a summary of the biological and pathological roles of inflammasomes in health and disease. We also highlight key evidence that suggests targeting inflammasomes could be a novel strategy for developing new disease-modifying therapies that may be effective in several conditions.
Collapse
Affiliation(s)
- Jing Yao
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yun Zhang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.
| | - Weihong Song
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Zhejiang Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and The Affiliated Kangning Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
34
|
Hoyer A, Chakraborty S, Lilienthal I, Konradsen JR, Katayama S, Söderhäll C. The functional role of CST1 and CCL26 in asthma development. Immun Inflamm Dis 2024; 12:e1162. [PMID: 38270326 PMCID: PMC10797655 DOI: 10.1002/iid3.1162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 12/18/2023] [Accepted: 01/10/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Asthma is the most common chronic disease in children with an increasing prevalence. Its development is caused by genetic and environmental factors and allergic sensitization is a known trigger. Dog allergens affect up to 30% of all children and dog dander-sensitized children show increased expression of cystatin-1 (CST1) and eotaxin-3 (CCL26) in nasal epithelium. The aim of our study was to investigate the functional mechanism of CST1 and CCL26 in the alveolar basal epithelial cell line A549. METHODS A549 cells were transfected with individual overexpression vectors for CST1 and CCL26 and RNA sequencing was performed to examine the transcriptomics. edgeR was used to identify differentially expressed genes (= DEG, |log2 FC | ≥ 2, FDR < 0.01). The protein expression levels of A549 cells overexpressing CST1 and CCL26 were analyzed using the Target 96 inflammation panel from OLINK (antibody-mediated proximity extension-based assay; OLINK Proteomics). Differentially expressed proteins were considered with a |log2 FC| ≥ 1, p < .05. RESULTS The overexpression of CST1 resulted in a total of 27 DEG (1 upregulated and 26 downregulated) and the overexpression of CCL26 in a total of 137 DEG (0 upregulated and 137 downregulated). The gene ontology enrichment analysis showed a significant downregulation of type I and III interferon signaling pathway genes as well as interferon-stimulated genes. At the protein level, overexpression of CST1 induced a significantly increased expression of CCL3, whereas CCL26 overexpression led to increased expression of HGF, and a decrease of CXCL11, CCL20, CCL3 and CXCL10. CONCLUSION Our results indicate that an overexpression of CST1 and CCL26 cause a downregulation of interferon related genes and inflammatory proteins. It might cause a higher disease susceptibility, mainly for allergic asthma, as CCL26 is an agonist for CCR-3-carrying cells, such as eosinophils and Th2 lymphocytes, mostly active in allergic asthma.
Collapse
Affiliation(s)
- Angela Hoyer
- Department of Women's and Children's HealthKarolinska InstitutetSolnaSweden
- Astrid Lindgren Children's HospitalKarolinska University HospitalSolnaSweden
| | - Sandip Chakraborty
- Department of Women's and Children's HealthKarolinska InstitutetSolnaSweden
- Astrid Lindgren Children's HospitalKarolinska University HospitalSolnaSweden
| | - Ingrid Lilienthal
- Childhood Cancer Research Unit, Department of Women's and Children's HealthKarolinska InstitutetSolnaSweden
| | - Jon R. Konradsen
- Department of Women's and Children's HealthKarolinska InstitutetSolnaSweden
- Astrid Lindgren Children's HospitalKarolinska University HospitalSolnaSweden
| | - Shintaro Katayama
- Department of Biosciences and NutritionKarolinska InstitutetHuddingeSweden
- Stem Cells and Metabolism Research ProgramUniversity of HelsinkiHelsinkiFinland
- Folkhälsan Research CenterHelsinkiFinland
| | - Cilla Söderhäll
- Department of Women's and Children's HealthKarolinska InstitutetSolnaSweden
- Astrid Lindgren Children's HospitalKarolinska University HospitalSolnaSweden
| |
Collapse
|
35
|
Meyer L, Duquénois I, Gellenoncourt S, Pellerin M, Marcadet-Hauss A, Pavio N, Doceul V. Identification of interferon-stimulated genes with modulated expression during hepatitis E virus infection in pig liver tissues and human HepaRG cells. Front Immunol 2023; 14:1291186. [PMID: 38058490 PMCID: PMC10696647 DOI: 10.3389/fimmu.2023.1291186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023] Open
Abstract
Introduction Hepatitis E virus (HEV) is a common cause of enterically transmitted acute hepatitis worldwide. The virus is transmitted by the fecal-oral route via the consumption of contaminated water supplies and is also a zoonotic foodborne pathogen. Swine are the main reservoir of zoonotic HEV. In humans, HEV infection is usually asymptomatic or causes acute hepatitis that is self-limited. However, fulminant hepatic failure and chronic cases of HEV infection can occur in some patients. In contrast, HEV infection in pigs remains asymptomatic, although the virus replicates efficiently, suggesting that swine are able to control the virus pathogenesis. Upon viral infection, IFN is secreted and activates cellular pathways leading to the expression of many IFN-stimulated genes (ISGs). ISGs can restrict the replication of specific viruses and establish an antiviral state within infected and neighboring cells. Methods In this study, we used PCR arrays to determine the expression level of up to 168 ISGs and other IFN-related genes in the liver tissues of pigs infected with zoonotic HEV-3c and HEV-3f and in human bipotent liver HepaRG cells persistently infected with HEV-3f. Results and discussion The expression of 12 and 25 ISGs was found to be up-regulated in infected swine livers and HepaRG cells, respectively. The expression of CXCL10, IFIT2, MX2, OASL and OAS2 was up-regulated in both species. Increased expression of IFI16 mRNA was also found in swine liver tissues. This study contributes to the identification of potential ISGs that could play a role in the control or persistence of HEV infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Virginie Doceul
- Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Agence Nationale de Sécurité Sanitaire de l’Alimentation, de l’Environnement et du Travail (ANSES), École Nationale Vétérinaire d'Alfort (ENVA), UMR Virology, Maisons-Alfort, France
| |
Collapse
|
36
|
Kong LZ, Kim SM, Wang C, Lee SY, Oh SC, Lee S, Jo S, Kim TD. Understanding nucleic acid sensing and its therapeutic applications. Exp Mol Med 2023; 55:2320-2331. [PMID: 37945923 PMCID: PMC10689850 DOI: 10.1038/s12276-023-01118-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 11/12/2023] Open
Abstract
Nucleic acid sensing is involved in viral infections, immune response-related diseases, and therapeutics. Based on the composition of nucleic acids, nucleic acid sensors are defined as DNA or RNA sensors. Pathogen-associated nucleic acids are recognized by membrane-bound and intracellular receptors, known as pattern recognition receptors (PRRs), which induce innate immune-mediated antiviral responses. PRR activation is tightly regulated to eliminate infections and prevent abnormal or excessive immune responses. Nucleic acid sensing is an essential mechanism in tumor immunotherapy and gene therapies that target cancer and infectious diseases through genetically engineered immune cells or therapeutic nucleic acids. Nucleic acid sensing supports immune cells in priming desirable immune responses during tumor treatment. Recent studies have shown that nucleic acid sensing affects the efficiency of gene therapy by inhibiting translation. Suppression of innate immunity induced by nucleic acid sensing through small-molecule inhibitors, virus-derived proteins, and chemical modifications offers a potential therapeutic strategy. Herein, we review the mechanisms and regulation of nucleic acid sensing, specifically covering recent advances. Furthermore, we summarize and discuss recent research progress regarding the different effects of nucleic acid sensing on therapeutic efficacy. This study provides insights for the application of nucleic acid sensing in therapy.
Collapse
Affiliation(s)
- Ling-Zu Kong
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Seok-Min Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Chunli Wang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Soo Yun Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Se-Chan Oh
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Sunyoung Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
- Department of Life Sciences, Korea University, Seoul, 02841, Korea
| | - Seona Jo
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Korea
| | - Tae-Don Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34113, Korea.
- Biomedical Mathematics Group, Institute for Basic Science (IBS), Daejeon, Republic of Korea.
- Department of Biopharmaceutical Convergence, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
37
|
Garrett TJ, Coatsworth H, Mahmud I, Hamerly T, Stephenson CJ, Ayers JB, Yazd HS, Miller MR, Lednicky JA, Dinglasan RR. Niclosamide as a chemical probe for analyzing SARS-CoV-2 modulation of host cell lipid metabolism. Front Microbiol 2023; 14:1251065. [PMID: 37901834 PMCID: PMC10603251 DOI: 10.3389/fmicb.2023.1251065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/15/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction SARS-CoV-2 subverts host cell processes to facilitate rapid replication and dissemination, and this leads to pathological inflammation. Methods We used niclosamide (NIC), a poorly soluble anti-helminth drug identified initially for repurposed treatment of COVID-19, which activates the cells' autophagic and lipophagic processes as a chemical probe to determine if it can modulate the host cell's total lipid profile that would otherwise be either amplified or reduced during SARS-CoV-2 infection. Results Through parallel lipidomic and transcriptomic analyses we observed massive reorganization of lipid profiles of SARS-CoV-2 infected Vero E6 cells, especially with triglycerides, which were elevated early during virus replication, but decreased thereafter, as well as plasmalogens, which were elevated at later timepoints during virus replication, but were also elevated under normal cell growth. These findings suggested a complex interplay of lipid profile reorganization involving plasmalogen metabolism. We also observed that NIC treatment of both low and high viral loads does not affect virus entry. Instead, NIC treatment reduced the abundance of plasmalogens, diacylglycerides, and ceramides, which we found elevated during virus infection in the absence of NIC, resulting in a significant reduction in the production of infectious virions. Unexpectedly, at higher viral loads, NIC treatment also resulted in elevated triglyceride levels, and induced significant changes in phospholipid metabolism. Discussion We posit that future screens of approved or new partner drugs should prioritize compounds that effectively counter SARS-CoV-2 subversion of lipid metabolism, thereby reducing virus replication, egress, and the subsequent regulation of key lipid mediators of pathological inflammation.
Collapse
Affiliation(s)
- Timothy J. Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
- Southeast Center for Integrated Metabolomics, Clinical and Translational Science Institute, University of Florida, Gainesville, FL, United States
| | - Heather Coatsworth
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Iqbal Mahmud
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
- Southeast Center for Integrated Metabolomics, Clinical and Translational Science Institute, University of Florida, Gainesville, FL, United States
| | - Timothy Hamerly
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Caroline J. Stephenson
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Jasmine B. Ayers
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - Hoda S. Yazd
- Department of Chemistry, University of Florida, Gainesville, FL, United States
| | - Megan R. Miller
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - John A. Lednicky
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Rhoel R. Dinglasan
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
38
|
Yan Q, Zhou J, Wang Z, Ding X, Ma X, Li W, Jia X, Gao SJ, Lu C. NAT10-dependent N 4-acetylcytidine modification mediates PAN RNA stability, KSHV reactivation, and IFI16-related inflammasome activation. Nat Commun 2023; 14:6327. [PMID: 37816771 PMCID: PMC10564894 DOI: 10.1038/s41467-023-42135-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 09/28/2023] [Indexed: 10/12/2023] Open
Abstract
N-acetyltransferase 10 (NAT10) is an N4-acetylcytidine (ac4C) writer that catalyzes RNA acetylation at cytidine N4 position on tRNAs, rRNAs and mRNAs. Recently, NAT10 and the associated ac4C have been reported to increase the stability of HIV-1 transcripts. Here, we show that NAT10 catalyzes ac4C addition to the polyadenylated nuclear RNA (PAN), a long non-coding RNA encoded by the oncogenic DNA virus Kaposi's sarcoma-associated herpesvirus (KSHV), triggering viral lytic reactivation from latency. Mutagenesis of ac4C sites in PAN RNA in the context of KSHV infection abolishes PAN ac4C modifications, downregulates the expression of viral lytic genes and reduces virion production. NAT10 knockdown or mutagenesis erases ac4C modifications of PAN RNA and increases its instability, and prevents KSHV reactivation. Furthermore, PAN ac4C modification promotes NAT10 recruitment of IFN-γ-inducible protein-16 (IFI16) mRNA, resulting in its ac4C acetylation, mRNA stability and translation, and eventual inflammasome activation. These results reveal a novel mechanism of viral and host ac4C modifications and the associated complexes as a critical switch of KSHV replication and antiviral immunity.
Collapse
Affiliation(s)
- Qin Yan
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, 210004, Nanjing, P. R. China
- Department of Microbiology, Nanjing Medical University, 211166, Nanjing, P. R. China
- Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, P. R. China
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, 211166, Nanjing, P. R. China
| | - Jing Zhou
- Department of Microbiology, Nanjing Medical University, 211166, Nanjing, P. R. China
| | - Ziyu Wang
- Department of Microbiology, Nanjing Medical University, 211166, Nanjing, P. R. China
| | - Xiangya Ding
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, 210004, Nanjing, P. R. China
| | - Xinyue Ma
- Department of Microbiology, Nanjing Medical University, 211166, Nanjing, P. R. China
| | - Wan Li
- Department of Microbiology, Nanjing Medical University, 211166, Nanjing, P. R. China
- Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, P. R. China
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, 211166, Nanjing, P. R. China
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, 210004, Nanjing, P. R. China.
| | - Shou-Jiang Gao
- Tumor Virology Program, UPMC Hillman Cancer Center, and Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, 15232, USA
| | - Chun Lu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, 210004, Nanjing, P. R. China.
- Department of Microbiology, Nanjing Medical University, 211166, Nanjing, P. R. China.
- Changzhou Medical Center, Nanjing Medical University, 211166, Nanjing, P. R. China.
- Key Laboratory of Pathogen Biology of Jiangsu Province, Nanjing Medical University, 211166, Nanjing, P. R. China.
| |
Collapse
|
39
|
Alalem M, Dabous E, Awad AM, Alalem N, Guirgis AA, El-Masry S, Khalil H. Influenza a virus regulates interferon signaling and its associated genes; MxA and STAT3 by cellular miR-141 to ensure viral replication. Virol J 2023; 20:183. [PMID: 37596622 PMCID: PMC10439583 DOI: 10.1186/s12985-023-02146-4] [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: 04/26/2023] [Accepted: 07/28/2023] [Indexed: 08/20/2023] Open
Abstract
The antiviral response against influenza A virus (IAV) infection includes the induction of the interferon (IFN) signaling pathway, including activation of the STATs protein family. Subsequently, antiviral myxovirus resistance (MxA) protein and other interferon-stimulated genes control virus replication; however, the molecular interaction of viral-mediated IFN signaling needs more investigation. Host microRNAs (miRNAs) are small non-coding molecules that posttranscriptionally regulate gene expression. Here, we sought to investigate the possible involvement of miR-141 in IAV-mediated IFN signaling. Accordingly, the microarray analysis of A549 cells transfected with precursor miR-141 (pre-miR-141) was used to capture the potentially regulated genes in response to miR-141 overexpression independent of IAV infection. The downregulation of targeted genes by miR-141, in addition to viral gene expression, was investigated by quantitative real-time PCR, western blot analysis, and flow cytometric assay. Our findings showed a significant upregulation of miR-141 in infected A549 cells with different strains of IAV. Notably, IAV replication was firmly interrupted in cells transfected with the miR-141 inhibitor. While its replication significantly increased in cells transfected with pre-miR-141 confirming the crucial role of miRNA-141 in supporting virus replication. Interestingly, the microarray data of miR-141 transduced A549 cells showed many downregulated genes, including MxA, STAT3, IFI27, and LAMP3. The expression profile of MxA and STAT3 was significantly depleted in infected cells transfected with the pre-miR-141, while their expression was restored in infected cells transfected with the miR-141 inhibitor. Unlike interleukin 6 (IL-6), the production of IFN-β markedly decreased in infected cells that transfected with pre-miR-141, while it significantly elevated in infected cells transfected with miR-141 inhibitor. These data provide evidence for the crucial role of miR-141 in regulating the antiviral gene expression induced by IFN and IL-6 signaling during IAV infection to ensure virus replication.
Collapse
Affiliation(s)
- Mai Alalem
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, 79, Egypt
| | - Emad Dabous
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, 79, Egypt
| | - Ahmed M Awad
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, 79, Egypt
| | - Nedaa Alalem
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, 79, Egypt
| | - Adel A Guirgis
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, 79, Egypt
| | - Samir El-Masry
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, 79, Egypt
| | - Hany Khalil
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, 79, Egypt.
| |
Collapse
|
40
|
Fu Y, Zhan X, You X, Nie D, Mai H, Chen Y, He S, Sheng J, Zeng Z, Li H, Li J, Hu S. USP12 promotes antiviral responses by deubiquitinating and stabilizing IFI16. PLoS Pathog 2023; 19:e1011480. [PMID: 37410794 PMCID: PMC10353808 DOI: 10.1371/journal.ppat.1011480] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/18/2023] [Accepted: 06/13/2023] [Indexed: 07/08/2023] Open
Abstract
Deubiquitinating enzymes (DUBs) regulate antiviral immune response through targeting DNA sensor signaling pathway members. As one of the DNA sensors, interferon (IFN)-γ inducible protein 16 (IFI16) play a major role in response to virus infections through activating the canonical STING/TBK-1/IRF3 signaling pathway. Only a few studies discuss the function of DUBs in IFI16-mediated antiviral response. Ubiquitin-specific protease 12 (USP12), which is one of the major members of the USP family, participates in various biological functions. However, whether USP12 regulates the nucleic acid sensor to modulate antiviral immune responses has not yet been elucidated. In this study, we found that knockout or knockdown of USP12 impaired the HSV-1-induced expressions of IFN-β, CCL-5, IL-6, and downstream interferon-stimulated genes (ISGs). Moreover, USP12 deficiency increased HSV-1 replication and host susceptibility to HSV-1 infection. Mechanistically, USP12 inhibited the proteasome-dependent degradation of IFI16 through its deubiquitinase activity, thereby maintaining IFI16 stability and promoting IFI16-STING-IRF3- and p65-mediated antiviral signaling. Overall, our findings demonstrate an essential role of USP12 in DNA-sensing signaling and contribute to the understanding of deubiquitination-mediated regulation of innate antiviral responses.
Collapse
Affiliation(s)
- Yuling Fu
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiaoxia Zhan
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaolong You
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Dingnai Nie
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Haiyan Mai
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yitian Chen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Shitong He
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Junli Sheng
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Zhijie Zeng
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hongwei Li
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jinlong Li
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Shengfeng Hu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
41
|
Gallegos-Rodarte C, Escobar-Chavarría O, Cantera-Bravo MM, Sarmiento-Silva RE, Benitez-Guzman A. NLRP3 Inflammasome Involved with Viral Replication in Cytopathic NADL BVDV Infection and IFI16 Inflammasome Connected with IL-1β Release in Non-Cytopathic NY-1 BVDV Infection in Bovine Macrophages. Viruses 2023; 15:1494. [PMID: 37515181 PMCID: PMC10386432 DOI: 10.3390/v15071494] [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: 05/01/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Inflammasomes are multiprotein complexes that play a role in the processing of proinflammatory cytokines such as interleukin 1 beta (IL-1β). The secretion of IL-1β in bovine macrophages infected with the bovine viral diarrhea virus (BVDV) cytopathic strain NADL (NADLcp-BVDV) is caspase 1-dependent. In the present study, we found that in macrophages infected with NADL, the NLRP3 inflammasome participated in the maturation of IL-1β as the level decreased from 4629.3 pg/mL to 897.0 pg/mL after treatment with cytokine release inhibitory drug 3 (CRID3). Furthermore, NLRP3 activation has implications regarding viral replication, as there was a decrease in the viral titer until 1 log of a supernatant of macrophages that were inhibited with CRID3 remained. In the case of the non-cytopathic BVDV strain NY-1 (NY-1 ncpBVDV), IL-1β secretion is not affected by NLRP3, but could be related to the IFI16 inflammasome; we found a colocalization of IFI16 with ASC using confocal microscopy in infected macrophages with the NY-1 ncp-BVDV biotype. To relate IFI16 activation to IL-1β release, we used ODN TTAGGG (A151), a competitive inhibitor of IFI16; the results show a decrease in its level from 248 pg/mL to 128.3 pg/mL. Additionally, we evaluated the caspase 1 activation downstream of IFI16 and found a decrease in the IL-1β from 252.9 pg/mL to 63.5 pg/mL when caspase 1 was inhibited with Y-VAD. Our results provide an improved understanding of the mechanisms involved in the viral replication, inflammation and pathogenesis of bovine viral diarrhea.
Collapse
Affiliation(s)
| | | | | | | | - Alejandro Benitez-Guzman
- Departamento de Microbiología e Inmunología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (C.G.-R.); (O.E.-C.); (M.M.C.-B.); (R.E.S.-S.)
| |
Collapse
|
42
|
Fu C, Zhu W, Cao N, Liu W, Lu Z, Wong Z, Guan K, Hu C, Han B, Zeng S, Fan S. Role of CIV NS1 Protein in Innate Immunity and Viral Replication. Int J Mol Sci 2023; 24:10056. [PMID: 37373204 DOI: 10.3390/ijms241210056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/23/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
The innate immune pathway serves as the first line of defense against viral infections and plays a crucial role in the host's immune response in clearing viruses. Prior research has indicated that the influenza A virus has developed various strategies to avoid host immune responses. Nevertheless, the role of the NS1 protein of the canine influenza virus (CIV) in the innate immune pathway remains unclear. In this study, eukaryotic plasmids of NS1, NP, PA, PB1, and PB2 were constructed, and it was found that these proteins interact with melanoma differentiation-associated gene 5 (MDA5) and antagonize the activation of IFN-β promoters by MDA5. We selected the NS1 protein for further study and found that NS1 does not affect the interaction between the viral ribonucleoprotein (RNP) subunit and MDA5, but that it downregulates the expression of the laboratory of genetics and physiology 2 (LGP2) and retinoic acid-inducible gene-I (RIG-I) receptors in the RIG-I pathway. Additionally, NS1 was found to inhibit the expression of several antiviral proteins and cytokines, including MX dynamin like GTPase 1 (MX1), 2'-5'oligoadenylate synthetase (OAS), Signal Transducers and Activators of Transcription (STAT1), tripartite motif 25 (TRIM25), interleukin-2 (IL-2), IFN, IL-8, and IL-1β. To further investigate the role of NS1, a recombinant H3N2 virus strain (rH3N2) and an NS1-null virus (rH3N2ΔNS1) were rescued using reverse-genetic technology. The rH3N2ΔNS1 virus exhibited lower viral titers compared to rH3N2, but had a stronger activation effect on the receptors LGP2 and RIG-I. Furthermore, when compared to rH3N2, rH3N2ΔNS1 exhibited a more pronounced activation of antiviral proteins such as MX1, OAS, STAT1, and TRIM25, as well as antiviral cytokines such as IL-6, IFN-β, and IL-1β. These findings suggest a new mechanism by which NS1, a nonstructural protein of CIV, facilitates innate immune signaling and provides new avenues for the development of antiviral strategies.
Collapse
Affiliation(s)
- Cheng Fu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Wenhui Zhu
- College of Veterinary Medicine, South China Agricultural University, No. 483, Wushan Road, Tianhe District, Guangzhou 510000, China
| | - Nan Cao
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Wenjun Liu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Zhier Lu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Ziyuan Wong
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Kaiting Guan
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Chunyan Hu
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Baoting Han
- College of Animal Science & Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510225, China
| | - Sen Zeng
- College of Veterinary Medicine, South China Agricultural University, No. 483, Wushan Road, Tianhe District, Guangzhou 510000, China
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, No. 483, Wushan Road, Tianhe District, Guangzhou 510000, China
| |
Collapse
|
43
|
Huang Y, Abdelgawad A, Turchinovich A, Queen S, Abreu CM, Zhu X, Batish M, Zheng L, Witwer KW. RNA landscapes of brain tissue and brain tissue-derived extracellular vesicles in simian immunodeficiency virus (SIV) infection and SIV-related central nervous system pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.01.535193. [PMID: 37034720 PMCID: PMC10081316 DOI: 10.1101/2023.04.01.535193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Introduction Antiretroviral treatment regimens can effectively control HIV replication and some aspects of disease progression. However, molecular events in end-organ diseases such as central nervous system (CNS) disease are not yet fully understood, and routine eradication of latent reservoirs is not yet in reach. Brain tissue-derived extracellular vesicles (bdEVs) act locally in the source tissue and may indicate molecular mechanisms in HIV CNS pathology. Regulatory RNAs from EVs have emerged as important participants in HIV disease pathogenesis. Using brain tissue and bdEVs from the simian immunodeficiency virus (SIV) model of HIV disease, we profiled messenger RNAs (mRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), seeking to identify possible networks of RNA interaction in SIV infection and neuroinflammation. Methods Postmortem occipital cortex tissue were collected from pigtailed macaques: uninfected controls and SIV-infected subjects (acute phase and chronic phase with or without CNS pathology). bdEVs were separated and characterized in accordance with international consensus standards. RNAs from bdEVs and source tissue were used for sequencing and qPCR to detect mRNA, miRNA, and circRNA levels. Results Multiple dysregulated bdEV RNAs, including mRNAs, miRNAs, and circRNAs, were identified in acute infection and chronic infection with pathology. Most dysregulated mRNAs in bdEVs reflected dysregulation in their source tissues. These mRNAs are disproportionately involved in inflammation and immune responses, especially interferon pathways. For miRNAs, qPCR assays confirmed differential abundance of miR-19a-3p, let-7a-5p, and miR-29a-3p (acute SIV infection), and miR-146a-5p and miR-449a-5p (chronic with pathology) in bdEVs. In addition, target prediction suggested that several circRNAs that were differentially abundant in source tissue might be responsible for specific differences in small RNA levels in bdEVs during SIV infection. Conclusions RNA profiling of bdEVs and source tissues reveals potential regulatory networks in SIV infection and SIV-related CNS pathology.
Collapse
Affiliation(s)
- Yiyao Huang
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ahmed Abdelgawad
- Department of Medical and Molecular Sciences, University of Delaware, Newark, DE, USA
| | - Andrey Turchinovich
- Division of Cancer Genome Research, German Cancer Research Center DKFZ, Heidelberg, Germany
- Heidelberg Biolabs GmbH, Mannheim, Germany
| | - Suzanne Queen
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Celina Monteiro Abreu
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xianming Zhu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mona Batish
- Department of Medical and Molecular Sciences, University of Delaware, Newark, DE, USA
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
44
|
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
|
45
|
Wang S, Li Z, Chen Y, Gao S, Qiao J, Liu H, Song H, Ao D, Sun X. ARIH1 inhibits influenza A virus replication and facilitates RIG-I dependent immune signaling by interacting with SQSTM1/p62. Virol J 2023; 20:58. [PMID: 37005687 PMCID: PMC10066941 DOI: 10.1186/s12985-023-02022-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/24/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND Multiple host factors are involved in modulating type I interferon expression induced by viruses; however, the mechanism is not fully elucidated. Influenza A virus infection causes severe respiratory symptoms and triggers a series of signaling cascades and host innate immune responses, including interferon production. The co-IP/MS technology was used to screen several antiviral factors in the early stage. Among these factors, ariadne-1 homolog (ARIH1) caught our attention. METHODS Western blot assay was performed to detect the level of proteins and software ImageJ was used to analyze the band intensities. Polymerase activity assay was conducted to evaluate the polymerase activity of influenza A virus. Tissue culture infective dose (TCID50) assay was performed to measure influenza A virus titers, and quantitative RT-PCR assay was applied to test the mRNA level of IFN-β, ISG56, and CXCL10. Luciferase reporter assay was used to confirm the target of ARIH1 in RIG-I signaling. Immunoprecipitation assay was performed to detect the interaction and the ubiquitination of the proteins. All data were analyzed by biostatistical methods and presented as means ± standard deviation from three independent experiments. Statistical significance was determined using two-tailed student's t test. A P value of less than 0.05 was considered statistically significant, and a P value of less than 0.01 was considered highly significant (ns, P ≥ 0.05; *, P < 0.05; and **, P < 0.01). RESULTS We found that ARIH1, a member of E3 ubiquitin ligases, enhanced cellular antiviral responses. Subsequent study showed that ARIH1 was up-regulated during influenza A virus infection. Further analysis showed that ARIH1 enhanced IFN-β and downstream gene expression by affecting the degradation of RIG-I through the SQSTM1/p62 signaling pathway. CONCLUSION This newly revealed mechanism shows that cellular response increases of ARIH1 and promotes IFN-β expression to boost host survival during viral infection.
Collapse
Affiliation(s)
- Shengyu Wang
- Key Laboratory of Infectious Disease and Biosafety, Provincial Department of Education, Guizhou, Institute of Life Sciences/ College of Preclinical Medicine, Zunyi Medical University, Zunyi, China
- Department of Microbiology, College of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Zhenrong Li
- Department of Microbiology, College of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Yaping Chen
- Department of Microbiology, College of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Sanli Gao
- Key Laboratory of Infectious Disease and Biosafety, Provincial Department of Education, Guizhou, Institute of Life Sciences/ College of Preclinical Medicine, Zunyi Medical University, Zunyi, China
- Department of Microbiology, College of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Junhua Qiao
- Department of Microbiology, College of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Haoru Liu
- Department of Microbiology, College of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Hong Song
- Department of Microbiology, College of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Dishu Ao
- Department of Microbiology, College of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Xin Sun
- Key Laboratory of Infectious Disease and Biosafety, Provincial Department of Education, Guizhou, Institute of Life Sciences/ College of Preclinical Medicine, Zunyi Medical University, Zunyi, China.
- Department of Microbiology, College of Preclinical Medicine, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
46
|
Zhang D, Ding Z, Xu X. Pathologic Mechanisms of the Newcastle Disease Virus. Viruses 2023; 15:v15040864. [PMID: 37112843 PMCID: PMC10143668 DOI: 10.3390/v15040864] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/18/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023] Open
Abstract
Newcastle disease (ND) has been a consistent risk factor to the poultry industry worldwide. Its pathogen, Newcastle disease virus (NDV), is also a promising antitumor treatment candidate. The pathogenic mechanism has intrigued the great curiosity of researchers, and advances in the last two decades have been summarized in this paper. The NDV’s pathogenic ability is highly related to the basic protein structure of the virus, which is described in the Introduction of this review. The overall clinical signs and recent findings pertaining to NDV-related lymph tissue damage are then described. Given the involvement of cytokines in the overall virulence of NDV, cytokines, particularly IL6 and IFN expressed during infection, are reviewed. On the other hand, the host also has its way of antagonizing the virus, which starts with the detection of the pathogen. Thus, advances in NDV’s physiological cell mechanism and the subsequent IFN response, autophagy, and apoptosis are summarized to provide a whole picture of the NDV infection process.
Collapse
|
47
|
Jahun AS, Sorgeloos F, Chaudhry Y, Arthur SE, Hosmillo M, Georgana I, Izuagbe R, Goodfellow IG. Leaked genomic and mitochondrial DNA contribute to the host response to noroviruses in a STING-dependent manner. Cell Rep 2023; 42:112179. [PMID: 36943868 DOI: 10.1016/j.celrep.2023.112179] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 10/11/2022] [Accepted: 02/12/2023] [Indexed: 03/23/2023] Open
Abstract
The cGAS-STING pathway is central to the interferon response against DNA viruses. However, recent studies are increasingly demonstrating its role in the restriction of some RNA viruses. Here, we show that the cGAS-STING pathway also contributes to the interferon response against noroviruses, currently the commonest causes of infectious gastroenteritis worldwide. We show a significant reduction in interferon-β induction and a corresponding increase in viral replication in norovirus-infected cells after deletion of STING, cGAS, or IFI16. Further, we find that immunostimulatory host genome-derived DNA and mitochondrial DNA accumulate in the cytosol of norovirus-infected cells. Lastly, overexpression of the viral NS4 protein is sufficient to drive the accumulation of cytosolic DNA. Together, our data find a role for cGAS, IFI16, and STING in the restriction of noroviruses and show the utility of host genomic DNA as a damage-associated molecular pattern in cells infected with an RNA virus.
Collapse
Affiliation(s)
- Aminu S Jahun
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK.
| | - Frederic Sorgeloos
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK; Université catholique de Louvain, de Duve Institute, MIPA-VIRO 74-49, 74 Avenue Hippocrate, B-1200 Brussels, Belgium
| | - Yasmin Chaudhry
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Sabastine E Arthur
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Myra Hosmillo
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Iliana Georgana
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Rhys Izuagbe
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Ian G Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK.
| |
Collapse
|
48
|
Expression of down-regulated ERV LTR elements associates with immune activation in human small-cell lung cancers. Mob DNA 2023; 14:2. [PMID: 36918931 PMCID: PMC10012523 DOI: 10.1186/s13100-023-00290-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
Small-cell lung cancer (SCLC) is an aggressive cancer characterized by immunosuppressive features leading to poor responses to current immunotherapies. Activation of transposable elements (TE) can trigger an innate immune response, which can synergize with immunotherapeutic protocols in patients. However, TE activity in relation to immune gene response is not fully known in human SCLC. Here, we compared TE expression in 104 human SCLC and 24 normal tissues and established their involvement in innate immune responses. We observed that different intergenic TEs, mainly endogenous retroviral (ERV) families, are deregulated in SCLC. Similarly to other cancers, we detected a subset of LTRs that correlate with innate immune gene signatures and cytosolic RNA sensors, such as RIG-I. These LTRs are downregulated in SCLC tumors vs. normal tissues, and are mainly located at transcriptional repressed regions, marked with H3K4me2 in different cell lines. Analyses of different genomic datasets show that chromatin repression is likely due to de-methylase LSD1 activity. Moreover, high expression levels of ERV LTRs predict a better survival upon chemotherapy of SCLC patients. The findings reveal a specific pattern of TE-mediated activation of innate immune genes in SCLC, which can be exploited to establish more effective immunotherapeutic combinations.
Collapse
|
49
|
Albarnaz JD, Weekes MP. Proteomic analysis of antiviral innate immunity. Curr Opin Virol 2023; 58:101291. [PMID: 36529073 DOI: 10.1016/j.coviro.2022.101291] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/03/2022] [Accepted: 11/17/2022] [Indexed: 12/23/2022]
Abstract
The capacity of host cells to detect and restrict an infecting virus rests on an array of cell-autonomous antiviral effectors and innate immune receptors that can trigger inflammatory processes at tissue and organismal levels. Dynamic changes in protein abundance, subcellular localisation, post-translational modifications and interactions with other biomolecules govern these processes. Proteomics is therefore an ideal experimental tool to discover novel mechanisms of host antiviral immunity. Additional information can be gleaned both about host and virus by systematic analysis of viral immune evasion strategies. In this review, we summarise recent advances in proteomic technologies and their application to antiviral innate immunity.
Collapse
Affiliation(s)
- Jonas D Albarnaz
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, CB2 0XY Cambridge, UK
| | - Michael P Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, CB2 0XY Cambridge, UK.
| |
Collapse
|
50
|
Villamayor L, Rivero V, López-García D, Topham DJ, Martínez-Sobrido L, Nogales A, DeDiego ML. Interferon alpha inducible protein 6 is a negative regulator of innate immune responses by modulating RIG-I activation. Front Immunol 2023; 14:1105309. [PMID: 36793726 PMCID: PMC9923010 DOI: 10.3389/fimmu.2023.1105309] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/05/2023] [Indexed: 01/31/2023] Open
Abstract
Interferons (IFNs), IFN-stimulated genes (ISGs), and inflammatory cytokines mediate innate immune responses, and are essential to establish an antiviral response. Within the innate immune responses, retinoic acid-inducible gene I (RIG-I) is a key sensor of virus infections, mediating the transcriptional induction of IFNs and inflammatory proteins. Nevertheless, since excessive responses could be detrimental to the host, these responses need to be tightly regulated. In this work, we describe, for the first time, how knocking-down or knocking-out the expression of IFN alpha-inducible protein 6 (IFI6) increases IFN, ISG, and pro-inflammatory cytokine expression after the infections with Influenza A Virus (IAV), Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), and Sendai Virus (SeV), or poly(I:C) transfection. We also show how overexpression of IFI6 produces the opposite effect, in vitro and in vivo, indicating that IFI6 negatively modulates the induction of innate immune responses. Knocking-out or knocking-down the expression of IFI6 diminishes the production of infectious IAV and SARS-CoV-2, most likely because of its effect on antiviral responses. Importantly, we report a novel interaction of IFI6 with RIG-I, most likely mediated through binding to RNA, that affects RIG-I activation, providing a molecular mechanism for the effect of IFI6 on negatively regulating innate immunity. Remarkably, these new functions of IFI6 could be targeted to treat diseases associated with an exacerbated induction of innate immune responses and to combat viral infections, such as IAV and SARS-CoV-2.
Collapse
Affiliation(s)
- Laura Villamayor
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Vanessa Rivero
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Darío López-García
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - David J. Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Luis Martínez-Sobrido
- Disease Intervention and Prevention and Population Health Programs, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Aitor Nogales
- Center for Animal Health Research, CISA-INIA-CSIC, Valdeolmos, Madrid, Spain
| | - Marta L. DeDiego
- Department of Molecular and Cell Biology. Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain,*Correspondence: Marta L. DeDiego,
| |
Collapse
|