1
|
Kesheh MM, Bayat M, Kobravi S, Lotfalizadeh MH, Heydari A, Memar MY, Baghi HB, Kermanshahi AZ, Ravaei F, Taghavi SP, Zarepour F, Nahand JS, Hashemian SMR, Mirzaei H. MicroRNAs and human viral diseases: A focus on the role of microRNA-29. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167500. [PMID: 39260679 DOI: 10.1016/j.bbadis.2024.167500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 06/01/2024] [Accepted: 08/01/2024] [Indexed: 09/13/2024]
Abstract
The viral replication can impress through cellular miRNAs. Indeed, either the antiviral responses or the viral infection changes through cellular miRNAs resulting in affecting many regulatory signaling pathways. One of the microRNA families that is effective in human cancers, diseases, and viral infections is the miR-29 family. Members of miR-29 family are effective in different viral infections as their roles have appeared in regulation of immunity pathways either in innate immunity including interferon and inflammatory pathways or in adaptive immunity including activation of T-cells and antibodies production. Although miR-29a affects viral replication by suppressing antiviral responses, it can inhibit the expression of viral mRNAs via binding to their 3'UTR. In the present work, we discuss the evidence related to miR-29a and viral infection through host immunity regulation. We also review roles of other miR-29 family members by focusing on their role as biomarkers for diagnosing and targets for viral diseases management.
Collapse
Affiliation(s)
- Mina Mobini Kesheh
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mobina Bayat
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepehr Kobravi
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Tehran Azad University, Tehran, Iran
| | | | - Azhdar Heydari
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran; Department of Physiology, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atefeh Zamani Kermanshahi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Ravaei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Zarepour
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Seyed Mohammad Reza Hashemian
- Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
2
|
Wang C, Huang C, Li Y, Bai J, Zhao K, Fang Z, Chen J. Hepatitis B surface antigen impairs TLR4 signaling by upregulating A20 expression in monocytes. Microbiol Spectr 2024; 12:e0090924. [PMID: 39248482 PMCID: PMC11448406 DOI: 10.1128/spectrum.00909-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Toll-like receptors (TLRs) play a crucial role in eliminating viral infection. Conversely, viruses have evolved various strategies to disrupt TLR signaling during chronic infection. In the case of hepatitis B virus (HBV), we previously reported that plasma hepatitis B surface antigen (HBsAg) is closely associated with impaired TLR responses in peripheral blood mononuclear cells from chronic hepatitis B (CHB) patients, but the reasons remain unclear. In this study, we investigated the mechanism by which HBsAg suppresses TLR4 signaling in monocyte cell lines. The monocyte cell line THP-1 was pretreated with HBsAg, followed by lipopolysaccharide (LPS) stimulation. Levels of proinflammatory cytokines and the activation of NF-κB, c-JNK, and ERK were examined. We found that HBsAg did not influence the LPS-induced activation of p65, but it disrupted NF-κB promoter activity through the ectopic expression of myeloid differentiation factor 88 (MyD88) and TAK1, suggesting that HBsAg can block downstream TLR4 signaling. Furthermore, we proved that LPS-induced polyubiquitination of tumor necrosis factor receptor-associated factor 6 (TRAF6) and the formation of the TRAF6-TAB2 complex were inhibited in HBsAg-pretreated cells. Interestingly, HBsAg led to a significant upregulation of A20, a ubiquitin-editing enzyme. Correspondingly, downregulation of A20 using siRNA restored LPS-mediated cytokines production, reflecting its crucial role in HBsAg-mediated inhibition of TLR4 signaling. These results demonstrated a novel mechanism by which HBsAg disrupts TLR4 signaling through the upregulation of A20, suggesting that targeting A20 may be a potential strategy to help restore monocyte functions. IMPORTANCE Clearance HBsAg indicates a functional cure of HBV infection, but in chronic hepatitis B (CHB), it is hard to achieve. HBsAg has been found to regulate anti-viral immune responses, such as the activation of TLR. Our previous jobs proved that HBsAg negatively correlates with TLR2/4 activation in monocytes from CHB patients and blocks TLR2 ligand-indcuced IL-12 production in monocytes. However, how TLR4 signaling is affected by HBsAg remains unknown. In this study, we not only observed impaired TLR4 activation after pretreated monocytes with HBsAg but also identified HBsAg-induced A20 play a role in this impairment, which suggests that targeting A20 may be a viable strategy to restore monocyte functions in CHB.
Collapse
Affiliation(s)
- Cong Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Chenlu Huang
- Liver Cancer Institute of Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Yaming Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection(CAMS, Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinjin Bai
- Liver Cancer Institute of Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Kuangjie Zhao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection(CAMS, Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhong Fang
- Liver Cancer Institute of Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Jieliang Chen
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection(CAMS, Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Kazzaz SA, Shaikh KA, White J, Zhou Q, Powell WH, Harhaj EW. Phosphorylation of aryl hydrocarbon receptor interacting protein by TBK1 negatively regulates IRF7 and the type I interferon response. J Biol Chem 2024; 300:105525. [PMID: 38043800 PMCID: PMC10792245 DOI: 10.1016/j.jbc.2023.105525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/09/2023] [Accepted: 11/22/2023] [Indexed: 12/05/2023] Open
Abstract
The innate antiviral response to RNA viruses is initiated by sensing of viral RNAs by RIG-I-like receptors and elicits type I interferon (IFN) production, which stimulates the expression of IFN-stimulated genes that orchestrate the antiviral response to prevent systemic infection. Negative regulation of type I IFN and its master regulator, transcription factor IRF7, is essential to maintain immune homeostasis. We previously demonstrated that AIP (aryl hydrocarbon receptor interacting protein) functions as a negative regulator of the innate antiviral immune response by binding to and sequestering IRF7 in the cytoplasm, thereby preventing IRF7 transcriptional activation and type I IFN production. However, it remains unknown how AIP inhibition of IRF7 is regulated. We show here that the kinase TBK1 phosphorylates AIP and Thr40 serves as the primary target for TBK1 phosphorylation. AIP Thr40 plays critical roles in regulating AIP stability and mediating its interaction with IRF7. The AIP phosphomimetic T40E exhibited increased proteasomal degradation and enhanced interaction with IRF7 compared with wildtype AIP. AIP T40E also blocked IRF7 nuclear translocation, which resulted in reduced type I IFN production and increased viral replication. In sharp contrast, AIP phosphonull mutant T40A had impaired IRF7 binding, and stable expression of AIP T40A in AIP-deficient mouse embryonic fibroblasts elicited a heightened type I IFN response and diminished RNA virus replication. Taken together, these results demonstrate that TBK1-mediated phosphorylation of AIP at Thr40 functions as a molecular switch that enables AIP to interact with and inhibit IRF7, thus preventing overactivation of type I IFN genes by IRF7.
Collapse
Affiliation(s)
- Sarah A Kazzaz
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Kashif A Shaikh
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Jesse White
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Qinjie Zhou
- Department of Microbiology and Immunology, Miller School of Medicine, The University of Miami, Miami, Florida, USA
| | - Wade H Powell
- Biology Department, Kenyon College, Gambier, Ohio, USA
| | - Edward W Harhaj
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
| |
Collapse
|
4
|
Sheveleva O, Protasova E, Nenasheva T, Butorina N, Melnikova V, Gerasimova T, Sakovnich O, Kurinov A, Grigor’eva E, Medvedev S, Lyadova I. A Model of iPSC-Derived Macrophages with TNFAIP3 Overexpression Reveals the Peculiarities of TNFAIP3 Protein Expression and Function in Human Macrophages. Int J Mol Sci 2023; 24:12868. [PMID: 37629049 PMCID: PMC10454046 DOI: 10.3390/ijms241612868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/29/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
Macrophages play a crucial role in the development and control of inflammation. Understanding the mechanisms balancing macrophage inflammatory activity is important to develop new strategies for treating inflammation-related diseases. TNF-α-induced protein 3 (TNFAIP3, A20) is a negative regulator of intracellular inflammatory cascades; its deficiency induces hyper-inflammatory reactions. Whether A20 overexpression can dampen macrophage inflammatory response remains unclear. Here, we generated human-induced pluripotent stem cells with tetracycline-inducible A20 expression and differentiated them into macrophages (A20-iMacs). A20-iMacs displayed morphology, phenotype, and phagocytic activity typical of macrophages, and they displayed upregulated A20 expression in response to doxycycline. A20 overexpression dampened the A20-iMac response to TNF-α, as shown by a decreased expression of IL1B and IL6 mRNA. A dynamic analysis of A20 expression following the generation of A20-iMacs and control iMacs showed that the expression declined in iMacs and that iMacs expressed a lower molecular weight form of the A20 protein (~70 kDa) compared with less differentiated cells (~90 kDa). A low-level expression of A20 and the predominance of a low-molecular-weight A20 form were also characteristic of monocyte-derived macrophages. The study for the first time developed a model for generating macrophages with an inducible expression of a target gene and identified the peculiarities of A20 expression in macrophages that likely underlie macrophage preparedness for inflammatory reactivity. It also suggested the possibility of mitigating inflammatory macrophage responses via A20 overexpression.
Collapse
Affiliation(s)
- Olga Sheveleva
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| | - Elena Protasova
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| | - Tatiana Nenasheva
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| | - Nina Butorina
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| | - Victoria Melnikova
- Laboratory of Comparative Developmental Physiology, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia;
| | - Tatiana Gerasimova
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| | - Olga Sakovnich
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| | - Alexander Kurinov
- Laboratory of Regeneration Problems, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia;
| | - Elena Grigor’eva
- Laboratory of Developmental Epigenetics, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Lavrentyev Ave., 10, 630090 Novosibirsk, Russia; (E.G.); (S.M.)
| | - Sergey Medvedev
- Laboratory of Developmental Epigenetics, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Lavrentyev Ave., 10, 630090 Novosibirsk, Russia; (E.G.); (S.M.)
| | - Irina Lyadova
- Laboratory of Cellular and Molecular Basis of Histogenesis, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Vavilova Str., 26, 119334 Moscow, Russia; (O.S.); (E.P.); (T.N.); (N.B.); (T.G.); (O.S.)
| |
Collapse
|
5
|
Małkowska P, Niedźwiedzka-Rystwej P. Factors affecting RIG-I-Like receptors activation - New research direction for viral hemorrhagic fevers. Front Immunol 2022; 13:1010635. [PMID: 36248895 PMCID: PMC9557057 DOI: 10.3389/fimmu.2022.1010635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Viral hemorrhagic fever (VHF) is a term referring to a group of life-threatening infections caused by several virus families (Arenaviridae, Bunyaviridae, Filoviridae and Flaviviridae). Depending on the virus, the infection can be mild and can be also characterized by an acute course with fever accompanied by hypervolemia and coagulopathy, resulting in bleeding and shock. It has been suggested that the course of the disease is strongly influenced by the activation of signaling pathways leading to RIG-I-like receptor-dependent interferon production. RIG-I-like receptors (RLRs) are one of two major receptor families that detect viral nucleic acid. RLR receptor activation is influenced by a number of factors that may have a key role in the differences that occur during the antiviral immune response in VHF. In the present study, we collected data on RLR receptors in viral hemorrhagic fevers and described factors that may influence the activation of the antiviral response. RLR receptors seem to be a good target for VHF research, which may contribute to better therapeutic and diagnostic strategies. However, due to the difficulty of conducting such studies in humans, we suggest using Lagovirus europaeus as an animal model for VHF.
Collapse
Affiliation(s)
- Paulina Małkowska
- Doctoral School, University of Szczecin, Szczecin, Poland
- Institute of Biology, University of Szczecin, Szczecin, Poland
- *Correspondence: Paulina Małkowska,
| | | |
Collapse
|
6
|
Zhao G, Song D, Wu J, Yang S, Shi S, Cui X, Ren H, Zhang B. Identification of OTUD6B as a new biomarker for prognosis and immunotherapy by pan-cancer analysis. Front Immunol 2022; 13:955091. [PMID: 36052059 PMCID: PMC9425067 DOI: 10.3389/fimmu.2022.955091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/15/2022] [Indexed: 11/15/2022] Open
Abstract
Background Ovarian-tumor (OTU) domain-containing protein 6B (OTUD6B), one of newly identified OTU deubiquitylating enzyme families, is proved to be associated with tumor progression. However, whether it plays a key role in pan-cancer still remains unknown. Methods The profiles of OTUD6B expression in multiple cancers were analyzed using The Cancer Genome Atlas (TCGA) database. Information of protein expression was performed based on the HPA, GeneCards, and String databases. K-M plotter and survival data analysis were used to analyze the prognostic value of OTUD6B expression, including overall survival (OS), disease-specific survival (DSS), disease-free interval (DFI), and progression-free interval (PFI). R package “clusterProfiler” was used for enrichment analysis of OTUD6B. Furthermore, we analyzed the correlation between the expression of OTUD6B, immune infiltration, and immune-related genes. Additionally, we preliminarily validated its tumorigenic effect in lung cancer cell lines. Findings OTUD6B expression was upregulated in most cancers, such as COAD, CHOL, and LUAD, and predicted poor prognosis in most cancers in TCGA. Results showed that OTUD6B expression was positively correlated with memory CD4+ T cells, Th1 CD4+ T cells, and CD8+ T cells. In terms of the immune-related genes, OTUD6B was found to be associated with most types of genes, such as immunostimulatory genes KDR, TGFBR1, and IL-10. Moreover, for most types of tumors, the immune score was found to be negatively correlated with OTUD6B expression. In addition, lung cancer cell lines with OTUD6B knockdown significantly inhibited proliferation and invasion ability of lung cancer cells. Conclusions The study indicated that OTUD6B is an oncogene and may serve as a new potential biomarker in various tumors. OTUD6B may play a part in TIME, which could be applied as a new target for cancer therapy.
Collapse
Affiliation(s)
- Guang Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Dingli Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jie Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Sanhu Yang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi’an, China
| | - Sien Shi
- Department of Thoracic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Xiaohai Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Hong Ren
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Hong Ren, ; Boxiang Zhang,
| | - Boxiang Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Hong Ren, ; Boxiang Zhang,
| |
Collapse
|
7
|
Huang JP, Li J, Xiao YP, Xu LG. BAG6 negatively regulates the RLR signaling pathway by targeting VISA/MAVS. Front Immunol 2022; 13:972184. [PMID: 36045679 PMCID: PMC9420869 DOI: 10.3389/fimmu.2022.972184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 07/25/2022] [Indexed: 12/03/2022] Open
Abstract
The virus-induced signaling adaptor protein VISA (also known as MAVS, ISP-1, Cardif) is a critical adaptor protein in the innate immune response to RNA virus infection. Upon viral infection, VISA self-aggregates to form a sizeable prion-like complex and recruits downstream signal components for signal transduction. Here, we discover that BAG6 (BCL2-associated athanogene 6, formerly BAT3 or Scythe) is an essential negative regulator in the RIG-I-like receptor signaling pathway. BAG6 inhibits the aggregation of VISA by promoting the K48-linked ubiquitination and specifically attenuates the recruitment of TRAF2 by VISA to inhibit RLR signaling. The aggregation of VISA and the interaction of VISA and TRAF2 are enhanced in BAG6-deficient cell lines after viral infection, resulting in the enhanced transcription level of downstream antiviral genes. Our research shows that BAG6 is a critical regulating factor in RIG-I/VISA-mediated innate immune response by targeting VISA.
Collapse
|
8
|
Song J, Li M, Li C, Liu K, Zhu Y, Zhang H. Friend or foe: RIG- I like receptors and diseases. Autoimmun Rev 2022; 21:103161. [PMID: 35926770 PMCID: PMC9343065 DOI: 10.1016/j.autrev.2022.103161] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 07/29/2022] [Indexed: 12/22/2022]
Abstract
Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), which are pivotal sensors of RNA virus invasions, mediate the transcriptional induction of genes encoding type I interferons (IFNs) and proinflammatory cytokines, successfully establishing host antiviral immune response. A few excellent reviews have elaborated on the structural biology of RLRs and the antiviral mechanisms of RLR activation. In this review, we give a basic understanding of RLR biology and summarize recent findings of how RLR signaling cascade is strictly controlled by host regulatory mechanisms, which include RLR-interacting proteins, post-translational modifications and microRNAs (miRNAs). Furthermore, we pay particular attention to the relationship between RLRs and diseases, especially how RLRs participate in SARS-CoV-2, malaria or bacterial infections, how single-nucleotide polymorphisms (SNPs) or mutations in RLRs and antibodies against RLRs lead to autoinflammatory diseases and autoimmune diseases, and how RLRs are involved in anti-tumor immunity. These findings will provide insights and guidance for antiviral and immunomodulatory therapies targeting RLRs.
Collapse
Affiliation(s)
- Jie Song
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China
| | - Muyuan Li
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha City, Hunan Province, China
| | - Caiyan Li
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China
| | - Ke Liu
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China
| | - Yaxi Zhu
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| | - Huali Zhang
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha City, Hunan Province, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha City, Hunan Province, China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha City, Hunan Province, China.
| |
Collapse
|
9
|
Zhang Q, Jia Q, Gao W, Zhang W. The Role of Deubiquitinases in Virus Replication and Host Innate Immune Response. Front Microbiol 2022; 13:839624. [PMID: 35283827 PMCID: PMC8908266 DOI: 10.3389/fmicb.2022.839624] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/12/2022] [Indexed: 11/13/2022] Open
Abstract
As a critical post-translational modification, ubiquitination is known to affect almost all the cellular processes including immunity, signaling pathways, cell death, cancer development, and viral infection by controlling protein stability. Deubiquitinases (DUBs) cleave ubiquitin from proteins and reverse the process of ubiquitination. Thus, DUBs play an important role in the deubiquitination process and serve as therapeutic targets for various diseases. DUBs are found in eukaryotes, bacteria, and viruses and influence various biological processes. Here, we summarize recent findings on the function of DUBs in modulating viral infection, the mechanism by which viral DUBs regulate host innate immune response, and highlight those DUBs that have recently been discovered as antiviral therapeutic targets.
Collapse
Affiliation(s)
- Qinglin Zhang
- College of Life Sciences of Jilin University, Changchun, China
| | - Qizhen Jia
- College of Life Sciences of Jilin University, Changchun, China
| | - Wenying Gao
- Center for Pathogen Biology and Infectious Diseases, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Wenyan Zhang
- Center for Pathogen Biology and Infectious Diseases, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
He FB, Khan H, Huttunen M, Kolehmainen P, Melén K, Maljanen S, Qu M, Jiang M, Kakkola L, Julkunen I. Filovirus VP24 Proteins Differentially Regulate RIG-I and MDA5-Dependent Type I and III Interferon Promoter Activation. Front Immunol 2022; 12:694105. [PMID: 35069519 PMCID: PMC8767557 DOI: 10.3389/fimmu.2021.694105] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022] Open
Abstract
Filovirus family consists of highly pathogenic viruses that have caused fatal outbreaks especially in many African countries. Previously, research focus has been on Ebola, Sudan and Marburg viruses leaving other filoviruses less well studied. Filoviruses, in general, pose a significant global threat since they are highly virulent and potentially transmissible between humans causing sporadic infections and local or widespread epidemics. Filoviruses have the ability to downregulate innate immunity, and especially viral protein 24 (VP24), VP35 and VP40 have variably been shown to interfere with interferon (IFN) gene expression and signaling. Here we systematically analyzed the ability of VP24 proteins of nine filovirus family members to interfere with retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated antigen 5 (MDA5) induced IFN-β and IFN-λ1 promoter activation. All VP24 proteins were localized both in the cell cytoplasm and nucleus in variable amounts. VP24 proteins of Zaire and Sudan ebolaviruses, Lloviu, Taï Forest, Reston, Marburg and Bundibugyo viruses (EBOV, SUDV, LLOV, TAFV, RESTV, MARV and BDBV, respectively) were found to inhibit both RIG-I and MDA5 stimulated IFN-β and IFN-λ1 promoter activation. The inhibition takes place downstream of interferon regulatory factor 3 phosphorylation suggesting the inhibition to occur in the nucleus. VP24 proteins of Mengla (MLAV) or Bombali viruses (BOMV) did not inhibit IFN-β or IFN-λ1 promoter activation. Six ebolavirus VP24s and Lloviu VP24 bound tightly, whereas MARV and MLAV VP24s bound weakly, to importin α5, the subtype that regulates the nuclear import of STAT complexes. MARV and MLAV VP24 binding to importin α5 was very weak. Our data provides new information on the innate immune inhibitory mechanisms of filovirus VP24 proteins, which may contribute to the pathogenesis of filovirus infections.
Collapse
Affiliation(s)
- Felix B. He
- Institute of Biomedicine/Virology, University of Turku, Turku, Finland
| | - Hira Khan
- Institute of Biomedicine/Virology, University of Turku, Turku, Finland
| | - Moona Huttunen
- Institute of Biomedicine/Virology, University of Turku, Turku, Finland
| | - Pekka Kolehmainen
- Institute of Biomedicine/Virology, University of Turku, Turku, Finland
| | - Krister Melén
- Expert Microbiology Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Sari Maljanen
- Institute of Biomedicine/Virology, University of Turku, Turku, Finland
| | - Mengmeng Qu
- Research Center for Clinical & Translational Medicine, Fifth Medical Center for General Hospital of People’s Liberation Army (PLA), Beijing, China
| | - Miao Jiang
- Expert Microbiology Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Laura Kakkola
- Institute of Biomedicine/Virology, University of Turku, Turku, Finland
| | - Ilkka Julkunen
- Institute of Biomedicine/Virology, University of Turku, Turku, Finland
- Turku University Hospital, Clinical Microbiology, Turku, Finland
| |
Collapse
|
11
|
Yalcinkaya M, Liu W, Islam MN, Kotini AG, Gusarova GA, Fidler TP, Papapetrou EP, Bhattacharya J, Wang N, Tall AR. Modulation of the NLRP3 inflammasome by Sars-CoV-2 Envelope protein. Sci Rep 2021; 11:24432. [PMID: 34952919 PMCID: PMC8709866 DOI: 10.1038/s41598-021-04133-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 12/09/2021] [Indexed: 01/04/2023] Open
Abstract
Despite the initial success of some drugs and vaccines targeting COVID-19, understanding the mechanism underlying SARS-CoV-2 disease pathogenesis remains crucial for the development of further approaches to treatment. Some patients with severe Covid-19 experience a cytokine storm and display evidence of inflammasome activation leading to increased levels of IL-1β and IL-18; however, other reports have suggested reduced inflammatory responses to Sars-Cov-2. In this study we have examined the effects of the Sars-Cov-2 envelope (E) protein, a virulence factor in coronaviruses, on inflammasome activation and pulmonary inflammation. In cultured macrophages the E protein suppressed inflammasome priming and NLRP3 inflammasome activation. Similarly, in mice transfected with E protein and treated with poly(I:C) to simulate the effects of viral RNA, the E protein, in an NLRP3-dependent fashion, reduced expression of pro-IL-1β, levels of IL-1β and IL-18 in broncho-alveolar lavage fluid, and macrophage infiltration in the lung. To simulate the effects of more advanced infection, macrophages were treated with both LPS and poly(I:C). In this setting the E protein increased NLRP3 inflammasome activation in both murine and human macrophages. Thus, the Sars-Cov-2 E protein may initially suppress the host NLRP3 inflammasome response to viral RNA while potentially increasing NLRP3 inflammasome responses in the later stages of infection. Targeting the Sars-Cov-2 E protein especially in the early stages of infection may represent a novel approach to Covid-19 therapy.
Collapse
Affiliation(s)
- Mustafa Yalcinkaya
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Wenli Liu
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Mohammad N Islam
- Lung Biology Lab, Division of Pulmonary, Allergy and Critical Care, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Andriana G Kotini
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Galina A Gusarova
- Lung Biology Lab, Division of Pulmonary, Allergy and Critical Care, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Trevor P Fidler
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Eirini P Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jahar Bhattacharya
- Lung Biology Lab, Division of Pulmonary, Allergy and Critical Care, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Nan Wang
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Alan R Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
12
|
Xu X, Li M, Deng Z, Jiang Z, Li D, Wang S, Hu C. cGASa and cGASb from grass carp (Ctenopharyngodon idellus) play opposite roles in mediating type I interferon response. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 125:104233. [PMID: 34403683 DOI: 10.1016/j.dci.2021.104233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/02/2021] [Accepted: 08/12/2021] [Indexed: 06/13/2023]
Abstract
Cyclic GMP-AMP synthase (cGAS) is known as a DNA sensor for the initiation of innate immune responses in human and other mammals. However, the knowledge about fish cGAS is limited. In this study, we identified two paralogs of cGAS genes from grass carp (Ctenopharyngodon idellus), namely, CicGASa and CicGASb. Grass carp cGASa and cGASb share some conservative domains with mammalian cGASs; however, cGASb contains a unique transmembrane domain. Grass carp cGASa and cGASb responded to GCRV and poly (dA:dT) infection, but they played opposite roles in the regulation of type I IFN response, i.e. cGASa served as an activator for ISGs and NF-κB in a dose-dependent manner, while cGASb acted as an inhibitor. We found that cGASa and cGASb interacted with STING. Similarly, cGASa is an activator for IRF7, but cGASb inhibited IRF7 expression. Both cGASa and STING can protect cells from GCRV infection. Grass carp cGASb inhibited cGASa-induced type I IFN response by the competitive interaction with STING, suggesting that cGASb may be a negative regulator of cGASa-STING-IRF7 axis.
Collapse
Affiliation(s)
- Xiaowen Xu
- College of Life Science, Nanchang University; Nanchang, 330031, Jiangxi, China.
| | - Meifeng Li
- College of Life Science, Nanchang University; Nanchang, 330031, Jiangxi, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Zeyin Jiang
- College of Life Science, Nanchang University; Nanchang, 330031, Jiangxi, China
| | - Dongming Li
- Fuzhou Medical College, Nanchang University, Fuzhou, 344000, China
| | - Shanghong Wang
- College of Life Science, Nanchang University; Nanchang, 330031, Jiangxi, China
| | - Chengyu Hu
- College of Life Science, Nanchang University; Nanchang, 330031, Jiangxi, China.
| |
Collapse
|
13
|
Abstract
Circular RNAs (circRNAs) are a newly discovered class of noncoding RNAs (ncRNAs) present in various tissues and cells. However, the functions of most circRNAs have not been verified experimentally. Here, using deltacoronavirus as a model, differentially expressed circRNAs in cells with or without deltacoronavirus infection were analyzed by RNA sequencing to characterize the cellular responses to RNA virus infection. More than 57,000 circRNA candidates were detected, and seven significantly dysregulated circRNAs were quantitated by real-time reverse transcription-PCR. We discovered a previously unidentified circRNA derived from the TNFAIP3 gene, named circTNFAIP3, which is distributed and expressed widely in various tissues. RNA viruses, including deltacoronaviruses, rather than DNA viruses tend to activate the expression of endogenous circTNFAIP3. Overexpression of circTNFAIP3 promoted deltacoronavirus replication by reducing the apoptosis, while silencing of circTNFAIP3 inhibited deltacoronavirus replication by enhancing the apoptosis. In summary, our work provides useful circRNA-related information to facilitate investigation of the underlying mechanism of deltacoronavirus infection and identifies a novel circTNFAIP3 that promotes deltacoronavirus replication via regulating apoptosis.
Collapse
|
14
|
Qian G, Zhu L, Li G, Liu Y, Zhang Z, Pan J, Lv H. An Integrated View of Deubiquitinating Enzymes Involved in Type I Interferon Signaling, Host Defense and Antiviral Activities. Front Immunol 2021; 12:742542. [PMID: 34707613 PMCID: PMC8542838 DOI: 10.3389/fimmu.2021.742542] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022] Open
Abstract
Viral infectious diseases pose a great challenge to human health around the world. Type I interferons (IFN-Is) function as the first line of host defense and thus play critical roles during virus infection by mediating the transcriptional induction of hundreds of genes. Nevertheless, overactive cytokine immune responses also cause autoimmune diseases, and thus, tight regulation of the innate immune response is needed to achieve viral clearance without causing excessive immune responses. Emerging studies have recently uncovered that the ubiquitin system, particularly deubiquitinating enzymes (DUBs), plays a critical role in regulating innate immune responses. In this review, we highlight recent advances on the diverse mechanisms of human DUBs implicated in IFN-I signaling. These DUBs function dynamically to calibrate host defenses against various virus infections by targeting hub proteins in the IFN-I signaling transduction pathway. We also present a future perspective on the roles of DUB-substrate interaction networks in innate antiviral activities, discuss the promises and challenges of DUB-based drug development, and identify the open questions that remain to be clarified. Our review provides a comprehensive description of DUBs, particularly their differential mechanisms that have evolved in the host to regulate IFN-I-signaling-mediated antiviral responses.
Collapse
Affiliation(s)
- Guanghui Qian
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Liyan Zhu
- Department of Experimental Center, Medical College of Soochow University, Suzhou, China
| | - Gen Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Ying Liu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Zimu Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Haitao Lv
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
15
|
Jami R, Mérour E, Lamoureux A, Bernard J, Millet JK, Biacchesi S. Deciphering the Fine-Tuning of the Retinoic Acid-Inducible Gene-I Pathway in Teleost Fish and Beyond. Front Immunol 2021; 12:679242. [PMID: 33995423 PMCID: PMC8113963 DOI: 10.3389/fimmu.2021.679242] [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: 03/11/2021] [Accepted: 04/07/2021] [Indexed: 11/13/2022] Open
Abstract
Interferons are the first lines of defense against viral pathogen invasion during the early stages of infection. Their synthesis is tightly regulated to prevent excessive immune responses and possible deleterious effects on the host organism itself. The RIG-I-like receptor signaling cascade is one of the major pathways leading to the production of interferons. This pathway amplifies danger signals and mounts an appropriate innate response but also needs to be finely regulated to allow a rapid return to immune homeostasis. Recent advances have characterized different cellular factors involved in the control of the RIG-I pathway. This has been most extensively studied in mammalian species; however, some inconsistencies remain to be resolved. The IFN system is remarkably well conserved in vertebrates and teleost fish possess all functional orthologs of mammalian RIG-I-like receptors as well as most downstream signaling molecules. Orthologs of almost all mammalian regulatory components described to date exist in teleost fish, such as the widely used zebrafish, making fish attractive and powerful models to study in detail the regulation and evolution of the RIG-I pathway.
Collapse
Affiliation(s)
- Raphaël Jami
- University Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Emilie Mérour
- University Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Annie Lamoureux
- University Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Julie Bernard
- University Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Jean K Millet
- University Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | | |
Collapse
|
16
|
Zong Z, Zhang Z, Wu L, Zhang L, Zhou F. The Functional Deubiquitinating Enzymes in Control of Innate Antiviral Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002484. [PMID: 33511009 PMCID: PMC7816709 DOI: 10.1002/advs.202002484] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/09/2020] [Indexed: 05/11/2023]
Abstract
Innate antiviral immunity is the first line of host defense against invading viral pathogens. Immunity activation primarily relies on the recognition of pathogen-associated molecular patterns (PAMPs) by pattern recognition receptors (PRRs). Viral proteins or nucleic acids mainly engage three classes of PRRs: Toll-like receptors (TLRs), retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), and DNA sensor cyclic GMP-AMP (cGAMP) synthase (cGAS). These receptors initiate a series of signaling cascades that lead to the production of proinflammatory cytokines and type I interferon (IFN-I) in response to viral infection. This system requires precise regulation to avoid aberrant activation. Emerging evidence has unveiled the crucial roles that the ubiquitin system, especially deubiquitinating enzymes (DUBs), play in controlling immune responses. In this review, an overview of the most current findings on the function of DUBs in the innate antiviral immune pathways is provided. Insights into the role of viral DUBs in counteracting host immune responses are also provided. Furthermore, the prospects and challenges of utilizing DUBs as therapeutic targets for infectious diseases are discussed.
Collapse
Affiliation(s)
- Zhi Zong
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003P. R. China
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
| | - Zhengkui Zhang
- Institute of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Liming Wu
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003P. R. China
| | - Long Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhou310003P. R. China
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
| | - Fangfang Zhou
- Institute of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| |
Collapse
|
17
|
Fischer L, Lucendo-Villarin B, Hay DC, O’Farrelly C. Human PSC-Derived Hepatocytes Express Low Levels of Viral Pathogen Recognition Receptors, but Are Capable of Mounting an Effective Innate Immune Response. Int J Mol Sci 2020; 21:ijms21113831. [PMID: 32481600 PMCID: PMC7312201 DOI: 10.3390/ijms21113831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 04/30/2020] [Accepted: 05/24/2020] [Indexed: 01/19/2023] Open
Abstract
Hepatocytes are key players in the innate immune response to liver pathogens but are challenging to study because of inaccessibility and a short half-life. Recent advances in in vitro differentiation of hepatocyte-like cells (HLCs) facilitated studies of hepatocyte-pathogen interactions. Here, we aimed to define the anti-viral innate immune potential of human HLCs with a focus on toll-like receptor (TLR)-expression and the presence of a metabolic switch. We analysed cytoplasmic pattern recognition receptor (PRR)- and endosomal TLR-expression and activity and adaptation of HLCs to an inflammatory environment. We found that transcript levels of retinoic acid inducible gene I (RIG-I), melanoma differentiation antigen 5 (MDA5), and TLR3 became downregulated during differentiation, indicating the acquisition of a more tolerogenic phenotype, as expected in healthy hepatocytes. HLCs responded to activation of RIG-I by producing interferons (IFNs) and IFN-stimulated genes. Despite low-level expression of TLR3, receptor expression was upregulated in an inflammatory environment. TLR3 signalling induced expression of proinflammatory cytokines at the gene level, indicating that several PRRs need to interact for successful innate immune activation. The inflammatory responsiveness of HLCs was accompanied by the downregulation of cytochrome P450 3A and 1A2 activity and decreased serum protein production, showing that the metabolic switch seen in primary hepatocytes during anti-viral responses is also present in HLCs.
Collapse
Affiliation(s)
- Lena Fischer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland;
| | | | - David C. Hay
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK;
- Correspondence: (D.C.H.); (C.O.)
| | - Cliona O’Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland;
- School of Medicine, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin 2, Ireland
- Correspondence: (D.C.H.); (C.O.)
| |
Collapse
|
18
|
Priem D, van Loo G, Bertrand MJM. A20 and Cell Death-driven Inflammation. Trends Immunol 2020; 41:421-435. [PMID: 32241683 DOI: 10.1016/j.it.2020.03.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/27/2020] [Accepted: 03/04/2020] [Indexed: 12/18/2022]
Abstract
A20 is a potent anti-inflammatory molecule, and mutations in TNFAIP3, the gene encoding A20, are associated with a wide panel of inflammatory pathologies, both in human and mouse. The anti-inflammatory properties of A20 are commonly attributed to its ability to suppress inflammatory NF-κB signaling by functioning as a ubiquitin-editing enzyme. However, A20 also protects cells from death, independently of NF-κB regulation, and recent work has demonstrated that cell death may drive some of the inflammatory conditions caused by A20 deficiency. Adding to the fact that the protective role of A20 does not primarily rely on its catalytic activities, these findings shed new light on A20 biology.
Collapse
Affiliation(s)
- Dario Priem
- Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Geert van Loo
- Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Mathieu J M Bertrand
- Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
19
|
Park YJ, Oanh NTK, Heo J, Kim SG, Lee HS, Lee H, Lee JH, Kang HC, Lim W, Yoo YS, Cho H. Dual targeting of RIG-I and MAVS by MARCH5 mitochondria ubiquitin ligase in innate immunity. Cell Signal 2020; 67:109520. [PMID: 31881323 DOI: 10.1016/j.cellsig.2019.109520] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 12/17/2022]
Abstract
The mitochondrial antiviral signaling (MAVS) protein on the mitochondrial outer membrane acts as a central signaling molecule in the RIG-I-like receptor (RLR) signaling pathway by linking upstream viral RNA recognition to downstream signal activation. We previously reported that mitochondrial E3 ubiquitin ligase, MARCH5, degrades the MAVS protein aggregate and prevents persistent downstream signaling. Since the activated RIG-I oligomer interacts and nucleates the MAVS aggregate, MARCH5 might also target this oligomer. Here, we report that MARCH5 targets and degrades RIG-I, but not its inactive phosphomimetic form (RIG-IS8E). The MARCH5-mediated reduction of RIG-I is restored in the presence of MG132, a proteasome inhibitor. Upon poly(I:C) stimulation, RIG-I forms an oligomer and co-expression of MARCH5 reduces the expression of this oligomer. The RING domain of MARCH5 is necessary for binding to the CARD domain of RIG-I. In an in vivo ubiquitination assay, MARCH5 transfers the Lys 48-linked polyubiquitin to Lys 193 and 203 residues of RIG-I. Thus, dual targeting of active RIG-I and MAVS protein oligomers by MARCH5 is an efficient way to switch-off RLR signaling. We propose that modulation of MARCH5 activity might be beneficial for the treatment of chronic immune diseases.
Collapse
Affiliation(s)
- Yeon-Ji Park
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Nguyen Thi Kim Oanh
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - June Heo
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Seong-Gwang Kim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Ho-Soo Lee
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyojoon Lee
- Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jae-Ho Lee
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea
| | - Ho Chul Kang
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea; Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Wonchung Lim
- Department of Sports Medicine, College of Health Science, Cheongju University, Republic of Korea
| | - Young-Suk Yoo
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea.
| | - Hyeseong Cho
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, Republic of Korea.
| |
Collapse
|
20
|
Martín-Vicente M, González-Sanz R, Cuesta I, Monzón S, Resino S, Martínez I. Downregulation of A20 Expression Increases the Immune Response and Apoptosis and Reduces Virus Production in Cells Infected by the Human Respiratory Syncytial Virus. Vaccines (Basel) 2020; 8:vaccines8010100. [PMID: 32102364 PMCID: PMC7157707 DOI: 10.3390/vaccines8010100] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 01/20/2023] Open
Abstract
Human respiratory syncytial virus (HRSV) causes severe lower respiratory tract infections in infants, the elderly, and immunocompromised adults. Regulation of the immune response against HRSV is crucial to limiting virus replication and immunopathology. The A20/TNFAIP3 protein is a negative regulator of nuclear factor kappa B (NF-κB) and interferon regulatory factors 3/7 (IRF3/7), which are key transcription factors involved in the inflammatory/antiviral response of epithelial cells to virus infection. Here, we investigated the impact of A20 downregulation or knockout on HRSV growth and the induction of the immune response in those cells. Cellular infections in which the expression of A20 was silenced by siRNAs or eliminated by gene knockout showed increased inflammatory/antiviral response and reduced virus production. Similar results were obtained when the expression of A20-interacting proteins, such as TAX1BP1 and ABIN1, was silenced. Additionally, downregulation of A20, TAX1BP1, and ABIN1 increased cell apoptosis in HRSV-infected cells. These results show that the downregulation of A20 expression might contribute in the control of HRSV infections by potentiating the early innate immune response and increasing apoptosis in infected cells.
Collapse
Affiliation(s)
- María Martín-Vicente
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (M.M.-V.); (R.G.-S.); (S.R.)
| | - Rubén González-Sanz
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (M.M.-V.); (R.G.-S.); (S.R.)
| | - Isabel Cuesta
- Unidad de Bioinformática, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (I.C.); (S.M.)
| | - Sara Monzón
- Unidad de Bioinformática, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (I.C.); (S.M.)
| | - Salvador Resino
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (M.M.-V.); (R.G.-S.); (S.R.)
| | - Isidoro Martínez
- Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain; (M.M.-V.); (R.G.-S.); (S.R.)
- Correspondence: ; Tel.: +34-91-8223272; Fax: +34-91-5097919
| |
Collapse
|
21
|
Imai T, Shiraishi A, Nishiyama K, Ishimura M, Ohga S. Lipopolysaccharide-induced monocyte death in a novel ZnF7 domain mutation of TNFAIP3. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 8:2071-2074.e5. [PMID: 32004749 DOI: 10.1016/j.jaip.2020.01.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Takashi Imai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Shiraishi
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kei Nishiyama
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Ishimura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
22
|
Lee HC, Chathuranga K, Lee JS. Intracellular sensing of viral genomes and viral evasion. Exp Mol Med 2019; 51:1-13. [PMID: 31827068 PMCID: PMC6906418 DOI: 10.1038/s12276-019-0299-y] [Citation(s) in RCA: 420] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/15/2019] [Accepted: 05/22/2019] [Indexed: 12/11/2022] Open
Abstract
During viral infection, virus-derived cytosolic nucleic acids are recognized by host intracellular specific sensors. The efficacy of this recognition system is crucial for triggering innate host defenses, which then stimulate more specific adaptive immune responses against the virus. Recent studies show that signal transduction pathways activated by sensing proteins are positively or negatively regulated by many modulators to maintain host immune homeostasis. However, viruses have evolved several strategies to counteract/evade host immune reactions. These systems involve viral proteins that interact with host sensor proteins and prevent them from detecting the viral genome or from initiating immune signaling. In this review, we discuss key regulators of cytosolic sensor proteins and viral proteins based on experimental evidence.
Collapse
Affiliation(s)
- Hyun-Cheol Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea
- Central Research Institute, Komipharm International Co., Ltd, Shiheung, 15094, Korea
| | - Kiramage Chathuranga
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, 34134, Korea.
| |
Collapse
|
23
|
Selenium Treatment Enhanced Clearance of Salmonella in Chicken Macrophages (HD11). Antioxidants (Basel) 2019; 8:antiox8110532. [PMID: 31703342 PMCID: PMC6912687 DOI: 10.3390/antiox8110532] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/28/2019] [Accepted: 11/01/2019] [Indexed: 11/21/2022] Open
Abstract
As an important micronutrient, selenium (Se) plays many essential roles in immune response and protection against pathogens in humans and animals, but underlying mechanisms of Se-based control of salmonella growth within macrophages remain poorly elucidated. In this study, using RNA-seq analyses, we demonstrate that Se treatment (at an appropriate concentration) can modulate the global transcriptome of chicken macrophages HD11. The bioinformatic analyses (KEGG pathway analysis) revealed that the differentially expressed genes (DEGs) were mainly enriched in retinol and glutathione metabolism, revealing that Se may be associated with retinol and glutathione metabolism. Meanwhile, Se treatment increased the number of salmonella invading the HD11 cells, but reduced the number of salmonella within HD11 cells, suggesting that enhanced clearance of salmonella within HD11 cells was potentially modulated by Se treatment. Furthermore, RNA-seq analyses also revealed that nine genes including SIVA1, FAS, and HMOX1 were differentially expressed in HD11 cells infected with salmonella following Se treatment, and GO enrichment analysis showed that these DEGs were mainly enriched in an extrinsic apoptotic signaling pathway. In summary, these results indicate that Se treatment may not only affect retinol and glutathione metabolism in macrophages, but could also inhibit salmonella-induced macrophage apoptosis via an extrinsic apoptotic signaling pathway involving SIVA1.
Collapse
|
24
|
Differential expression of innate immunity regulation genes in chronic HIV-1 infected adults. Cytokine 2019; 126:154871. [PMID: 31629104 DOI: 10.1016/j.cyto.2019.154871] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/23/2019] [Accepted: 09/30/2019] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Chronic activation of the innate immune system plays a central role in HIV-1 disease progression. Negative regulation of innate immunity is critical in preventing the effects of this excessive activation; however, the molecules involved in this process remain to be identified. In this study, we compared the expression of immune regulation genes between HIV-1 infected individuals and healthy control participants to identify genes involved in the regulation of innate immunity in HIV-1 infection. METHODS We conducted gene expression analysis of a series of immune regulatory genes in viremic treatment-naïve HIV-positive donors, patients receiving highly active antiretroviral therapy (HAART) and HIV-negative healthy control participants. Reverse transcription-quantitative PCR (RT-qPCR) was conducted to determine the expression levels of genes in peripheral blood mononuclear cells isolated from all participants. The spearman correlation test and linear regression analysis were performed to evaluate the correlation between gene expression level and viral load. RESULTS The following differentially expressed genes were identified: A20, CYLD, DDX24, MARCH5, MKRN2, PTP1B, RNF125, S1PR1, SOCS1, IFI35, RBCK1, TTLL12 and USP18. The three most differentially expressed genes were A20, S1PR1, and USP18. USP18 correlated positively with viral load. CONCLUSION Thirteen immune regulation genes were identified in comparisons of viremic treatment-naïve HIV-positive donors, HAART-treated patients and healthy control participants, indicating the potential of these genes as therapeutic targets.
Collapse
|
25
|
Li SN, Ling T, Yang YX, Huang JP, Xu LG. CHID1 positively regulates RLR antiviral signaling by targeting the RIG-I/VISA signalosome. J Med Virol 2019; 91:1668-1678. [PMID: 31106867 DOI: 10.1002/jmv.25508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/09/2019] [Accepted: 05/09/2019] [Indexed: 11/09/2022]
Abstract
Retinoic acid-inducible gene-I (RIG-I) belongs to the RIGI-like receptors (RLRs), a class of primary pattern recognition receptors. It senses viral double-strand RNA in the cytoplasm and delivers the activated signal to its adaptor virus-induced signaling adapter (VISA), which then recruits the downstream TNF receptor-associated factors and kinases, triggering a downstream signal cascade that leads to the production of proinflammatory cytokines and antiviral interferons (IFNs). However, the mechanism of RIG-I-mediated antiviral signaling is not fully understood. Here, we demonstrate that chitinase domain-containing 1 (CHID1), a member of the chitinase family, positively regulates the RLR antiviral signaling pathway by targeting the RIG-I/VISA signalosome. CHID1 overexpression enhances the activation of nuclear factor κB (NF-кB) and interferon regulatory factor 3 (IRF3) triggered by Sendai virus (SeV) by promoting the polyubiquitination of RIG-I and VISA, thereby potentiating IFN-β production. CHID1 knockdown in human 239T cells inhibits SeV-induced activation of IRF3 and NF-κB and the induction of IFN-β. These results indicate that CHID1 positively regulates RLR antiviral signal, revealing the novel mechanism of the RIG-I antiviral signaling pathway.
Collapse
Affiliation(s)
- Sheng-Na Li
- Key Laboratory of Functional Small Organic Molecules, Ministry of Education and College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi, China
| | - Ting Ling
- Key Laboratory of Functional Small Organic Molecules, Ministry of Education and College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi, China
| | - Ya-Xian Yang
- Key Laboratory of Functional Small Organic Molecules, Ministry of Education and College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi, China
| | - Jing-Ping Huang
- Key Laboratory of Functional Small Organic Molecules, Ministry of Education and College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi, China
| | - Liang-Guo Xu
- Key Laboratory of Functional Small Organic Molecules, Ministry of Education and College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi, China
| |
Collapse
|
26
|
Yang Q, Bai SY, Li LF, Li S, Zhang Y, Munir M, Qiu HJ. Human Hemoglobin Subunit Beta Functions as a Pleiotropic Regulator of RIG-I/MDA5-Mediated Antiviral Innate Immune Responses. J Virol 2019; 93:e00718-19. [PMID: 31167908 PMCID: PMC6675906 DOI: 10.1128/jvi.00718-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 05/20/2019] [Indexed: 12/28/2022] Open
Abstract
Hemoglobin is an important oxygen-carrying protein and plays crucial roles in establishing host resistance against pathogens and in regulating innate immune responses. The hemoglobin subunit beta (HB) is an essential component of hemoglobin, and we have previously demonstrated that the antiviral role of the porcine HB (pHB) is mediated by promoting type I interferon pathways. Thus, considering the high homology between human HB (hHB) and pHB, we hypothesized that hHB also plays an important role in the antiviral innate immunity. In this study, we characterized hHB as a regulatory factor for the replication of RNA viruses by differentially regulating the RIG-I- and MDA5-mediated antiviral signaling pathways. Furthermore, we showed that hHB directly inhibited MDA5-mediated signaling by reducing the MDA5-double-stranded RNA (dsRNA) interaction. Additionally, hHB required hHB-induced reactive oxygen species (ROS) to promote RIG-I-mediated signaling through enhancement of K63-linked RIG-I ubiquitination. Taken together, our findings suggest that hHB is a pleiotropic regulator of RIG-I/MDA5-mediated antiviral responses and further highlight the importance of the intercellular microenvironment, including the redox state, in regulating antiviral innate immune responses.IMPORTANCE Hemoglobin, the most important oxygen-carrying protein, is involved in the regulation of innate immune responses. We have previously reported that the porcine hemoglobin subunit beta (HB) exerts antiviral activity through regulation of type I interferon production. However, the antiviral activities and the underlying mechanisms of HBs originating from other animals have been poorly understood. Here, we identified human HB (hHB) as a pleiotropic regulator of the replication of RNA viruses through regulation of RIG-I/MDA5-mediated signaling pathways. hHB enhances RIG-I-mediated antiviral responses by promoting RIG-I ubiquitination depending on the hHB-induced reactive oxygen species (ROS), while it blocks MDA5-mediated antiviral signaling by suppressing the MDA5-dsRNA interaction. Our results contribute to an understanding of the crucial roles of hHB in the regulation of the RIG-I/MDA5-mediated signaling pathways. We also provide novel insight into the correlation of the intercellular redox state with the regulation of antiviral innate immunity.
Collapse
Affiliation(s)
- Qian Yang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Si-Yu Bai
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Lian-Feng Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Su Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yuexiu Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Muhammad Munir
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, United Kingdom
| | - Hua-Ji Qiu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
27
|
Woo B, Baek KH. Regulatory interplay between deubiquitinating enzymes and cytokines. Cytokine Growth Factor Rev 2019; 48:40-51. [PMID: 31208841 PMCID: PMC7108389 DOI: 10.1016/j.cytogfr.2019.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 06/07/2019] [Indexed: 02/06/2023]
Abstract
Deubiquitinating enzymes (DUBs) are cysteine protease proteins that reverse the ubiquitination by removing ubiquitins from the target protein. With over 100 DUBs identified and categorized into at least 7 families, many DUBs interact with one or more cytokines, influencing cellular processes, such as antiviral responses, inflammatory responses, apoptosis, etc. While some DUBs influence cytokine pathway or production, some DUBs are cytokine-inducible. In this article, we summarize a list of DUBs, their interaction with cytokines, target proteins and mechanisms of action.
Collapse
Affiliation(s)
- Bean Woo
- Department of Biomedical Science, CHA University, Bundang CHA General Hospital, Gyeonggi-Do, 13488, Republic of Korea; University of Alabama at Birmingham School of Medicine, Birmingham, AL, 35233, USA
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA University, Bundang CHA General Hospital, Gyeonggi-Do, 13488, Republic of Korea.
| |
Collapse
|
28
|
Read SA, Obeid S, Ahlenstiel C, Ahlenstiel G. The Role of Zinc in Antiviral Immunity. Adv Nutr 2019; 10:696-710. [PMID: 31305906 PMCID: PMC6628855 DOI: 10.1093/advances/nmz013] [Citation(s) in RCA: 415] [Impact Index Per Article: 69.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/17/2019] [Accepted: 01/22/2019] [Indexed: 12/16/2022] Open
Abstract
Zinc is an essential trace element that is crucial for growth, development, and the maintenance of immune function. Its influence reaches all organs and cell types, representing an integral component of approximately 10% of the human proteome, and encompassing hundreds of key enzymes and transcription factors. Zinc deficiency is strikingly common, affecting up to a quarter of the population in developing countries, but also affecting distinct populations in the developed world as a result of lifestyle, age, and disease-mediated factors. Consequently, zinc status is a critical factor that can influence antiviral immunity, particularly as zinc-deficient populations are often most at risk of acquiring viral infections such as HIV or hepatitis C virus. This review summarizes current basic science and clinical evidence examining zinc as a direct antiviral, as well as a stimulant of antiviral immunity. An abundance of evidence has accumulated over the past 50 y to demonstrate the antiviral activity of zinc against a variety of viruses, and via numerous mechanisms. The therapeutic use of zinc for viral infections such as herpes simplex virus and the common cold has stemmed from these findings; however, there remains much to be learned regarding the antiviral mechanisms and clinical benefit of zinc supplementation as a preventative and therapeutic treatment for viral infections.
Collapse
Affiliation(s)
- Scott A Read
- Blacktown Medical School, Western Sydney University, Blacktown, New South Wales, Australia
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, New South Wales, Australia
| | - Stephanie Obeid
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Chantelle Ahlenstiel
- The Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Golo Ahlenstiel
- Blacktown Medical School, Western Sydney University, Blacktown, New South Wales, Australia
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Westmead, New South Wales, Australia
| |
Collapse
|
29
|
Abstract
Healthy tissues of the body express relatively low basal levels of interferons. However, following detection of microbial invasion by sentinel receptors, a cascade of events initiates leading to the transcriptional induction of interferon genes. Interferons are secreted and act primarily as paracrine cytokines to bind neighboring cell surface receptors. Binding to interferon receptors activates a signal pathway to the nucleus inducing a set of interferon-stimulated genes. The biological activity of these genes confers the unique antiviral and innate immune response of interferons. The rapid induction of interferons is critical to survival, and equally critical is the recovery from this defensive state. Either an aberrant response to infection or an inherited genetic disorder that leads to sustained or increased interferon levels can tip the balance towards pathogenesis.
Collapse
Affiliation(s)
- Nancy C Reich
- Stony Brook University, Dept Molecular Genetics & Microbiology, 11796 Stony Brook, NY, USA.
| |
Collapse
|
30
|
Mérour E, Jami R, Lamoureux A, Bernard J, Brémont M, Biacchesi S. A20 (tnfaip3) is a negative feedback regulator of RIG-I-Mediated IFN induction in teleost. FISH & SHELLFISH IMMUNOLOGY 2019; 84:857-864. [PMID: 30385247 DOI: 10.1016/j.fsi.2018.10.082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/01/2018] [Accepted: 10/29/2018] [Indexed: 06/08/2023]
Abstract
Interferon production is tightly regulated in order to prevent excessive immune responses. The RIG-I signaling pathway, which is one of the major pathways inducing the production of interferon, is therefore finely regulated through the participation of different molecules such as A20 (TNFAIP3). A20 is a negative key regulatory factor of the immune response. Although A20 has been identified and actively studied in mammals, nothing is known about its putative function in lower vertebrates. In this study, we sought to define the involvement of fish A20 orthologs in the regulation of RIG-I signaling. We showed that A20 completely blocked the activation of IFN and ISG promoters mediated by RIG-I. Furthermore, A20 expression in fish cells was sufficient to reverse the antiviral state induced by the expression of a constitutively active form of RIG-I, thus allowing the efficient replication of a fish rhabdovirus, the viral hemorrhagic septicemia virus (VHSV). We brought evidence that A20 interrupted RIG-I signaling at the level of TBK1 kinase, a critical point of convergence for many different pathways that activates important transcription factors involved in the expression of many cytokines. Finally, we showed that A20 expression was directly induced by the RIG-I pathway demonstrating that fish A20 acts as a negative feedback regulator of this key pathway for the establishment of an antiviral state.
Collapse
Affiliation(s)
- Emilie Mérour
- VIM, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Raphaël Jami
- VIM, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Annie Lamoureux
- VIM, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Julie Bernard
- VIM, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Michel Brémont
- VIM, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | | |
Collapse
|
31
|
Yuan H, You J, You H, Zheng C. Herpes Simplex Virus 1 UL36USP Antagonizes Type I Interferon-Mediated Antiviral Innate Immunity. J Virol 2018; 92:e01161-18. [PMID: 29997210 PMCID: PMC6146802 DOI: 10.1128/jvi.01161-18] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 12/22/2022] Open
Abstract
Type I interferons (IFNs), as major components of the innate immune system, play a vital role in host resistance to a variety of pathogens. Canonical signaling mediated by type I IFNs activates the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway through binding to the IFN-α/β receptor (IFNAR), resulting in transcription of IFN-stimulated genes (ISGs). However, viruses have evolved multiple strategies to evade this process. Here, we report that herpes simplex virus 1 (HSV-1) ubiquitin-specific protease (UL36USP) abrogates the type I IFN-mediated signaling pathway independent of its deubiquitinase (DUB) activity. In this study, ectopically expressed UL36USP inhibited IFN-β-induced activation of ISRE promoter and transcription of ISGs, and overexpression of UL36USP lacking DUB activity did not influence this effect. Furthermore, UL36USP was demonstrated to antagonize IFN-β-induced activation of JAKs and STATs via specifically binding to the IFNAR2 subunit and blocking the interaction between JAK1 and IFNAR2. More importantly, knockdown of HSV-1 UL36USP restored the formation of JAK1-IFNAR2 complex. These findings underline the roles of UL36USP-IFNAR2 interaction in counteracting the type I IFN-mediated signaling pathway and reveal a novel evasion mechanism of antiviral innate immunity by HSV-1.IMPORTANCE Type I IFNs mediate transcription of numerous antiviral genes, creating a remarkable antiviral state in the host. Viruses have evolved various mechanisms to evade this response. Our results indicated that HSV-1 encodes a ubiquitin-specific protease (UL36USP) as an antagonist to subvert type I IFN-mediated signaling. UL36USP was identified to significantly inhibit IFN-β-induced signaling independent of its deubiquitinase (DUB) activity. The underlying mechanism of UL36USP antagonizing type I IFN-mediated signaling was to specifically bind with IFNAR2 and disassociate JAK1 from IFNAR2. For the first time, we identify UL36USP as a crucial suppressor for HSV-1 to evade type I IFN-mediated signaling. Our findings also provide new insights into the innate immune evasion by HSV-1 and will facilitate our understanding of host-virus interplay.
Collapse
Affiliation(s)
- Hui Yuan
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Center of Clinical Laboratory, The Fifth People's Hospital of Wuxi, Affiliated with Jiangnan University, Wuxi, China
| | - Jia You
- The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Hongjuan You
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
32
|
Association of TNFAIP3 and TNIP1 polymorphisms with systemic lupus erythematosus risk: A meta-analysis. Gene 2018; 668:155-165. [PMID: 29783072 DOI: 10.1016/j.gene.2018.05.062] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 05/12/2018] [Accepted: 05/16/2018] [Indexed: 02/01/2023]
Abstract
OBJECT With the development of GWAS, both TNFAIP3 and TNIP1 were revealed to be susceptibility genes of SLE. However, some other studies revealed no association between TNFAIP3, TNIP1 and SLE susceptibility. In order to estimate such association more precisely and systemically, a meta-analysis was conducted. METHOD Studies on the association between TNFAIP3 rs2230926, TNIP1 rs7708392 and SLE risk were carefully selected via searching 3 databases (Pubmed, Embase, and Web of Science). A fixed- or random-effect model was used according to the heterogeneity, and a subgroup analysis by ethnicity was also performed. RESULTS 26 studies from 18 articles involving a total of 21,372 patients and 30,165 controls were analyzed for TNFAIP3 rs2230926. A significant association between the minor G allele of TNFAIP3 rs2230926 and SLE risk was found via a random-effect model (OR = 1.643, 95% CI = (1.462, 1.847), p < 0.01). In the subgroup analysis by ethnicity, significant correlations were also found in all Caucasians, Asians, and Africans (OR = 1.675, 95% CI = (1.353, 2.074), p < 0.01; OR = 1.738, 95% CI = (1.557, 1.940), p < 0.01; OR = 1.324, 95% CI = (1.029, 1.704), p < 0.05). As for TNIP1 rs7708392, 21 studies from 12 articles involving 24,716 cases and 32,200 controls were analyzed. A significant association of the minor C allele of TNIP1 rs7708392 and SLE risk was found via a random-effect model (OR = 1.247, 95% CI = (1.175, 1.323), p < 0.01). In the subgroup analysis by ethnicity, significant correlations were found in Caucasians, and Africans (OR = 1.317, 95% CI = (1.239, 1.401), p < 0.01; OR = 1.210, 95% CI = (1.108, 1.322), p < 0.01). However, there was no significant association in Asians (OR = 1.122, 95% CI = (0.953, 1.321), p > 0.05). CONCLUSION The minor G allele of TNFAIP3 rs2230926 was associated with increased risk of SLE in all Caucasians, Asians, and Africans. The minor C allele of TNIP1 rs7708392 was associated with the increased risk of SLE in Caucasians and Africans, while it was not associated with SLE susceptibility in Asians.
Collapse
|
33
|
Van Quickelberghe E, Martens A, Goeminne LJE, Clement L, van Loo G, Gevaert K. Identification of Immune-Responsive Gene 1 (IRG1) as a Target of A20. J Proteome Res 2018; 17:2182-2191. [DOI: 10.1021/acs.jproteome.8b00139] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | - Arne Martens
- VIB-UGent Center
for Inflammation Research, B-9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | | | | | - Geert van Loo
- VIB-UGent Center
for Inflammation Research, B-9052 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Kris Gevaert
- VIB-UGent Center
for Medical Biotechnology, B-9000 Ghent, Belgium
| |
Collapse
|
34
|
Czerkies M, Korwek Z, Prus W, Kochańczyk M, Jaruszewicz-Błońska J, Tudelska K, Błoński S, Kimmel M, Brasier AR, Lipniacki T. Cell fate in antiviral response arises in the crosstalk of IRF, NF-κB and JAK/STAT pathways. Nat Commun 2018; 9:493. [PMID: 29402958 PMCID: PMC5799375 DOI: 10.1038/s41467-017-02640-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 12/14/2017] [Indexed: 12/24/2022] Open
Abstract
The innate immune system processes pathogen-induced signals into cell fate decisions. How information is turned to decision remains unknown. By combining stochastic mathematical modelling and experimentation, we demonstrate that feedback interactions between the IRF3, NF-κB and STAT pathways lead to switch-like responses to a viral analogue, poly(I:C), in contrast to pulse-like responses to bacterial LPS. Poly(I:C) activates both IRF3 and NF-κB, a requirement for induction of IFNβ expression. Autocrine IFNβ initiates a JAK/STAT-mediated positive-feedback stabilising nuclear IRF3 and NF-κB in first responder cells. Paracrine IFNβ, in turn, sensitises second responder cells through a JAK/STAT-mediated positive feedforward pathway that upregulates the positive-feedback components: RIG-I, PKR and OAS1A. In these sensitised cells, the 'live-or-die' decision phase following poly(I:C) exposure is shorter-they rapidly produce antiviral responses and commit to apoptosis. The interlinked positive feedback and feedforward signalling is key for coordinating cell fate decisions in cellular populations restricting pathogen spread.
Collapse
Affiliation(s)
- Maciej Czerkies
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | - Zbigniew Korwek
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | - Wiktor Prus
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | - Marek Kochańczyk
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | | | - Karolina Tudelska
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | - Sławomir Błoński
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | - Marek Kimmel
- Departments of Statistics and Bioengineering, and Program in Systems, Synthetic, and Physical Biology, Rice University, Houston, TX, 77005, USA
- Systems Engineering Group, Silesian University of Technology, Gliwice, 44-100, Poland
| | - Allan R Brasier
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, 77555-1060, USA
| | - Tomasz Lipniacki
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, 02-106, Poland.
| |
Collapse
|
35
|
Hu YW, Zhang J, Wu XM, Cao L, Nie P, Chang MX. TANK-Binding Kinase 1 (TBK1) Isoforms Negatively Regulate Type I Interferon Induction by Inhibiting TBK1-IRF3 Interaction and IRF3 Phosphorylation. Front Immunol 2018; 9:84. [PMID: 29441066 PMCID: PMC5797597 DOI: 10.3389/fimmu.2018.00084] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/11/2018] [Indexed: 12/21/2022] Open
Abstract
TANK-binding kinase 1 (TBK1) is an important serine/threonine-protein kinase that mediates phosphorylation and nuclear translocation of IRF3, which contributes to induction of type I interferons (IFNs) in the innate antiviral response. In mammals, TBK1 spliced isoform negatively regulates the virus-triggered IFN-β signaling pathway by disrupting the interaction between retinoic acid-inducible gene I (RIG-I) and mitochondria antiviral-signaling protein (MAVS). However, it is still unclear whether alternative splicing patterns and the function of TBK1 isoform(s) exist in teleost fish. In this study, we identify two alternatively spliced isoforms of TBK1 from zebrafish, termed TBK1_tv1 and TBK1_tv2. Both TBK1_tv1 and TBK1_tv2 contain an incomplete STKc_TBK1 domain. Moreover, the UBL_TBK1_like domain is also missing for TBK1_tv2. TBK1_tv1 and TBK1_tv2 are expressed in zebrafish larvae. Overexpression of TBK1_tv1 and TBK1_tv2 inhibits RIG-I-, MAVS-, TBK1-, and IRF3-mediated activation of IFN promoters in response to spring viremia of carp virus infection. Also, TBK1_tv1 and TBK1_tv2 inhibit expression of IFNs and IFN-stimulated genes induced by MAVS and TBK1. Mechanistically, TBK1_tv1 and TBK1_tv2 competitively associate with TBK1 and IRF3 to disrupt the formation of a functional TBK1-IRF3 complex, impeding the phosphorylation of IRF3 mediated by TBK1. Collectively, these results demonstrate that TBK1 spliced isoforms are dominant negative regulators in the RIG-I/MAVS/TBK1/IRF3 antiviral pathway by targeting the functional TBK1-IRF3 complex formation. Identification and functional characterization of piscine TBK1 spliced isoforms may contribute to understanding the role of TBK1 expression in innate antiviral response.
Collapse
Affiliation(s)
- Yi Wei Hu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Xiao Man Wu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Lu Cao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Pin Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan, China
| | - Ming Xian Chang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan, China
| |
Collapse
|
36
|
Arimoto KI, Miyauchi S, Stoner SA, Fan JB, Zhang DE. Negative regulation of type I IFN signaling. J Leukoc Biol 2018; 103:1099-1116. [PMID: 29357192 DOI: 10.1002/jlb.2mir0817-342r] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/15/2017] [Accepted: 12/19/2017] [Indexed: 12/15/2022] Open
Abstract
Type I IFNs (α, β, and others) are a family of cytokines that are produced in physiological conditions as well as in response to the activation of pattern recognition receptors. They are critically important in controlling the host innate and adaptive immune response to viral and some bacterial infections, cancer, and other inflammatory stimuli. However, dysregulation of type I IFN production or response can contribute to immune pathologies termed "interferonopathies", pointing to the importance of balanced activating signals with tightly regulated mechanisms of tuning this signaling. Here, we summarize the recent advances of how type I IFN production and response are controlled at multiple levels of the type I IFN signaling cascade.
Collapse
Affiliation(s)
- Kei-Ichiro Arimoto
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Sayuri Miyauchi
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Samuel A Stoner
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Jun-Bao Fan
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Dong-Er Zhang
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, USA
- Division of Biological Sciences, University of California San Diego, La Jolla, California, USA
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
37
|
|
38
|
Abstract
Host anti-viral innate-immune signalling pathways are regulated by a variety of post-translation modifications including ubiquitination, which is critical to regulate various signalling pathways for synthesis of anti-viral molecules. A homeostasis of host immune responses, induced due to viral infection and further ubiquitination, is maintained by the action of deubiquitinases (DUB). Infecting viruses utilize the process of deubiquitination for tricking host immune system wherein viral DUBs compete with host DUBs for inhibition of innate-immune anti-viral signalling pathways, which instead of maintaining an immune homeostasis bring about virus-mediated pathogenesis. This suggests that viruses co-evolve with their hosts to acquire similar machinery for tricking immune surveillance and establishing infection.
Collapse
Affiliation(s)
- Puja Kumari
- a Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology , Indian Institute of Science Education and Research (IISER) Bhopal , Bhopal , India
| | - Himanshu Kumar
- a Department of Biological Sciences, Laboratory of Immunology and Infectious Disease Biology , Indian Institute of Science Education and Research (IISER) Bhopal , Bhopal , India
| |
Collapse
|
39
|
Quicke KM, Diamond MS, Suthar MS. Negative regulators of the RIG-I-like receptor signaling pathway. Eur J Immunol 2017; 47:615-628. [PMID: 28295214 DOI: 10.1002/eji.201646484] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/18/2017] [Accepted: 03/01/2017] [Indexed: 12/13/2022]
Abstract
Upon recognition of specific molecular patterns on microbes, host cells trigger an innate immune response, which culminates in the production of type I interferons, proinflammatory cytokines and chemokines, and restricts pathogen replication and spread within the host. At each stage of this response, there are stimulatory and inhibitory signals that regulate the magnitude, quality, and character of the response. Positive regulation promotes an antiviral state to control and eventually clear infection, whereas negative regulation dampens inflammation and prevents immune-mediated tissue damage. An overexuberant innate response can lead to cell and tissue destruction, and the development of spontaneous autoimmunity. The retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), RIG-I and melanoma differentiation-associated gene 5 (MDA5), belong to a family of cytosolic host RNA helicases that recognize distinct nonself RNA signatures and trigger innate immune responses against several RNA viruses by signaling through the essential adaptor protein mitochondrial antiviral signaling (MAVS). The RLR signaling pathway is tightly regulated to maximize antiviral immunity and minimize immune-mediated pathology. This review highlights contemporary findings on negative regulators of the RLR signaling pathway, with specific focus on the proteins and biological processes that directly regulate RIG-I, MDA5 and MAVS signaling function.
Collapse
Affiliation(s)
- Kendra M Quicke
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.,Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA.,Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Mehul S Suthar
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA.,Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA, USA
| |
Collapse
|
40
|
Abstract
Apoptosis is an important component of normal tissue physiology, and the prompt removal of apoptotic cells is equally essential to avoid the undesirable consequences of their accumulation and disintegration. Professional phagocytes are highly specialized for engulfing apoptotic cells. The recent ability to track cells that have undergone apoptosis in situ has revealed a division of labor among the tissue resident phagocytes that sample them. Macrophages are uniquely programmed to process internalized apoptotic cell-derived fatty acids, cholesterol and nucleotides, as a reflection of their dominant role in clearing the bulk of apoptotic cells. Dendritic cells carry apoptotic cells to lymph nodes where they signal the emergence and expansion of highly suppressive regulatory CD4 T cells. A broad suppression of inflammation is executed through distinct phagocyte-specific mechanisms. A clever induction of negative regulatory nodes is notable in dendritic cells serving to simultaneously shut down multiple pathways of inflammation. Several of the genes and pathways modulated in phagocytes in response to apoptotic cells have been linked to chronic inflammatory and autoimmune diseases such as atherosclerosis, inflammatory bowel disease and systemic lupus erythematosus. Our collective understanding of old and new phagocyte functions after apoptotic cell phagocytosis demonstrates the enormity of ways to mediate immune suppression and enforce tissue homeostasis.
Collapse
Affiliation(s)
- J Magarian Blander
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| |
Collapse
|
41
|
Feng J, Cao Z, Wang L, Wan Y, Peng N, Wang Q, Chen X, Zhou Y, Zhu Y. Inducible GBP5 Mediates the Antiviral Response via Interferon-Related Pathways during Influenza A Virus Infection. J Innate Immun 2017; 9:419-435. [PMID: 28376501 DOI: 10.1159/000460294] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/08/2017] [Indexed: 12/18/2022] Open
Abstract
Guanylate binding protein (GBP) 5 belongs to the GBP family, which is involved in important cellular processes, including signal transduction, translation, vesicle trafficking, and exocytosis. Structurally, GBPs display a high degree of homology and share highly conserved GTP-binding or hydrolysis domains. GBP5 was reported to be a critical cellular factor in inflammasome assembly. However, little is known about its role in the host antiviral innate immune response. In this study, we found that GBP5 expression was significantly elevated in influenza patients and influenza A virus-infected A549 human lung epithelial cells. The overexpression of GBP5 inhibited virus replication by enhancing the expression of virus-induced interferon (IFN) and IFN-related effectors. Knockdown of GBP5 had the opposite effect. Moreover, GBP5 enhanced endogenous IFN expression by interacting with the NF-κB-essential modulator complex and stimulating NF-κB signaling. Additionally, the expression of proinflammatory factors, such as IL-6, IL-8, tumor necrosis factor-α, cyclooxygenase-2, and inducible nitric oxide synthase, was also activated by GBP5. Taken together, our results reveal that GBP5 inhibited virus replication through the activation of IFN signaling and proinflammatory factors.
Collapse
Affiliation(s)
- Jian Feng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Rao Y, Wan Q, Yang C, Su J. Grass Carp Laboratory of Genetics and Physiology 2 Serves As a Negative Regulator in Retinoic Acid-Inducible Gene I- and Melanoma Differentiation-Associated Gene 5-Mediated Antiviral Signaling in Resting State and Early Stage of Grass Carp Reovirus Infection. Front Immunol 2017; 8:352. [PMID: 28396670 PMCID: PMC5366347 DOI: 10.3389/fimmu.2017.00352] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/13/2017] [Indexed: 12/22/2022] Open
Abstract
Laboratory of genetics and physiology 2 (LGP2) is a key component of RIG-I-like receptors (RLRs). However, the lack of the caspase recruitment domains (CARDs) results in its controversial functional performance as a negative or positive regulator in antiviral responses. Especially, no sufficient evidence uncovers the functional mechanisms of LGP2 in RLR signaling pathways in teleost. Here, negative regulation mechanism of LGP2 in certain situations in retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5)-mediated antiviral responses was identified in Ctenopharyngodon idella kidney cells. LGP2 overexpression inhibits synthesis and phosphorylation of interferon regulatory factor 3/7 (IRF3/7), and mRNA levels and promoter activities of IFNs and NF-κBs in resting state and early phase of grass carp reovirus (GCRV) infection. Knockdown of LGP2 obtains opposite effects. Luciferase report assay indicates that LGP2 works at the upstream of RIG-I and MDA5. LGP2 binds to RIG-I and MDA5 with diverse domain preference and which is independent of GCRV infection. Furthermore, LGP2 restrains K63-linked ubiquitination of RIG-I and MDA5 in various degrees. These differences result in disparate repressive mechanisms of LGP2 to RIG-I- and MDA5-mediated signal activations of IFN-β promoter stimulator 1 and mediator of IRF3 activation. Interestingly, LGP2 also inhibits K48-linked RIG-I and MDA5 ubiquitination to suppress proteins degradation, which guarantees the basal protein levels for subsequently rapid signal activation. All these results reveal a mechanism that LGP2 functions as a suppressor in RLR signaling pathways to maintain cellular homeostasis in resting state and early phase during GCRV infection.
Collapse
Affiliation(s)
- Youliang Rao
- College of Fisheries, Huazhong Agricultural University , Wuhan , China
| | - Quanyuan Wan
- College of Fisheries, Huazhong Agricultural University , Wuhan , China
| | - Chunrong Yang
- College of Veterinary Medicine, Huazhong Agricultural University , Wuhan , China
| | - Jianguo Su
- College of Fisheries, Huazhong Agricultural University , Wuhan , China
| |
Collapse
|
43
|
Soonthornvacharin S, Rodriguez-Frandsen A, Zhou Y, Galvez F, Huffmaster NJ, Tripathi S, Balasubramaniam VRMT, Inoue A, de Castro E, Moulton H, Stein DA, Sánchez-Aparicio MT, De Jesus PD, Nguyen Q, König R, Krogan NJ, García-Sastre A, Yoh SM, Chanda SK. Systems-based analysis of RIG-I-dependent signalling identifies KHSRP as an inhibitor of RIG-I receptor activation. Nat Microbiol 2017; 2:17022. [PMID: 28248290 PMCID: PMC5338947 DOI: 10.1038/nmicrobiol.2017.22] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 01/23/2017] [Indexed: 01/05/2023]
Abstract
Retinoic acid-inducible gene I (RIG-I) receptor recognizes 5'-triphosphorylated RNA and triggers a signalling cascade that results in the induction of type-I interferon (IFN)-dependent responses. Its precise regulation represents a pivotal balance between antiviral defences and autoimmunity. To elucidate the cellular cofactors that regulate RIG-I signalling, we performed two global RNA interference analyses to identify both positive and negative regulatory nodes operating on the signalling pathway during virus infection. These factors were integrated with experimentally and computationally derived interactome data to build a RIG-I protein interaction network. Our analysis revealed diverse cellular processes, including the unfolded protein response, Wnt signalling and RNA metabolism, as critical cellular components governing innate responses to non-self RNA species. Importantly, we identified K-Homology Splicing Regulatory Protein (KHSRP) as a negative regulator of this pathway. We find that KHSRP associates with the regulatory domain of RIG-I to maintain the receptor in an inactive state and attenuate its sensing of viral RNA (vRNA). Consistent with increased RIG-I antiviral signalling in the absence of KHSRP, viral replication is reduced when KHSRP expression is knocked down both in vitro and in vivo. Taken together, these data indicate that KHSRP functions as a checkpoint regulator of the innate immune response to pathogen challenge.
Collapse
Affiliation(s)
- Stephen Soonthornvacharin
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
- The San Diego Center for Systems Biology (SDCSB), La Jolla, California 92093, USA
- Graduate School of Biomedical Sciences, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Ariel Rodriguez-Frandsen
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
- The San Diego Center for Systems Biology (SDCSB), La Jolla, California 92093, USA
| | - Yingyao Zhou
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, California 92121, USA
| | - Felipe Galvez
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
- The San Diego Center for Systems Biology (SDCSB), La Jolla, California 92093, USA
| | - Nicholas J Huffmaster
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
- The San Diego Center for Systems Biology (SDCSB), La Jolla, California 92093, USA
| | - Shashank Tripathi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
- Global Health and Emerging Pathogen Institute, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
| | - Vinod R M T Balasubramaniam
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
- Global Health and Emerging Pathogen Institute, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
| | - Atsushi Inoue
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Elisa de Castro
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
- Global Health and Emerging Pathogen Institute, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
| | - Hong Moulton
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University Corvallis, 450 SW 30th Street, Oregon 97331, USA
| | - David A Stein
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University Corvallis, 450 SW 30th Street, Oregon 97331, USA
| | - María Teresa Sánchez-Aparicio
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
- Global Health and Emerging Pathogen Institute, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
| | - Paul D De Jesus
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
- The San Diego Center for Systems Biology (SDCSB), La Jolla, California 92093, USA
| | - Quy Nguyen
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
- The San Diego Center for Systems Biology (SDCSB), La Jolla, California 92093, USA
| | - Renate König
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
- Host-Pathogen-Interactions, Paul-Ehrlich-Institute, German Center for Infection Research (DZIF), Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, 1700 4th Street, Byers Hall 308D, Box 2530, San Francisco, California 94158, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
- Global Health and Emerging Pathogen Institute, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, New York 10029, USA
| | - Sunnie M Yoh
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
- The San Diego Center for Systems Biology (SDCSB), La Jolla, California 92093, USA
| | - Sumit K Chanda
- Immunity and Pathogenesis Program, Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
- The San Diego Center for Systems Biology (SDCSB), La Jolla, California 92093, USA
| |
Collapse
|
44
|
Martínez I, Oliveros JC, Cuesta I, de la Barrera J, Ausina V, Casals C, de Lorenzo A, García E, García-Fojeda B, Garmendia J, González-Nicolau M, Lacoma A, Menéndez M, Moranta D, Nieto A, Ortín J, Pérez-González A, Prat C, Ramos-Sevillano E, Regueiro V, Rodriguez-Frandsen A, Solís D, Yuste J, Bengoechea JA, Melero JA. Apoptosis, Toll-like, RIG-I-like and NOD-like Receptors Are Pathways Jointly Induced by Diverse Respiratory Bacterial and Viral Pathogens. Front Microbiol 2017; 8:276. [PMID: 28298903 PMCID: PMC5331050 DOI: 10.3389/fmicb.2017.00276] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/09/2017] [Indexed: 12/18/2022] Open
Abstract
Lower respiratory tract infections are among the top five leading causes of human death. Fighting these infections is therefore a world health priority. Searching for induced alterations in host gene expression shared by several relevant respiratory pathogens represents an alternative to identify new targets for wide-range host-oriented therapeutics. With this aim, alveolar macrophages were independently infected with three unrelated bacterial (Streptococcus pneumoniae, Klebsiella pneumoniae, and Staphylococcus aureus) and two dissimilar viral (respiratory syncytial virus and influenza A virus) respiratory pathogens, all of them highly relevant for human health. Cells were also activated with bacterial lipopolysaccharide (LPS) as a prototypical pathogen-associated molecular pattern. Patterns of differentially expressed cellular genes shared by the indicated pathogens were searched by microarray analysis. Most of the commonly up-regulated host genes were related to the innate immune response and/or apoptosis, with Toll-like, RIG-I-like and NOD-like receptors among the top 10 signaling pathways with over-expressed genes. These results identify new potential broad-spectrum targets to fight the important human infections caused by the bacteria and viruses studied here.
Collapse
Affiliation(s)
- Isidoro Martínez
- Centro Nacional de Microbiología, Instituto de Salud Carlos IIIMadrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | | | - Isabel Cuesta
- Centro Nacional de Microbiología, Instituto de Salud Carlos III Madrid, Spain
| | - Jorge de la Barrera
- Centro Nacional de Microbiología, Instituto de Salud Carlos III Madrid, Spain
| | - Vicente Ausina
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Badalona, Institut d' Investigació Germans Trias i Pujol, Universitat Autònoma de BarcelonaBarcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Cristina Casals
- Departmento de Bioquímica y Biología Molecular I, Universidad ComplutenseMadrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Alba de Lorenzo
- Departmento de Bioquímica y Biología Molecular I, Universidad ComplutenseMadrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Ernesto García
- Centro de Investigaciones Biológicas (CSIC)Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Belén García-Fojeda
- Departmento de Bioquímica y Biología Molecular I, Universidad ComplutenseMadrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Junkal Garmendia
- Instituto de Agrobiotecnología, CSIC-Universidad Pública de Navarra-GobNavarra, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Mar González-Nicolau
- Fundación de Investigación Sanitaria de las Islas Baleares, Instituto de Investigación Sanitaria de PalmaPalma, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Alicia Lacoma
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Badalona, Institut d' Investigació Germans Trias i Pujol, Universitat Autònoma de BarcelonaBarcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Margarita Menéndez
- Instituto de Química Física Rocasolano (CSIC)Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - David Moranta
- Fundación de Investigación Sanitaria de las Islas Baleares, Instituto de Investigación Sanitaria de PalmaPalma, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Amelia Nieto
- Centro Nacional de Biotecnología (CSIC)Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Juan Ortín
- Centro Nacional de Biotecnología (CSIC)Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Alicia Pérez-González
- Centro Nacional de Biotecnología (CSIC)Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Cristina Prat
- Servei de Microbiologia, Hospital Universitari Germans Trias i Pujol, Badalona, Institut d' Investigació Germans Trias i Pujol, Universitat Autònoma de BarcelonaBarcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Elisa Ramos-Sevillano
- Centro de Investigaciones Biológicas (CSIC)Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Verónica Regueiro
- Fundación de Investigación Sanitaria de las Islas Baleares, Instituto de Investigación Sanitaria de PalmaPalma, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Ariel Rodriguez-Frandsen
- Centro Nacional de Biotecnología (CSIC)Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - Dolores Solís
- Instituto de Química Física Rocasolano (CSIC)Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - José Yuste
- Centro de Investigaciones Biológicas (CSIC)Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - José A Bengoechea
- Fundación de Investigación Sanitaria de las Islas Baleares, Instituto de Investigación Sanitaria de PalmaPalma, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| | - José A Melero
- Centro Nacional de Microbiología, Instituto de Salud Carlos IIIMadrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos IIIMadrid, Spain
| |
Collapse
|
45
|
Pulmonary innate inflammatory responses to agricultural occupational contaminants. Cell Tissue Res 2017; 367:627-642. [PMID: 28168324 DOI: 10.1007/s00441-017-2573-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/09/2017] [Indexed: 10/20/2022]
Abstract
Agricultural workers are exposed to many contaminants and suffer from respiratory and other symptoms. Dusts, gases, microbial products and pesticide residues from farms have been linked to effects on the health of agricultural workers. Growing sets of data from in vitro and in vivo models demonstrate the role of the innate immune system, especially Toll-like receptor 4 (TLR4) and TLR9, in lung inflammation induced following exposure to contaminants in agricultural environments. Interestingly, inflammation and lung function changes appear to be discordant indicating the complexity of inflammatory responses to exposures. Whereas the recent development of rodent models and exposure systems have yielded valuable data, we need new systems to examine the combined effects of multiple contaminants in order to increase our understanding of farm-exposure-induced negative health effects.
Collapse
|
46
|
The Linear Ubiquitin Assembly Complex Modulates Latent Membrane Protein 1 Activation of NF-κB and Interferon Regulatory Factor 7. J Virol 2017; 91:JVI.01138-16. [PMID: 27903798 DOI: 10.1128/jvi.01138-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 11/14/2016] [Indexed: 12/11/2022] Open
Abstract
Recently, linear ubiquitin assembly complex (LUBAC)-mediated linear ubiquitination has come into focus due to its emerging role in activation of NF-κB in different biological contexts. However, the role of LUBAC in LMP1 signaling leading to NF-κB and interferon regulatory factor 7 (IRF7) activation has not been investigated. We show here that RNF31, the key component of LUBAC, interacts with LMP1 and IRF7 in Epstein-Barr virus (EBV)-transformed cells and that LUBAC stimulates linear ubiquitination of NEMO and IRF7. Consequently, LUBAC is required for LMP1 signaling to full activation of NF-κB but inhibits LMP1-stimulated IRF7 transcriptional activity. The protein levels of RNF31 and LMP1 are correlated in EBV-transformed cells. Knockdown of RNF31 in EBV-transformed IB4 cells by RNA interference negatively regulates the expression of the genes downstream of LMP1 signaling and results in a decrease of cell proliferation. These lines of evidence indicate that LUBAC-mediated linear ubiquitination plays crucial roles in regulating LMP1 signaling and functions. IMPORTANCE We show here that LUBAC-mediated linear ubiquitination is required for LMP1 activation of NF-κB but inhibits LMP1-mediated IRF7 activation. Our findings provide novel mechanisms underlying EBV-mediated oncogenesis and may have a broad impact on IRF7-mediated immune responses.
Collapse
|
47
|
Influenza a virus NS1 protein induced A20 contributes to viral replication by suppressing interferon-induced antiviral response. Biochem Biophys Res Commun 2016; 482:1107-1113. [PMID: 27914808 DOI: 10.1016/j.bbrc.2016.11.166] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 11/29/2016] [Indexed: 11/23/2022]
Abstract
The innate immune response provides the first line of defense against viruses and other pathogens by responding to specific microbial molecules. A20 is a cytoplasmic ubiquitin-editing protein that negatively regulates the retinoic acid-inducible gene I (RIG-I)-mediated activation of interferon regulatory factors (IRF) 3. Here, we found that influenza A virus (IAV) non-structural protein (NS) 1 dramatically induced the protein level of A20 in A549 cells whose expression levels were positively associated with the viral virulence. A20 overexpression in A549 cells significantly suppressed IAV-induced the activation of IRF3 and interferon (IFN) promoter, resulted in downregulation of IFNβ and IFN-stimulated genes (ISGs) mRNA. Conversely, silencing A20 expression markedly enhanced IRF3-mediated innate antiviral responses. Furthermore, we demonstrated that A20 overexpression in A549 cells obviously promoted IAV replication, and conversely, knockdown of A20 inhibited the viral replication. Overall, the findings described in this study support and extend previous results on interferon-antagonistic strategies of IAV NS1 by showing an induced host target A20, which restricts IAV-induced host innate immune antiviral responses and thereby facilitates viral replication.
Collapse
|
48
|
Lin W, Zhang J, Lin H, Li Z, Sun X, Xin D, Yang M, Sun L, Li L, Wang H, Chen D, Sun Q. Syndecan-4 negatively regulates antiviral signalling by mediating RIG-I deubiquitination via CYLD. Nat Commun 2016; 7:11848. [PMID: 27279133 PMCID: PMC4906230 DOI: 10.1038/ncomms11848] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 05/05/2016] [Indexed: 12/25/2022] Open
Abstract
Retinoic acid-inducible gene I (RIG-I) plays important roles in pathogen recognition and antiviral signalling transduction. Here we show that syndecan-4 (SDC4) is a RIG-I-interacting partner identified in a yeast two-hybrid screen. We find that SDC4 negatively regulates the RIG-I-mediated antiviral signalling in a feedback-loop control manner. The genetic evidence obtained by using knockout mice further emphasizes this biological role of SDC4 in antiviral signalling. Mechanistically, we show that SDC4 interacts with both RIG-I and deubiquitinase CYLD via its carboxyl-terminal intracellular region. SDC4 likely promotes redistribution of RIG-I and CYLD in a perinuclear pattern post viral infection, and thus enhances the RIG-I-CYLD interaction and potentiates the K63-linked deubiquitination of RIG-I. Collectively, our findings uncover a mechanism by which SDC4 antagonizes the activation of RIG-I in a CYLD-mediated deubiquitination-dependent process, thereby balancing antiviral signalling to avoid deleterious effects on host cells.
Collapse
Affiliation(s)
- Wei Lin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Jing Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Haiyan Lin
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Zexing Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Xiaofeng Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Di Xin
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Meng Yang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Liwei Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Lin Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Hongmei Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Dahua Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Qinmiao Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| |
Collapse
|
49
|
Abstract
Ubiquitination has emerged as a crucial mechanism that regulates signal transduction in diverse biological processes, including different aspects of immune functions. Ubiquitination regulates pattern-recognition receptor signaling that mediates both innate immune responses and dendritic cell maturation required for initiation of adaptive immune responses. Ubiquitination also regulates the development, activation, and differentiation of T cells, thereby maintaining efficient adaptive immune responses to pathogens and immunological tolerance to self-tissues. Like phosphorylation, ubiquitination is a reversible reaction tightly controlled by the opposing actions of ubiquitin ligases and deubiquitinases. Deregulated ubiquitination events are associated with immunological disorders, including autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Hongbo Hu
- Department of Rheumatology and Immunology, State Key Laboratory of Biotherapy & Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Unit 902, Houston, TX 77030, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
50
|
The inhibitory effect of A20 on the inflammatory reaction of epidermal keratinocytes. Int J Mol Med 2016; 37:1099-104. [DOI: 10.3892/ijmm.2016.2514] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 02/29/2016] [Indexed: 11/05/2022] Open
|