1
|
Ramnani B, Devale T, Manivannan P, Haridas A, Malathi K. DHX15 and Rig-I Coordinate Apoptosis and Innate Immune Signaling by Antiviral RNase L. Viruses 2024; 16:1913. [PMID: 39772220 PMCID: PMC11680366 DOI: 10.3390/v16121913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
During virus infection, the activation of the antiviral endoribonuclease, ribonuclease L (RNase L), by a unique ligand 2'-5'-oilgoadenylate (2-5A) causes the cleavage of single-stranded viral and cellular RNA targets, restricting protein synthesis, activating stress response pathways, and promoting cell death to establish broad antiviral effects. The immunostimulatory dsRNA cleavage products of RNase L activity (RL RNAs) recruit diverse dsRNA sensors to activate signaling pathways to amplify interferon (IFN) production and activate inflammasome, but the sensors that promote cell death are not known. In this study, we found that DEAH-box polypeptide 15 (DHX15) and retinoic acid-inducible gene I (Rig-I) are essential for apoptosis induced by RL RNAs and require mitochondrial antiviral signaling (MAVS), c-Jun amino terminal kinase (JNK), and p38 mitogen-activated protein kinase (p38 MAPK) for caspase-3-mediated intrinsic apoptosis. In RNase L-activated cells, DHX15 interacts with Rig-I and MAVS, and cells lacking MAVS expression were resistant to apoptosis. RL RNAs induced the transcription of genes for IFN and proinflammatory cytokines by interferon regulatory factor 3 (IRF-3) and nuclear factor kB (NF-kB), while cells lacking both DHX15 and Rig-I showed a reduced induction of cytokines. However, apoptotic cell death is independent of both IRF-3 and NF-kB, suggesting that cytokine and cell death induction by RL RNAs are uncoupled. The RNA binding of both DHX15 and Rig-I is required for apoptosis induction, and the expression of both single proteins in cells lacking both DHX15 and Rig-I is insufficient to promote cell death by RL RNAs. Cell death induced by RL RNAs suppressed Coxsackievirus B3 (CVB3) replication, and inhibiting caspase-3 activity or cells lacking IRF-3 showed that the induction of apoptosis directly resulted in the CVB3 antiviral effect, and the effects were independent of the role of IRF-3.
Collapse
Affiliation(s)
- Barkha Ramnani
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA; (B.R.); (T.D.); (P.M.); (A.H.)
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Trupti Devale
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA; (B.R.); (T.D.); (P.M.); (A.H.)
| | - Praveen Manivannan
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA; (B.R.); (T.D.); (P.M.); (A.H.)
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Aiswarya Haridas
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA; (B.R.); (T.D.); (P.M.); (A.H.)
| | - Krishnamurthy Malathi
- Department of Biological Sciences, University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA; (B.R.); (T.D.); (P.M.); (A.H.)
| |
Collapse
|
2
|
Zhang J, Zhang L, Liu D, Shi H, Zhang X, Chen J, Yang X, Zeng M, Zhang J, Feng T, Zhu X, Jing Z, Ji Z, Shi D, Feng L. Helicase protein DDX11 as a novel antiviral factor promoting RIG-I-MAVS-mediated signaling pathway. mBio 2024; 15:e0202824. [PMID: 39470258 PMCID: PMC11633105 DOI: 10.1128/mbio.02028-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
Type Ι interferon (IFN) production mediated by retinoic acid-inducible gene 1 (RIG-I) and mitochondrial antiviral signaling protein (MAVS) is essential for antiviral innate immune responses. Here, we report the identification of a novel co-sensor for cytosolic nucleic acids: DEAD/H-box helicase 11 (DDX11), a member of the DExD/H (Asp-Glu-x-Asp/His)-box helicase family. Knockdown or knockout of DDX11 attenuated the ability of cells to increase IFN-β, IFN-stimulated gene 56, and C-X-C motif chemokine ligand 10 in response to SeV and poly (I:C) by blocking the activation of TANK-binding kinase 1 and IFN regulatory factor 3. Nucleic acid sensing by DDX11 was independent of the stimulator of IFN genes but was dependent on RIG-I and MAVS. DDX11 regulated RIG-I-MAVS-mediated IFN signaling by specifically interacting with nucleic acid, RIG-I, and MAVS to enhance RIG-I-double-strand RNA and RIG-I-MAVS binding affinity. Overall, our results identified a critical role for DDX11 in the innate immune response and provided molecular insights into the mechanisms by which DDX11 recognized cytosolic nucleic acid and interacted with RIG-Ι and MAVS for potent IFN signaling and antiviral immunity. IMPORTANCE Innate immunity is the first and most rapid host defense against virus infection. Recognition of viral RNA by the retinoic acid-inducible gene 1 (RIG-I)-like receptors (RLRs) initiates innate antiviral immune responses. How the binding of viral RNA to and activation of the RLRs are regulated remains enigmatic. In this study, we identified DEAD/H-box helicase 11 (DDX11) as a positive regulator of the RIG-I-mitochondrial antiviral signaling protein (MAVS)-mediated signaling pathways. Mechanistically, we demonstrated that DDX11 bound to viral RNA, interacted with RIG-I, and promoted their binding to viral RNA. DDX11 also promoted the interaction between RIG-I and MAVS and activation of RIG-I-MAVS signaling. Overall, our results elucidate the role of DDX11 in RIG-I-MAVS-dependent signaling pathways and may shed light on innate immune gene regulation.
Collapse
Affiliation(s)
- Jiyu Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Liaoyuan Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Dakai Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hongyan Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xin Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jianfei Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xiaoman Yang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Miaomiao Zeng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jialin Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Tingshuai Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xiaoyuan Zhu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhaoyang Jing
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhaoyang Ji
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Da Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Li Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
3
|
Alekseeva ON, Hoa LT, Vorobyev PO, Kochetkov DV, Gumennaya YD, Naberezhnaya ER, Chuvashov DO, Ivanov AV, Chumakov PM, Lipatova AV. Receptors and Host Factors for Enterovirus Infection: Implications for Cancer Therapy. Cancers (Basel) 2024; 16:3139. [PMID: 39335111 PMCID: PMC11430599 DOI: 10.3390/cancers16183139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Enteroviruses, with their diverse clinical manifestations ranging from mild or asymptomatic infections to severe diseases such as poliomyelitis and viral myocarditis, present a public health threat. However, they can also be used as oncolytic agents. This review shows the intricate relationship between enteroviruses and host cell factors. Enteroviruses utilize specific receptors and coreceptors for cell entry that are critical for infection and subsequent viral replication. These receptors, many of which are glycoproteins, facilitate virus binding, capsid destabilization, and internalization into cells, and their expression defines virus tropism towards various types of cells. Since enteroviruses can exploit different receptors, they have high oncolytic potential for personalized cancer therapy, as exemplified by the antitumor activity of certain enterovirus strains including the bioselected non-pathogenic Echovirus type 7/Rigvir, approved for melanoma treatment. Dissecting the roles of individual receptors in the entry of enteroviruses can provide valuable insights into their potential in cancer therapy. This review discusses the application of gene-targeting techniques such as CRISPR/Cas9 technology to investigate the impact of the loss of a particular receptor on the attachment of the virus and its subsequent internalization. It also summarizes the data on their expression in various types of cancer. By understanding how enteroviruses interact with specific cellular receptors, researchers can develop more effective regimens of treatment, offering hope for more targeted and efficient therapeutic strategies.
Collapse
Affiliation(s)
- Olga N. Alekseeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Le T. Hoa
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Pavel O. Vorobyev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Dmitriy V. Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Yana D. Gumennaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Elizaveta R. Naberezhnaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Denis O. Chuvashov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Peter M. Chumakov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| | - Anastasia V. Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.A.); (P.O.V.); (D.V.K.); (Y.D.G.); (E.R.N.); (D.O.C.); (P.M.C.)
| |
Collapse
|
4
|
Haycock J, Maehr T, Dastjerdi A, Steinbach F. Immunostimulation of Asian elephant ( Elephas maximus) blood cells by parapoxvirus ovis and CpG motif-containing bacterial plasmid DNA upregulates innate immune gene expression. Front Immunol 2024; 15:1329820. [PMID: 38590526 PMCID: PMC10999609 DOI: 10.3389/fimmu.2024.1329820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
The immune system of Asian elephants (Elephas maximus) is poorly studied, compared to that of livestock, rodents or humans. The innate immune response has become a focus of interest in relation to Elephant endotheliotropic herpesviruses (EEHVs). EEHVs cause a fatal hemorrhagic disease (EEHV-HD) and are a significant threat to captive Asian elephant populations worldwide. Similar to other herpesvirus infections, nearly all animals become infected, but only some develop disease. As progression to EEHV-HD is often acute, a robust innate immune response is crucial to control EEHV infections. This is invariably true of the host in the first instance, but it can also potentially be modulated by intervention strategies. Here, two immunostimulant veterinary medicinal products, authorized for use in domestic species, were tested for their ability to induce innate anti-viral immune responses in Asian elephant blood cells. Sequence data were obtained for a range of previously unidentified Asian elephant immune genes, including C-X-C motif chemokine ligand 10 (CXCL10), interferon stimulated gene 15 (ISG15) and myxovirus GTPase 1 (Mx1), and were employed in the design of species-specific qPCR assays. These assays were subsequently used in analyses to determine fold changes in gene expression over a period of 24 hours. This study demonstrates that both immunostimulant medications are capable of inducing significant innate anti-viral immune responses which suggests that both could be beneficial in controlling EEHV infections in Asian elephants.
Collapse
Affiliation(s)
- Jonathan Haycock
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- Department of Virology, Animal and Plant Health Agency, Addlestone, United Kingdom
| | - Tanja Maehr
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Akbar Dastjerdi
- Department of Virology, Animal and Plant Health Agency, Addlestone, United Kingdom
| | - Falko Steinbach
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
- Department of Virology, Animal and Plant Health Agency, Addlestone, United Kingdom
| |
Collapse
|
5
|
Zhou P, Liu A, Chen D, Wu W, Zhang Q, Chen H, Zhou H, Luo R. Molecular cloning and functional characterization of pigeon IKKε. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 151:105103. [PMID: 38000488 DOI: 10.1016/j.dci.2023.105103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/19/2023] [Accepted: 11/19/2023] [Indexed: 11/26/2023]
Abstract
Inhibitor of nuclear factor kappa-B kinase ε (IKKε), a member of the non-canonical IκB kinase family, plays a critical role in connecting various signaling pathways associated with the initiation of type I interferon (IFN) production. Although the importance of IKKε in innate immunity has been well established in mammals and fish, its characterization and function in pigeons have remained largely unexplored. In this study, we successfully cloned pigeon IKKε (piIKKε) from pigeon embryo fibroblasts (PEFs) for the first time. This gene encodes 722 amino acids and shares high amino acid similarity with its duck and goose counterparts. piIKKε showed a diffuse cytoplasmic distribution and broad expression in all tissues examined. Overexpression of piIKKε in PEFs significantly activated the IFN-β promoter, with both the kinase and CC domains of piIKKε playing key roles in initiating IFN-β expression. Knockdown of piIKKε using small interfering RNA significantly reduced the levels of IFN-β induced by NDV, AIV, poly (I:C), or SeV. Furthermore, the presence of piIKKε resulted in a remarkable reduction in the replication of both avian influenza virus (AIV) H9N2 and Newcastle disease virus (NDV) in PEFs. Our results demonstrate that piIKKε plays a critical role in mediating antiviral innate immunity in pigeons.
Collapse
Affiliation(s)
- Peng Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
| | - Aixin Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
| | - Dong Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
| | - Wanrong Wu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
| | - Qingxiang Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, 510640, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
| | - Hongbo Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, Guangdong, 510640, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China.
| |
Collapse
|
6
|
He S, Gou H, Zhou Y, Wu C, Ren X, Wu X, Guan G, Jin B, Huang J, Jin Z, Zhao T. The SARS-CoV-2 nucleocapsid protein suppresses innate immunity by remodeling stress granules to atypical foci. FASEB J 2023; 37:e23269. [PMID: 37889852 DOI: 10.1096/fj.202201973rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 08/10/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023]
Abstract
Viruses deploy multiple strategies to suppress the host innate immune response to facilitate viral replication and pathogenesis. Typical G3BP1+ stress granules (SGs) are usually formed in host cells after virus infection to restrain viral translation and to stimulate innate immunity. Thus, viruses have evolved various mechanisms to inhibit SGs or to repurpose SG components such as G3BP1. Previous studies showed that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection inhibited host immunity during the early stage of COVID-19. However, the precise mechanism is not yet well understood. Here we showed that the SARS-CoV-2 nucleocapsid (SARS2-N) protein suppressed the double-stranded RNA (dsRNA)-induced innate immune response, concomitant with inhibition of SGs and the induction of atypical SARS2-N+ /G3BP1+ foci (N+ foci). The SARS2-N protein-induced formation of N+ foci was dependent on the ability of its ITFG motif to hijack G3BP1, which contributed to suppress the innate immune response. Importantly, SARS2-N protein facilitated viral replication by inducing the formation of N+ foci. Viral mutations within SARS2-N protein that impair the formation of N+ foci are associated with the inability of the SARS2-N protein to suppress the immune response. Taken together, our study has revealed a novel mechanism by which SARS-CoV-2 suppresses the innate immune response via induction of atypical N+ foci. We think that this is a critical strategy for viral pathogenesis and has potential therapeutic implications.
Collapse
Affiliation(s)
- Su He
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Hongwei Gou
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Yulin Zhou
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Chunxiu Wu
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Xinxin Ren
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Xiajunpeng Wu
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Guanwen Guan
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Boxing Jin
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Jinhua Huang
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhigang Jin
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Tiejun Zhao
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Wang SH, Du J, Yu J, Zhao Y, Wang Y, Hua S, Zhao K. Coxsackievirus A6 2C protein antagonizes IFN-β production through MDA5 and RIG-I depletion. J Virol 2023; 97:e0107523. [PMID: 37847581 PMCID: PMC10688345 DOI: 10.1128/jvi.01075-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/09/2023] [Indexed: 10/19/2023] Open
Abstract
IMPORTANCE Coxsackievirus A6 (CV-A6) is a major emerging pathogen associated with atypical hand, foot, and mouth disease and can cause serious complications such as encephalitis, acute flaccid paralysis, and neurorespiratory syndrome. Therefore, revealing the associated pathogenic mechanisms could benefit the control of CV-A6 infections. In this study, we demonstrate that the nonstructural 2CCV-A6 suppresses IFN-β production, which supports CV-A6 infection. This is achieved by depleting RNA sensors such as melanoma differentiation-associated gene 5 and retinoic acid-inducible gene I (RIG-I) through the lysosomal pathway. Such a function is shared by 2CEV-A71 and 2CCV-B3 but not 2CCV-A16, suggesting the latter might have an alternative way to promote viral replication. This study broadens our understanding of enterovirus 2C protein regulation of the RIG-I-like receptor signaling pathway and reveals a novel mechanism by which CV-A6 and other enteroviruses evade the host innate immune response. These findings on 2C may provide new therapeutic targets for the development of effective inhibitors against CV-A6 and other enterovirus infections.
Collapse
Affiliation(s)
- Shao-Hua Wang
- Center of Infectious Diseases and Pathogen Biology, First Hospital of Jilin University, Changchun, China
- Institute of Virology and AIDS Research, First Hospital of Jilin University, Changchun, China
| | - Juan Du
- Center of Infectious Diseases and Pathogen Biology, First Hospital of Jilin University, Changchun, China
- Institute of Virology and AIDS Research, First Hospital of Jilin University, Changchun, China
| | - Jinghua Yu
- Center of Infectious Diseases and Pathogen Biology, First Hospital of Jilin University, Changchun, China
- Institute of Virology and AIDS Research, First Hospital of Jilin University, Changchun, China
| | - Yifei Zhao
- Center of Infectious Diseases and Pathogen Biology, First Hospital of Jilin University, Changchun, China
- Institute of Virology and AIDS Research, First Hospital of Jilin University, Changchun, China
| | - Yu Wang
- Center of Infectious Diseases and Pathogen Biology, First Hospital of Jilin University, Changchun, China
- Institute of Virology and AIDS Research, First Hospital of Jilin University, Changchun, China
| | - Shucheng Hua
- Department of Respiratory Medicine, First Hospital of Jilin University, Changchun, China
| | - Ke Zhao
- Center of Infectious Diseases and Pathogen Biology, First Hospital of Jilin University, Changchun, China
- Institute of Virology and AIDS Research, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
Weichert L, Düsedau HP, Fritzsch D, Schreier S, Scharf A, Grashoff M, Cebulski K, Michaelsen-Preusse K, Erck C, Lienenklaus S, Dunay IR, Kröger A. Astrocytes evoke a robust IRF7-independent type I interferon response upon neurotropic viral infection. J Neuroinflammation 2023; 20:213. [PMID: 37737190 PMCID: PMC10515022 DOI: 10.1186/s12974-023-02892-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 09/06/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Type I interferons (IFN-I) are fundamental in controlling viral infections but fatal interferonopathy is restricted in the immune-privileged central nervous system (CNS). In contrast to the well-established role of Interferon Regulatory Factor 7 (IRF7) in the regulation of IFN-I response in the periphery, little is known about the specific function in the CNS. METHODS To investigate the role for IRF7 in antiviral response during neurotropic virus infection, mice deficient for IRF3 and IRF7 were infected systemically with Langat virus (LGTV). Viral burden and IFN-I response was analyzed in the periphery and the CNS by focus formation assay, RT-PCR, immunohistochemistry and in vivo imaging. Microglia and infiltration of CNS-infiltration of immune cells were characterized by flow cytometry. RESULTS Here, we demonstrate that during infection with the neurotropic Langat virus (LGTV), an attenuated member of the tick-borne encephalitis virus (TBEV) subgroup, neurons do not rely on IRF7 for cell-intrinsic antiviral resistance and IFN-I induction. An increased viral replication in IRF7-deficient mice suggests an indirect antiviral mechanism. Astrocytes rely on IRF7 to establish a cell-autonomous antiviral response. Notably, the loss of IRF7 particularly in astrocytes resulted in a high IFN-I production. Sustained production of IFN-I in astrocytes is independent of an IRF7-mediated positive feedback loop. CONCLUSION IFN-I induction in the CNS is profoundly regulated in a cell type-specific fashion.
Collapse
Affiliation(s)
- Loreen Weichert
- Molecular Microbiology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke-University Magdeburg, 39120, Magdeburg, Germany
- Innate Immunity and Infection, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Henning Peter Düsedau
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, 39120, Magdeburg, Germany
| | - David Fritzsch
- Molecular Microbiology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke-University Magdeburg, 39120, Magdeburg, Germany
| | - Sarah Schreier
- Molecular Microbiology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke-University Magdeburg, 39120, Magdeburg, Germany
| | - Annika Scharf
- Molecular Microbiology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke-University Magdeburg, 39120, Magdeburg, Germany
- Innate Immunity and Infection, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Martina Grashoff
- Molecular Microbiology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke-University Magdeburg, 39120, Magdeburg, Germany
- Innate Immunity and Infection, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Kristin Cebulski
- Molecular Microbiology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke-University Magdeburg, 39120, Magdeburg, Germany
| | | | - Christian Erck
- Cellular Proteome Research, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Stefan Lienenklaus
- Institute for Laboratory Animal Science, Hanover Medical School, 30625, Hannover, Germany
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Otto-von-Guericke-University Magdeburg, 39120, Magdeburg, Germany
- Health Campus Immunology, Infectiology, and inflammation (GC-I3), Magdeburg, Germany
- Center for Behavioral Braun Science (CBBS), 39106, Magdeburg, Germany
| | - Andrea Kröger
- Molecular Microbiology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke-University Magdeburg, 39120, Magdeburg, Germany.
- Innate Immunity and Infection, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany.
- Health Campus Immunology, Infectiology, and inflammation (GC-I3), Magdeburg, Germany.
- Center for Behavioral Braun Science (CBBS), 39106, Magdeburg, Germany.
| |
Collapse
|
9
|
Li M, Ayyanathan K, Dittmar M, Miller J, Tapescu I, Lee JS, McGrath ME, Xue Y, Vashee S, Schultz DC, Frieman MB, Cherry S. SARS-CoV-2 ORF6 protein does not antagonize interferon signaling in respiratory epithelial Calu-3 cells during infection. mBio 2023; 14:e0119423. [PMID: 37377442 PMCID: PMC10470815 DOI: 10.1128/mbio.01194-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused millions of deaths, posing a substantial threat to global public health. Viruses evolve different strategies to antagonize or evade host immune responses. While ectopic expression of SARS-CoV-2 accessory protein ORF6 blocks interferon (IFN) production and downstream IFN signaling, the role of ORF6 in IFN signaling during bona fide viral infection of respiratory cells is unclear. By comparing wild-type (WT) and ORF6-deleted (ΔORF6) SARS-CoV-2 infection and IFN signaling in respiratory cells, we found that ΔORF6 SARS-CoV-2 replicates more efficiently than WT virus and, thus, stimulates more robust immune signaling. Loss of ORF6 does not alter innate signaling in infected cells: both WT and ΔORF6 virus induce delayed IFN responses only in bystander cells. Moreover, expression of ORF6 in the context of SARS-CoV-2 infection has no effect on Sendai virus-stimulated IFN induction: robust translocation of IRF3 is observed in both SARS-CoV-2 infected and bystander cells. Furthermore, IFN pretreatment potently blocks WT and ΔORF6 virus replication similarly, and both viruses fail to suppress the induction of interferon-stimulated genes (ISGs) upon IFN-β treatment. However, upon treatment with IFN-β, only bystander cells induce STAT1 translocation during infection with WT virus, whereas ΔORF6 virus-infected cells now show translocation. This suggests that under conditions of high IFN activation, ORF6 can attenuate STAT1 activation. These data provide evidence that ORF6 is not sufficient to antagonize IFN production or IFN signaling in SARS-CoV-2-infected respiratory cells but may impact the efficacy of therapeutics that stimulate innate immune pathways. IMPORTANCE Previous studies identified several SARS-CoV-2 proteins, including ORF6, that antagonize host innate immune responses in the context of overexpression of viral proteins in non-respiratory cells. We set out to determine the role of ORF6 in IFN responses during SARS-CoV-2 infection of respiratory cells. Using a deletion strain, we observed no reduction of infection and no difference in evasion of IFN signaling, with responses limited to bystander cells. Moreover, stimulation of Sendai virus-induced IFN production or IFN-β-stimulated ISG expression was comparable between SARS-CoV-2 virus and SARS-CoV-2 lacking ORF6 virus, suggesting that ORF6 is not sufficient to counteract IFN induction or IFN signaling during viral infection.
Collapse
Affiliation(s)
- Minghua Li
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Kasirajan Ayyanathan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark Dittmar
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jesse Miller
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Iulia Tapescu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jae Seung Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marisa E. McGrath
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yong Xue
- J Craig Venter Institute, Rockville, Maryland, USA
| | | | - David C. Schultz
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Matthew B. Frieman
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
Quach HQ, Goergen KM, Grill DE, Haralambieva IH, Ovsyannikova IG, Poland GA, Kennedy RB. Virus-specific and shared gene expression signatures in immune cells after vaccination in response to influenza and vaccinia stimulation. Front Immunol 2023; 14:1168784. [PMID: 37600811 PMCID: PMC10436507 DOI: 10.3389/fimmu.2023.1168784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Background In the vaccine era, individuals receive multiple vaccines in their lifetime. Host gene expression in response to antigenic stimulation is usually virus-specific; however, identifying shared pathways of host response across a wide spectrum of vaccine pathogens can shed light on the molecular mechanisms/components which can be targeted for the development of broad/universal therapeutics and vaccines. Method We isolated PBMCs, monocytes, B cells, and CD8+ T cells from the peripheral blood of healthy donors, who received both seasonal influenza vaccine (within <1 year) and smallpox vaccine (within 1 - 4 years). Each of the purified cell populations was stimulated with either influenza virus or vaccinia virus. Differentially expressed genes (DEGs) relative to unstimulated controls were identified for each in vitro viral infection, as well as for both viral infections (shared DEGs). Pathway enrichment analysis was performed to associate identified DEGs with KEGG/biological pathways. Results We identified 2,906, 3,888, 681, and 446 DEGs in PBMCs, monocytes, B cells, and CD8+ T cells, respectively, in response to influenza stimulation. Meanwhile, 97, 120, 20, and 10 DEGs were identified as gene signatures in PBMCs, monocytes, B cells, and CD8+ T cells, respectively, upon vaccinia stimulation. The majority of DEGs identified in PBMCs were also found in monocytes after either viral stimulation. Of the virus-specific DEGs, 55, 63, and 9 DEGs occurred in common in PBMCs, monocytes, and B cells, respectively, while no DEGs were shared in infected CD8+ T cells after influenza and vaccinia. Gene set enrichment analysis demonstrated that these shared DEGs were over-represented in innate signaling pathways, including cytokine-cytokine receptor interaction, viral protein interaction with cytokine and cytokine receptor, Toll-like receptor signaling, RIG-I-like receptor signaling pathways, cytosolic DNA-sensing pathways, and natural killer cell mediated cytotoxicity. Conclusion Our results provide insights into virus-host interactions in different immune cells, as well as host defense mechanisms against viral stimulation. Our data also highlights the role of monocytes as a major cell population driving gene expression in ex vivo PBMCs in response to viral stimulation. The immune response signaling pathways identified in this study may provide specific targets for the development of novel virus-specific therapeutics and improved vaccines for vaccinia and influenza. Although influenza and vaccinia viruses have been selected in this study as pathogen models, this approach could be applicable to other pathogens.
Collapse
Affiliation(s)
- Huy Quang Quach
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Krista M. Goergen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Diane E. Grill
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Iana H. Haralambieva
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Inna G. Ovsyannikova
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Gregory A. Poland
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Richard B. Kennedy
- Mayo Clinic Vaccine Research Group, Division of General Internal Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
11
|
Antigen-Specific T Cells and SARS-CoV-2 Infection: Current Approaches and Future Possibilities. Int J Mol Sci 2022; 23:ijms232315122. [PMID: 36499448 PMCID: PMC9737069 DOI: 10.3390/ijms232315122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
COVID-19, a significant global health threat, appears to be an immune-related disease. Failure of effective immune responses in initial stages of infection may contribute to development of cytokine storm and systemic inflammation with organ damage, leading to poor clinical outcomes. Disease severity and the emergence of new SARS-CoV-2 variants highlight the need for new preventative and therapeutic strategies to protect the immunocompromised population. Available data indicate that these people may benefit from adoptive transfer of allogeneic SARS-CoV-2-specific T cells isolated from convalescent individuals. This review first provides an insight into the mechanism of cytokine storm development, as it is directly related to the exhaustion of T cell population, essential for viral clearance and long-term antiviral immunity. Next, we describe virus-specific T lymphocytes as a promising and efficient approach for the treatment and prevention of severe COVID-19. Furthermore, other potential cell-based therapies, including natural killer cells, regulatory T cells and mesenchymal stem cells are mentioned. Additionally, we discuss fast and effective ways of producing clinical-grade antigen-specific T cells which can be cryopreserved and serve as an effective "off-the-shelf" approach for rapid treatment of SARS-CoV-2 infection in case of sudden patient deterioration.
Collapse
|
12
|
Xie W, Tian S, Yang J, Cai S, Jin S, Zhou T, Wu Y, Chen Z, Ji Y, Cui J. OTUD7B deubiquitinates SQSTM1/p62 and promotes IRF3 degradation to regulate antiviral immunity. Autophagy 2022; 18:2288-2302. [PMID: 35100065 PMCID: PMC9542415 DOI: 10.1080/15548627.2022.2026098] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 01/01/2022] [Accepted: 01/03/2022] [Indexed: 02/02/2023] Open
Abstract
Deubiquitination plays an important role in the regulation of the crosstalk between macroautophagy/autophagy and innate immune signaling, yet its regulatory mechanisms are not fully understood. Here we identify the deubiquitinase OTUD7B as a negative regulator of antiviral immunity by targeting IRF3 (interferon regulatory factor 3) for selective autophagic degradation. Mechanistically, OTUD7B interacts with IRF3, and activates IRF3-associated cargo receptor SQSTM1/p62 (sequestosome 1) by removing its K63-linked poly-ubiquitin chains at lysine 7 (K7) to enhance SQSTM1 oligomerization. Moreover, viral infection increased the expression of OTUD7B, which forms a negative feedback loop by promoting IRF3 degradation to balance type I interferon (IFN) signaling. Taken together, our study reveals a specific role of OTUD7B in mediating the activation of cargo receptors in a substrate-dependent manner, which could be a potential target against excessive immune responses.Abbreviations: Baf A1: bafilomycin A1; CGAS: cyclic GMP-AMP synthase; DDX58/RIG-I: DExD/H-box helicase 58; DSS: dextran sodium sulfate; DUBs: deubiquitinating enzymes; GFP: green fluorescent protein; IFN: interferon; IKKi: IKBKB/IkappaB kinase inhibitor; IRF3: interferon regulatory factor 3; ISGs: interferon-stimulated genes; MAVS: mitochondrial antiviral signaling protein; MOI: multiplicity of infection; PAMPs: pathogen-associated molecular patterns; SeV: Sendai virus; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; STING1: stimulator of interferon response cGAMP interactor 1; TBK1: TANK binding kinase 1; Ub: ubiquitin; WT: wild-type; VSV: vesicular stomatitis virus.
Collapse
Affiliation(s)
- Weihong Xie
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
- Huizhou Municipal Central Hospital, Huizhou, P.R.China
| | - Shuo Tian
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Jiahui Yang
- Huizhou Municipal Central Hospital, Huizhou, P.R.China
| | - Sihui Cai
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Shouheng Jin
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Tao Zhou
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Yaoxing Wu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Zhiyun Chen
- Huizhou Municipal Central Hospital, Huizhou, P.R.China
| | - Yanqin Ji
- Huizhou Municipal Central Hospital, Huizhou, P.R.China
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| |
Collapse
|
13
|
Antia A, Pinski AN, Ding S. Re-Examining Rotavirus Innate Immune Evasion: Potential Applications of the Reverse Genetics System. mBio 2022; 13:e0130822. [PMID: 35699371 PMCID: PMC9426431 DOI: 10.1128/mbio.01308-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Rotaviruses represent one of the most successful pathogens in the world, with high infectivity and efficient transmission between the young of many animal species, including humans. To overcome host defenses, rotaviruses have evolved a plethora of strategies to effectively evade the innate immune response, establish initial infection in the small intestine, produce progeny, and shed into the environment. Previously, studying the roles and relative contributions of specific rotaviral factors in innate immune evasion had been challenging without a plasmid-only reverse genetics system. Although still in its infancy, current reverse genetics technology will help address important research questions regarding rotavirus innate immune evasion, host range restriction, and viral pathogenesis. In this review, we summarize the current knowledge about the antiviral host innate immune defense mechanisms, countermeasures of rotavirus-encoded factors, and strategies to better understand these interactions using the rotavirus reverse genetics system.
Collapse
Affiliation(s)
- Avan Antia
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Amanda N. Pinski
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
14
|
Type I and Type II Interferon Antagonism Strategies Used by Paramyxoviridae: Previous and New Discoveries, in Comparison. Viruses 2022; 14:v14051107. [PMID: 35632848 PMCID: PMC9145045 DOI: 10.3390/v14051107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/15/2022] [Accepted: 05/18/2022] [Indexed: 02/04/2023] Open
Abstract
Paramyxoviridae is a viral family within the order of Mononegavirales; they are negative single-strand RNA viruses that can cause significant diseases in both humans and animals. In order to replicate, paramyxoviruses–as any other viruses–have to bypass an important protective mechanism developed by the host’s cells: the defensive line driven by interferon. Once the viruses are recognized, the cells start the production of type I and type III interferons, which leads to the activation of hundreds of genes, many of which encode proteins with the specific function to reduce viral replication. Type II interferon is produced by active immune cells through a different signaling pathway, and activates a diverse range of genes with the same objective to block viral replication. As a result of this selective pressure, viruses have evolved different strategies to avoid the defensive function of interferons. The strategies employed by the different viral species to fight the interferon system include a number of sophisticated mechanisms. Here we analyzed the current status of the various strategies used by paramyxoviruses to subvert type I, II, and III interferon responses.
Collapse
|
15
|
Xu L, Yu Z, Uekusa Y, Kawaguchi S, Kikuchi H, Daitoku K, Minagawa M, Motomura S, Furukawa KI, Oshima Y, Seya K, Imaizumi T. Elucidation of the inhibitory effect of (+)-hopeaphenol on polyinosinic–polycytidylic acid-induced innate immunity activation in human cerebral microvascular endothelial cells. J Pharmacol Sci 2022; 149:147-157. [DOI: 10.1016/j.jphs.2022.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/14/2022] [Accepted: 04/28/2022] [Indexed: 11/26/2022] Open
|
16
|
Wang Z, Chen J, Zhang QG, Huang K, Ma D, Du Q, Tong D, Huang Y. Porcine circovirus type 2 infection inhibits the activation of type I interferon signaling via capsid protein and host gC1qR. Vet Microbiol 2022; 266:109354. [DOI: 10.1016/j.vetmic.2022.109354] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/20/2022] [Accepted: 01/20/2022] [Indexed: 12/12/2022]
|
17
|
Pan T, Cao G, Tang E, Zhao Y, Penaloza-MacMaster P, Fang Y, Huang J. A single-cell atlas reveals shared and distinct immune responses and metabolism during SARS-CoV-2 and HIV-1 infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.01.10.475725. [PMID: 35043114 PMCID: PMC8764725 DOI: 10.1101/2022.01.10.475725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
UNLABELLED SARS-CoV-2 and HIV-1 are RNA viruses that have killed millions of people worldwide. Understanding the similarities and differences between these two infections is critical for understanding disease progression and for developing effective vaccines and therapies, particularly for 38 million HIV-1 + individuals who are vulnerable to SARS-CoV-2 co-infection. Here, we utilized single-cell transcriptomics to perform a systematic comparison of 94,442 PBMCs from 7 COVID-19 and 9 HIV-1 + patients in an integrated immune atlas, in which 27 different cell types were identified using an accurate consensus single-cell annotation method. While immune cells in both cohorts show shared inflammation and disrupted mitochondrial function, COVID-19 patients exhibit stronger humoral immunity, broader IFN-I signaling, elevated Rho GTPase and mTOR pathway activities, and downregulated mitophagy. Our results elucidate transcriptional signatures associated with COVID-19 and HIV-1 that may reveal insights into fundamental disease biology and potential therapeutic targets to treat these viral infections. HIGHLIGHTS COVID-19 and HIV-1 + patients show disease-specific inflammatory immune signatures COVID-19 patients show more productive humoral responses than HIV-1 + patients SARS-CoV-2 elicits more enriched IFN-I signaling relative to HIV-IDivergent, impaired metabolic programs distinguish SARS-CoV-2 and HIV-1 infections.
Collapse
|
18
|
Uddin MB, Sajib EH, Hoque SF, Hassan MM, Ahmed SSU. Macrophages in respiratory system. RECENT ADVANCEMENTS IN MICROBIAL DIVERSITY 2022:299-333. [DOI: 10.1016/b978-0-12-822368-0.00014-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
19
|
Gao Y, Huo X, Wang Z, Yuan G, Liu X, Ai T, Su J. Oral Administration of Bacillus subtilis Subunit Vaccine Significantly Enhances the Immune Protection of Grass Carp against GCRV-II Infection. Viruses 2021; 14:v14010030. [PMID: 35062234 PMCID: PMC8779733 DOI: 10.3390/v14010030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 12/16/2022] Open
Abstract
Grass carp reovirus (GCRV) is a severe virus that causes great losses to grass carp culture every year, and GCRV-II is the current popular and fatal strain. VP56, fibrin on the outer surface of GCRV-II, mediates cell attachment. In this study, we firstly divided the VP56 gene into four fragments to screen the optimal antigen by enzyme-linked immunosorbent assay and neutralizing antibody methods. The second fragment VP56-2 demonstrates the optimal efficiency and was employed as an antigen in the following experiments. Bacillus subtilis were used as a carrier, and VP56-2 was expressed on the surface of the spores. Then, we performed the oral immunization for grass carp and the challenge with GCRV-II. The survival rate was remarkably raised, and mRNA expressions of IgM were significantly up-regulated in spleen and head kidney tissues in the B. s-CotC-VP56-2 group. Three crucial immune indexes (complement C3, lysozyme and total superoxide dismutase) in the sera were also significantly enhanced. mRNA expressions of four important genes (TNF-α, IL-1β, IFN1 and MHC-II) were significantly strengthened. Tissue lesions were obviously attenuated by histopathological slide examination in trunk kidney and spleen tissues. Tissue viral burdens were significantly reduced post-viral challenge. These results indicated that the oral recombinant B. subtilis VP56-2 subunit vaccine is effective for controlling GCRV infection and provides a feasible strategy for the control of fish virus diseases.
Collapse
Affiliation(s)
- Yang Gao
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (Y.G.); (X.H.); (Z.W.); (G.Y.); (X.L.)
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Xingchen Huo
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (Y.G.); (X.H.); (Z.W.); (G.Y.); (X.L.)
| | - Zhensheng Wang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (Y.G.); (X.H.); (Z.W.); (G.Y.); (X.L.)
| | - Gailing Yuan
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (Y.G.); (X.H.); (Z.W.); (G.Y.); (X.L.)
| | - Xiaoling Liu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (Y.G.); (X.H.); (Z.W.); (G.Y.); (X.L.)
| | - Taoshan Ai
- Wuhan Chopper Fishery Bio-Tech Co., Ltd., Wuhan Academy of Agricultural Science, Wuhan 430207, China;
| | - Jianguo Su
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (Y.G.); (X.H.); (Z.W.); (G.Y.); (X.L.)
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
- Correspondence: ; Tel./Fax: +86-27-87282227
| |
Collapse
|
20
|
Sajewicz-Krukowska J, Jastrzębski JP, Grzybek M, Domańska-Blicharz K, Tarasiuk K, Marzec-Kotarska B. Transcriptome Sequencing of the Spleen Reveals Antiviral Response Genes in Chickens Infected with CAstV. Viruses 2021; 13:2374. [PMID: 34960643 PMCID: PMC8708055 DOI: 10.3390/v13122374] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022] Open
Abstract
Astrovirus infections pose a significant problem in the poultry industry, leading to multiple adverse effects such as a decreased egg production, breeding disorders, poor weight gain, and even increased mortality. The commonly observed chicken astrovirus (CAstV) was recently reported to be responsible for the "white chicks syndrome" associated with an increased embryo/chick mortality. CAstV-mediated pathogenesis in chickens occurs due to complex interactions between the infectious pathogen and the immune system. Many aspects of CAstV-chicken interactions remain unclear, and there is no information available regarding possible changes in gene expression in the chicken spleen in response to CAstV infection. We aim to investigate changes in gene expression triggered by CAstV infection. Ten 21-day-old SPF White Leghorn chickens were divided into two groups of five birds each. One group was inoculated with CAstV, and the other used as the negative control. At 4 days post infection, spleen samples were collected and immediately frozen at -70 °C for RNA isolation. We analyzed the isolated RNA, using RNA-seq to generate transcriptional profiles of the chickens' spleens and identify differentially expressed genes (DEGs). The RNA-seq findings were verified by quantitative reverse-transcription PCR (qRT-PCR). A total of 31,959 genes was identified in response to CAstV infection. Eventually, 45 DEGs (p-value < 0.05; log2 fold change > 1) were recognized in the spleen after CAstV infection (26 upregulated DEGs and 19 downregulated DEGs). qRT-PCR performed on four genes (IFIT5, OASL, RASD1, and DDX60) confirmed the RNA-seq results. The most differentially expressed genes encode putative IFN-induced CAstV restriction factors. Most DEGs were associated with the RIG-I-like signaling pathway or more generally with an innate antiviral response (upregulated: BLEC3, CMPK2, IFIT5, OASL, DDX60, and IFI6; downregulated: SPIK5, SELENOP, HSPA2, TMEM158, RASD1, and YWHAB). The study provides a global analysis of host transcriptional changes that occur during CAstV infection in vivo and proves that, in the spleen, CAstV infection in chickens predominantly affects the cell cycle and immune signaling.
Collapse
Affiliation(s)
- Joanna Sajewicz-Krukowska
- Department of Poultry Diseases, National Veterinary Research Institute, 24-100 Puławy, Poland; (K.D.-B.); (K.T.)
| | - Jan Paweł Jastrzębski
- Department of Plant Physiology, Genetics and Biotechnology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland;
| | - Maciej Grzybek
- Department of Tropical Parasitology, Institute of Maritime and Tropical Medicine, Medical University of Gdansk, 81-519 Gdynia, Poland;
| | - Katarzyna Domańska-Blicharz
- Department of Poultry Diseases, National Veterinary Research Institute, 24-100 Puławy, Poland; (K.D.-B.); (K.T.)
| | - Karolina Tarasiuk
- Department of Poultry Diseases, National Veterinary Research Institute, 24-100 Puławy, Poland; (K.D.-B.); (K.T.)
| | - Barbara Marzec-Kotarska
- Department of Clinical Pathomorphology, The Medical University of Lublin, 20-090 Lublin, Poland;
| |
Collapse
|
21
|
Liang S, Wu YS, Li DY, Tang JX, Liu HF. Autophagy in Viral Infection and Pathogenesis. Front Cell Dev Biol 2021; 9:766142. [PMID: 34722550 PMCID: PMC8554085 DOI: 10.3389/fcell.2021.766142] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 09/17/2021] [Indexed: 12/14/2022] Open
Abstract
As an evolutionarily conserved cellular process, autophagy plays an essential role in the cellular metabolism of eukaryotes as well as in viral infection and pathogenesis. Under physiological conditions, autophagy is able to meet cellular energy needs and maintain cellular homeostasis through degrading long-lived cellular proteins and recycling damaged organelles. Upon viral infection, host autophagy could degrade invading viruses and initial innate immune response and facilitate viral antigen presentation, all of which contribute to preventing viral infection and pathogenesis. However, viruses have evolved a variety of strategies during a long evolutionary process, by which they can hijack and subvert host autophagy for their own benefits. In this review, we highlight the function of host autophagy in the key regulatory steps during viral infections and pathogenesis and discuss how the viruses hijack the host autophagy for their life cycle and pathogenesis. Further understanding the function of host autophagy in viral infection and pathogenesis contributes to the development of more specific therapeutic strategies to fight various infectious diseases, such as the coronavirus disease 2019 epidemic.
Collapse
Affiliation(s)
- Shan Liang
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yun-Shan Wu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Dong-Yi Li
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Ji-Xin Tang
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Shunde Women and Children's Hospital, Guangdong Medical University (Foshan Shunde Maternal and Child Healthcare Hospital), Foshan, China
| | - Hua-Feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
22
|
Farooq M, Batool M, Kim MS, Choi S. Toll-Like Receptors as a Therapeutic Target in the Era of Immunotherapies. Front Cell Dev Biol 2021; 9:756315. [PMID: 34671606 PMCID: PMC8522911 DOI: 10.3389/fcell.2021.756315] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/13/2021] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptors (TLRs) are the pattern recognition receptors, which are activated by foreign and host molecules in order to initiate the immune response. They play a crucial role in the regulation of innate immunity, and several studies have shown their importance in bacterial, viral, and fungal infections, autoimmune diseases, and cancers. The consensus view from an immunological perspective is that TLR agonists can serve either as a possible therapeutic agent or as a vaccine adjuvant toward cancers or infectious diseases and that TLR inhibitors may be a promising approach to the treatment of autoimmune diseases, some cancers, bacterial, and viral infections. These notions are based on the fact that TLR agonists stimulate the secretion of proinflammatory cytokines and in general, the development of proinflammatory responses. Some of the TLR-based inhibitory agents have shown to be efficacious in preclinical models and have now entered clinical trials. Therefore, TLRs seem to hold the potential to serve as a perfect target in the era of immunotherapies. We offer a perspective on TLR-based therapeutics that sheds light on their usefulness and on combination therapies. We also highlight various therapeutics that are in the discovery phase or in clinical trials.
Collapse
Affiliation(s)
- Mariya Farooq
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Maria Batool
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
- S&K Therapeutics, Suwon, South Korea
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
- S&K Therapeutics, Suwon, South Korea
| |
Collapse
|
23
|
Hameete BC, Fernández-Calleja JM, de Groot MW, Oppewal TR, Tiemessen MM, Hogenkamp A, de Vries RB, Groenink L. The poly(I:C)-induced maternal immune activation model; a systematic review and meta-analysis of cytokine levels in the offspring. Brain Behav Immun Health 2021; 11:100192. [PMID: 34589729 PMCID: PMC8474626 DOI: 10.1016/j.bbih.2020.100192] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/19/2022] Open
Abstract
The maternal polyinosinic:polycytidylic acid (poly(I:C)) animal model is frequently used to study how maternal immune activation may impact neuro development in the offspring. Here, we present the first systematic review and meta-analysis on the effects of maternal poly(I:C) injection on immune mediators in the offspring and provide an openly accessible systematic map of the data including methodological characteristics. Pubmed and EMBASE were searched for relevant publications, yielding 45 unique papers that met inclusion criteria. We extracted data on immune outcomes and methodological characteristics, and assessed the risk of bias. The descriptive summary showed that most studies reported an absence of effect, with an equal number of studies reporting an increase or decrease in the immune mediator being studied. Meta-analysis showed increased IL-6 concentrations in the offspring of poly(I:C) exposed mothers. This effect appeared larger prenatally than post-weaning. Furthermore, poly(I:C) administration during mid-gestation was associated with higher IL-6 concentrations in the offspring. Maternal poly(I:C) induced changes in IL-1β, Il-10 and TNF-α concentrations were small and could not be associated with age of offspring, gestational period or sampling location. Finally, quality of reporting of potential measures to minimize bias was low, which stresses the importance of adherence to publication guidelines. Since neurodevelopmental disorders in humans tend to be associated with lifelong changes in cytokine concentrations, the absence of these effects as identified in this systematic review may suggest that combining the model with other etiological factors in future studies may provide further insight in the mechanisms through which maternal immune activation affects neurodevelopment. Long-term effects of maternal poly(I:C) on immune mediators in the offspring appear limited. Prenatal measurements and mid gestation poly(I:C) injection are associated with increases in IL-6 concentrations. Variety in methodological conduct hampers identification of key elements that affect cytokine concentrations. The quality of reporting of potential measures to minimize bias is poor.
Collapse
Affiliation(s)
- Bart C. Hameete
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - José M.S. Fernández-Calleja
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Martje W.G.D.M. de Groot
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Titia Rixt Oppewal
- University College Utrecht (UCU), Campusplein 1, Utrecht, 3584 ED, the Netherlands
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 4, Groningen, 9747 AG, the Netherlands
| | - Machteld M. Tiemessen
- Research & Innovation, GCoE Immunology, Danone Nutricia Research, Uppsalalaan 12, Utrecht, 3584 CT, the Netherlands
| | - Astrid Hogenkamp
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
| | - Rob B.M. de Vries
- SYstematic Review Center for Laboratory (Animal) Experimentation, Department for Health Evidence, Radboud University Medical Center, Geert Grooteplein zuid 10, Nijmegen, 6525 GA, the Netherlands
| | - Lucianne Groenink
- Department of Pharmacology, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, the Netherlands
- Corresponding author.
| |
Collapse
|
24
|
Dong L, Cao Y, Hou Y, Liu G. N 6 -methyladenosine RNA methylation: A novel regulator of the development and function of immune cells. J Cell Physiol 2021; 237:329-345. [PMID: 34515345 DOI: 10.1002/jcp.30576] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022]
Abstract
N6 -methyladenosine (m6 A) RNA methylation is a reversible posttranscriptional modification in eukaryotes involving three types of functional proteins: "writers", "erasers", and "readers". m6 A regulates the metabolism of messenger RNAs and noncoding RNAs through RNA structure, splicing, stability, export, and translation, thereby participating in various physiological and pathological processes. Here, we summarize the current state of m6 A methylation researches, focusing on how these modifications modulate the fate decisions of innate and adaptive immune cells and regulate immune responses in immune-associated diseases, including viral infections and cancer. These studies showed that m6 A modifications and m6 A modifying proteins play a critical role in pathogen recognition, immune cell activation, immune cell fate decisions, and immune reactions. m6 A is a novel regulator of immune system homeostasis and activation.
Collapse
Affiliation(s)
- Lin Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yejin Cao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yueru Hou
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
25
|
Mahesh KC, Ngunjiri JM, Ghorbani A, Abundo MEC, Wilbanks KQ, Lee K, Lee CW. Assessment of TLR3 and MDA5-Mediated Immune Responses Using Knockout Quail Fibroblast Cells. Avian Dis 2021; 65:419-428. [PMID: 34427417 DOI: 10.1637/0005-2086-65.3.419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/21/2021] [Indexed: 11/05/2022]
Abstract
Toll-like receptor 3 (TLR3) and melanoma differentiation-associated gene 5 (MDA5) are double-stranded RNA (dsRNA)-recognizing receptors that mediate innate immune responses to virus infection. However, the roles played by these receptors in the pathogenesis of avian viruses are poorly understood. In this study, we generated TLR3 and MDA5 single knockout (SKO) and TLR3-MDA5 double knockout (DKO) quail fibroblast cells and examined dsRNA receptor-mediated innate immune responses in vitro. The knockout cells were then stimulated with a synthetic dsRNA ligand polyinosinic:polycytidylic acid [poly(I:C)] or influenza A virus. Endosomal stimulation of TLR3 by adding poly(I:C) in cell culture media or cytoplasmic stimulation of MDA5 by transfecting poly(I:C) resulted in significant increases of TLR3, MDA5, interferon (IFN) β, and interleukin 8 gene expression levels in wild type (WT) cells. Endosomal poly(I:C) treatment induced a higher level expression of most of the genes tested in MDA5 SKO cells compared with WT cells, but not in TLR3 SKO and DKO cells. Cytoplasmic transfection of poly(I:C) led to significant upregulation of all four genes in WT, TLR3 SKO, and MDA5 SKO cells at 8 hr posttransfection and negligible gene expression changes in TLR3-MDA5 DKO cells. Upon infection with a strain of influenza virus with compromised IFN antagonistic capability, WT cells produced the highest amount of biologically active type I IFN followed by TLR3 SKO and MDA5 SKO cells. DKO cells did not produce detectable amounts of type I IFN. However, the IFN did not induce an antiviral state fast enough to block virus replication, even in WT cells under the experimental conditions employed. In summary, our data demonstrate that TLR3 and MDA5 are the key functional dsRNA receptors in quail and imply their coordinated roles in the induction of innate immune responses upon virus infection.
Collapse
Affiliation(s)
- K C Mahesh
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691.,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210
| | - John M Ngunjiri
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691
| | - Amir Ghorbani
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691.,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210
| | - Michael E C Abundo
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691
| | | | - Kichoon Lee
- Department of Animal Sciences, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Columbus, OH 43210
| | - Chang-Won Lee
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, .,Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
26
|
Chen DD, Jiang JY, Lu LF, Zhang C, Zhou XY, Li ZC, Zhou Y, Li S. Zebrafish Uba1 Degrades IRF3 through K48-Linked Ubiquitination to Inhibit IFN Production. THE JOURNAL OF IMMUNOLOGY 2021; 207:512-522. [PMID: 34193603 DOI: 10.4049/jimmunol.2100125] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/19/2021] [Indexed: 01/03/2023]
Abstract
Fish IFN regulatory factor 3 (IRF3) is a crucial transcription factor in the IFN activation signaling pathway, which leads to IFN production and a positive cycle. Unrestricted IFN expression results in hyperimmune responses and therefore, IFN must be tightly regulated. In the current study, we found that zebrafish Ub-activating enzyme (Uba1) negatively regulated IRF3 via the K-48 ubiquitin proteasome degradation of IRF3. First, ifn expression stimulated by spring viraemia of carp virus infection was blunted by the overexpression of Uba1 and enhanced by Uba1 knockdown. Afterward, we found that Uba1 was localized in the cytoplasm, where it interacted with and degraded IRF3. Functional domains analysis revealed that the C-terminal ubiquitin-fold domain was necessary for IRF3 degradation by Uba1 and the N-terminal DNA-binding domain of IRF3 was indispensable for the degradation by Uba1.The degradation of IRF3 was subsequently impaired by treatment with MG132, a ubiquitin proteasome inhibitor. Further mechanism analysis revealed that Uba1 induced the K48-linked Ub-proteasomal degradation of IRF3. Finally, the antiviral capacity of IRF3 was significantly attenuated by Uba1. Taken together, our study reveals that zebrafish Uba1 interacts with and activates the ubiquitinated degradation of IRF3, providing evidence of the IFN immune balance mechanism in fish.
Collapse
Affiliation(s)
- Dan-Dan Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan, China
| | - Jing-Yu Jiang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China; and
| | - Long-Feng Lu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan, China
| | - Can Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China; and
| | - Xiao-Yu Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Zhuo-Cong Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China; and
| | - Yu Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China; and
| | - Shun Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; .,Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan, China
| |
Collapse
|
27
|
Chibssa TR, Kangethe RT, Berguido FJ, Settypalli TBK, Liu Y, Grabherr R, Loitsch A, Sassu EL, Pichler R, Cattoli G, Diallo A, Wijewardana V, Lamien CE. Innate Immune Responses to Wildtype and Attenuated Sheeppox Virus Mediated Through RIG-1 Sensing in PBMC In-Vitro. Front Immunol 2021; 12:666543. [PMID: 34211465 PMCID: PMC8240667 DOI: 10.3389/fimmu.2021.666543] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022] Open
Abstract
Sheeppox (SPP) is a highly contagious disease of small ruminants caused by sheeppox virus (SPPV) and predominantly occurs in Asia and Africa with significant economic losses. SPPV is genetically and immunologically closely related to goatpox virus (GTPV) and lumpy skin disease virus (LSDV), which infect goats and cattle respectively. SPPV live attenuated vaccines (LAVs) are used for vaccination against SPP and goatpox (GTP). Mechanisms related to innate immunity elicited by SPPV are unknown. Although adaptive immunity is responsible for long-term immunity, it is the innate responses that prevent viral invasion and replication before LAVs generate specific long-term protection. We analyzed the relative expression of thirteen selected genes that included pattern recognition receptors (PRRs), Nuclear factor-κβ p65 (NF-κβ), and cytokines to understand better the interaction between SPPV and its host. The transcripts of targeted genes in sheep PBMC incubated with either wild type (WT) or LAV SPPV were analyzed using quantitative PCR. Among PRRs, we observed a significantly higher expression of RIG-1 in PBMC incubated with both WT and LAV, with the former producing the highest expression level. However, there was high inter-individual variability in cytokine transcripts levels among different donors, with the expression of TNFα, IL-15, and IL-10 all significantly higher in both PBMC infected with either WT or LAV compared to control PBMC. Correlation studies revealed a strong significant correlation between RIG-1 and IL-10, between TLR4, TNFα, and NF-κβ, between IL-18 and IL-15, and between NF-κβ and IL-10. There was also a significant negative correlation between RIG-1 and IFNγ, between TLR3 and IL-1 β, and between TLR4 and IL-15 (P< 0.05). This study identified RIG-1 as an important PRR in the signaling pathway of innate immune activation during SPPV infection, possibly through intermediate viral dsRNA. The role of immunomodulatory molecules produced by SPPV capable of inhibiting downstream signaling activation following RIG-1 upregulation is discussed. These findings advance our knowledge of the induction of immune responses by SPPV and will help develop safer and more potent vaccines against SPP and GTP.
Collapse
Affiliation(s)
- Tesfaye Rufael Chibssa
- Animal Production and Health Laboratory, Joint FAO/IAEA Agricultural and Biotechnology Laboratory, Division of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria.,Institute of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria.,National Animal Health Diagnostic and Investigation Center (NAHDIC), Sebeta, Ethiopia
| | - Richard Thiga Kangethe
- Animal Production and Health Laboratory, Joint FAO/IAEA Agricultural and Biotechnology Laboratory, Division of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Francisco J Berguido
- Animal Production and Health Laboratory, Joint FAO/IAEA Agricultural and Biotechnology Laboratory, Division of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Tirumala Bharani K Settypalli
- Animal Production and Health Laboratory, Joint FAO/IAEA Agricultural and Biotechnology Laboratory, Division of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Yang Liu
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Reingard Grabherr
- Institute of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Angelika Loitsch
- Austrian Agency for Health and Food Safety (AGES), Vienna, Austria
| | - Elena Lucia Sassu
- Animal Production and Health Laboratory, Joint FAO/IAEA Agricultural and Biotechnology Laboratory, Division of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria.,Department for Farm Animals and Veterinary Public Health, University Clinic for Swine, University of Veterinary Medicine, Vienna, Austria
| | - Rudolf Pichler
- Animal Production and Health Laboratory, Joint FAO/IAEA Agricultural and Biotechnology Laboratory, Division of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Giovanni Cattoli
- Animal Production and Health Laboratory, Joint FAO/IAEA Agricultural and Biotechnology Laboratory, Division of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Adama Diallo
- Laboratoire National d'Elevage et de Recherches Vétérinaires, Institut Sénégalais de Recherches Agricoles (ISRA), Dakar, Sénégal.,UMR CIRAD INRA, Animal, Santé, Territoires, Risques et Ecosystèmes (ASTRE), Montpellier, France
| | - Viskam Wijewardana
- Animal Production and Health Laboratory, Joint FAO/IAEA Agricultural and Biotechnology Laboratory, Division of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Charles Euloge Lamien
- Animal Production and Health Laboratory, Joint FAO/IAEA Agricultural and Biotechnology Laboratory, Division of Nuclear Techniques in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| |
Collapse
|
28
|
Yang J, Zhou M, Zhong Y, Xu L, Zeng C, Zhao X, Zhang M. Gene duplication and adaptive evolution of Toll-like receptor genes in birds. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:103990. [PMID: 33422554 DOI: 10.1016/j.dci.2020.103990] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/27/2020] [Accepted: 12/27/2020] [Indexed: 06/12/2023]
Abstract
Toll-like receptors (TLRs) play an important role in innate immune through recognizes pathogens. In order to reveal the evolutionary patterns and adaptive evolution of avian TLRs, we examined 66 representative bird species in 26 orders. Phylogenetic results indicated that TLR1A and TLR1B may have differentiated functionally. Evolutionary analysis showed that the TLR genes in birds under strong Purification selection (0.165-0.4265). A total of 126 common positively selected codons were identified in 10 TLR genes of avian, and most sites were located in the extracellular leucine-rich repeat (LRR) functional domains, and both environment and feeding habits were external factors driving the evolution of avian TLR genes. Environmental pressures had a greater effect on TLR1B, TLR2B, TLR3 and TLR4, while feeding habits were active in affecting TLR2A, TLR2B, TLR15 and TLR21. Our data suggested that TLR genes have been subjected to different selective pressures in the diversification of birds and that these changes enabled them to respond differently to pathogens from diverse sources.
Collapse
Affiliation(s)
- Jiandong Yang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Southwest China Wildlife Rsources Conservation (Ministry of Education), China West Normal University, Nanchong, 637009, PR China.
| | - Ming Zhou
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China; Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Yun Zhong
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Liqun Xu
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Changjun Zeng
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaoling Zhao
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Ming Zhang
- College of Animal Sciences and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| |
Collapse
|
29
|
Wang Y, Yang F, Yin H, He Q, Lu Y, Zhu Q, Lan X, Zhao X, Li D, Liu Y, Xu H. Chicken interferon regulatory factor 7 (IRF7) can control ALV-J virus infection by triggering type I interferon production through affecting genes related with innate immune signaling pathway. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:104026. [PMID: 33497733 DOI: 10.1016/j.dci.2021.104026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 06/12/2023]
Abstract
In order to breed new birds with strong disease resistance, it is necessary to first understand the mechanism of avian antiviral response. Interferon regulatory factor 7 (IRF7) is not only a member of type I interferons (IFNs) regulatory factor (IRFs) family, but also a major regulator of the IFN response in mammals. However, whether IRF7 is involved in the host innate immune response remains unclear in poultry, due to the absence of IRF3. Here, we first observed by HE stains that with the increase of the time of ALV-J challenge, the thymus was obviously loose and swollen, the arrangement of liver cell was disordered, and the bursa of fabricius formed vacuolated. Real-time PCR detection showed that the expression level of IRF7 gene and related immune genes in ALV-J group was significantly higher than that in control group (P < 0.05). To further study the role of chicken IRF7 during avian leukosis virus subgroup J (ALV-J) infection, we constructed an induced IRF7 overexpression and interfered chicken embryo fibroblasts (CEFs) cell and performed in vitro infection using low pathogenic ALV-J and virus analog poly(I:C). In ALV-J and poly(I:C) stimulated CEFs cells, the expression level of STAT1, IFN-α, IFN-β, TLR3 and TLR7 were increased after IRF7 overexpressed, while the results were just the opposite after IRF7 interfered, which indicating that IRF7 may be associated with Toll-like receptor signaling pathway and JAK-STAT signaling pathway. These findings suggest that chicken IRF7 is an important regulator of IFN and is involved in chicken anti-ALV-J innate immunity.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, 211# Huimin Road, Chengdu, 611130, China
| | - Fuling Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, 211# Huimin Road, Chengdu, 611130, China
| | - Huadong Yin
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, 211# Huimin Road, Chengdu, 611130, China
| | - Qijian He
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, 211# Huimin Road, Chengdu, 611130, China
| | - Yuxiang Lu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, 211# Huimin Road, Chengdu, 611130, China
| | - Qing Zhu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, 211# Huimin Road, Chengdu, 611130, China
| | - Xi Lan
- College of Animal Science and Technology, Southwest University, 2# Tiansheng Road, Beibei District Chongqing, 400715, China
| | - Xiaoling Zhao
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, 211# Huimin Road, Chengdu, 611130, China
| | - Diyan Li
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, 211# Huimin Road, Chengdu, 611130, China
| | - Yiping Liu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, 211# Huimin Road, Chengdu, 611130, China
| | - Hengyong Xu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, 211# Huimin Road, Chengdu, 611130, China.
| |
Collapse
|
30
|
Vathiotis IA, Johnson JM, Argiris A. Enhancing programmed cell death protein 1 axis inhibition in head and neck squamous cell carcinoma: Combination immunotherapy. Cancer Treat Rev 2021; 97:102192. [PMID: 33819755 DOI: 10.1016/j.ctrv.2021.102192] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 02/06/2023]
Abstract
Anti-programmed cell death protein 1 immunotherapy has become the new standard in the treatment of patients with recurrent or metastatic head and neck squamous cell carcinoma (HNSCC). However, the population that benefits is small, warranting drug combinations and novel approaches. HNSCC is a profoundly immunosuppressive disease, characterized by the interplay among different immune regulatory pathways. As clinical trials evaluating immunotherapy combinations in patients with HNSCC have started producing preliminary results, preclinical evidence on potential new targets for combination immunotherapy continues to accumulate. This review summarizes emerging clinical and preclinical data on immunotherapy combinations for the treatment of HNSCC.
Collapse
Affiliation(s)
- Ioannis A Vathiotis
- Department of Pathology, Yale University, 310 Cedar Street, New Haven, CT, USA
| | - Jennifer M Johnson
- Department of Medical Oncology, Thomas Jefferson University, 1025 Walnut Street, Suite 700, Philadelphia, PA, USA
| | - Athanassios Argiris
- Department of Medical Oncology, Thomas Jefferson University, 1025 Walnut Street, Suite 700, Philadelphia, PA, USA
| |
Collapse
|
31
|
Lu LF, Zhang C, Li ZC, Zhou XY, Jiang JY, Chen DD, Zhang YA, Xiong F, Zhou F, Li S. A novel role of Zebrafish TMEM33 in negative regulation of interferon production by two distinct mechanisms. PLoS Pathog 2021; 17:e1009317. [PMID: 33600488 PMCID: PMC7891750 DOI: 10.1371/journal.ppat.1009317] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/14/2021] [Indexed: 01/17/2023] Open
Abstract
The transmembrane protein 33 (TMEM33) was originally identified as an endoplasmic reticulum (ER) protein that influences the tubular structure of the ER and modulates intracellular calcium homeostasis. However, the role of TMEM33 in antiviral immunity in vertebrates has not been elucidated. In this article, we demonstrate that zebrafish TMEM33 is a negative regulator of virus-triggered interferon (IFN) induction via two mechanisms: mitochondrial antiviral signaling protein (MAVS) ubiquitination and a decrease in the kinase activity of TANK binding kinase 1 (TBK1). Upon stimulation with viral components, tmem33 was remarkably upregulated in the zebrafish liver cell line. The IFNφ1 promoter (IFNφ1pro) activity and mRNA level induced by retinoic acid-inducible gene (RIG)-I-like receptors (RLRs) were significantly inhibited by TMEM33. Knockdown of TMEM33 increased host ifn transcription. Subsequently, we found that TMEM33 was colocalized in the ER and interacted with the RLR cascades, whereas MAVS was degraded by TMEM33 during the K48-linked ubiquitination. On the other hand, TMEM33 reduced the phosphorylation of mediator of IFN regulatory factor 3 (IRF3) activation (MITA)/IRF3 by acting as a decoy substrate of TBK1, which was also phosphorylated. A functional domain assay revealed that the N-terminal transmembrane domain 1 (TM1) and TM2 regions of TMEM33 were necessary for IFN suppression. Finally, TMEM33 significantly attenuated the host cellular antiviral capacity by blocking the IFN response. Taken together, our findings provide insight into the different mechanisms employed by TMEM33 in cellular IFN-mediated antiviral process.
Collapse
Affiliation(s)
- Long-Feng Lu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Can Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhuo-Cong Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiao-Yu Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Jing-Yu Jiang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Dan-Dan Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Feng Xiong
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Fang Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Shun Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
32
|
Chamera K, Trojan E, Kotarska K, Szuster-Głuszczak M, Bryniarska N, Tylek K, Basta-Kaim A. Role of Polyinosinic:Polycytidylic Acid-Induced Maternal Immune Activation and Subsequent Immune Challenge in the Behaviour and Microglial Cell Trajectory in Adult Offspring: A Study of the Neurodevelopmental Model of Schizophrenia. Int J Mol Sci 2021; 22:ijms22041558. [PMID: 33557113 PMCID: PMC7913889 DOI: 10.3390/ijms22041558] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/22/2021] [Accepted: 02/01/2021] [Indexed: 12/21/2022] Open
Abstract
Multiple lines of evidence support the pathogenic role of maternal immune activation (MIA) in the occurrence of the schizophrenia-like disturbances in offspring. While in the brain the homeostatic role of neuron-microglia protein systems is well documented, the participation of the CX3CL1-CX3CR1 and CD200-CD200R dyads in the adverse impact of MIA often goes under-recognized. Therefore, in the present study, we examined the effect of MIA induced by polyinosinic:polycytidylic acid (Poly I:C) on the CX3CL1-CX3CR1 and CD200-CD200R axes, microglial trajectory (MhcII, Cd40, iNos, Il-1β, Tnf-α, Il-6, Arg1, Igf-1, Tgf-β and Il-4), and schizophrenia-like behaviour in adult male offspring of Sprague-Dawley rats. Additionally, according to the “two-hit” hypothesis of schizophrenia, we evaluated the influence of acute challenge with Poly I:C in adult prenatally MIA-exposed animals on the above parameters. In the present study, MIA evoked by Poly I:C injection in the late period of gestation led to the appearance of schizophrenia-like disturbances in adult offspring. Our results revealed the deficits manifested as a diminished number of aggressive interactions, presence of depressive-like episodes, and increase of exploratory activity, as well as a dichotomy in the sensorimotor gating in the prepulse inhibition (PPI) test expressed as two behavioural phenotypes (MIAPPI-low and MIAPPI-high). Furthermore, in the offspring rats subjected to a prenatal challenge (i.e., MIA) we noticed the lack of modulation of behavioural changes after the additional acute immune stimulus (Poly I:C) in adulthood. The important finding reported in this article is that MIA affects the expression and levels of the neuron-microglia proteins in the frontal cortex and hippocampus of adult offspring. We found that the changes in the CX3CL1-CX3CR1 axis could affect microglial trajectory, including decreased hippocampal mRNA level of MhcII and elevated cortical expression of Igf-1 in the MIAPPI-high animals and/or could cause the up-regulation of an inflammatory response (Il-6, Tnf-α, iNos) after the “second hit” in both examined brain regions and, at least in part, might differentiate behavioural disturbances in adult offspring. Consequently, the future effort to identify the biological background of these interactions in the Poly I:C-induced MIA model in Sprague-Dawley rats is desirable to unequivocally clarify this issue.
Collapse
|
33
|
Wang Z, Li L, Liu P, Wang C, Lu Q, Liu L, Yang Y, Luo Q, Shao H. Host innate immune responses of geese infected with goose origin nephrotic astrovirus. Microb Pathog 2021; 152:104753. [PMID: 33516903 DOI: 10.1016/j.micpath.2021.104753] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 11/28/2022]
Abstract
A novel goose astrovirus (GoAstV) outbreak in goslings, characterized by severe articular and visceral gout with high mortality, occurred in China. Although the pathogenesis of GoAstV-infected goslings has been explored in several studies, the host-immune response remains unclear. In this study, a goose astrovirus was isolated from goslings in Xiaogan, and designated as the HBXG strain. The full-length genome of HBXG was 7170 nt. A sequence analysis and phylogenetic trees revealed HBXG belonged to the novel GoAstV. We evaluated the viral distribution systematically and estimated immune related gene expression in HBXG-infected goslings. Results showed that GoAstV replicated quickly in many tissues and the highest titer was observed in the kidney, which reached 109.6 copies. TLR3, RIG-I and MDA5 were involved in the host-immune response to GoAstV, and the expression of IFN types I (IFN-α, IFN-β), inflammatory cytokines (IL-8, IL-10, TNF-α), antiviral proteins (Mx, OASL, PKR) and MHC-I were also upregulated during the infection. In contrast, the expression of proinflammatory cytokines (IL-1β, IL-6) and MHC-II were inhibited at 3 dpi. This study suggests that GoAstV is highly pathogenic to goslings, causing multiple systemic infections in tissues and the host-immune response is activated early in infection. However, rapid viral replication, suppression of inflammatory cytokines (IL-1β, IL-6) and MHC-II expressions were the possible reasons why the host-immune response cannot provide enough protection against GoAstV infection. This study is the first report to illuminate the immune response in goslings infected with GoAstV and offers insight into the pathogenesis of GoAstV.
Collapse
Affiliation(s)
- Zui Wang
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Special One, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China.
| | - Li Li
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Special One, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China.
| | - Peng Liu
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Special One, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis, Special 1, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China.
| | - Chen Wang
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Special One, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis, Special 1, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China.
| | - Qin Lu
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Special One, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China.
| | - Lina Liu
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Special One, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China
| | - Yuying Yang
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Special One, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China; College of Animal Sciences, Yangtze University, Jingzhou, 434025, China.
| | - Qingping Luo
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Special One, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis, Special 1, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China.
| | - Huabin Shao
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Special One, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China; Key Laboratory of Prevention and Control Agents for Animal Bacteriosis, Special 1, Nanhuyaoyuan, Hongshan District, Wuhan, 430064, China.
| |
Collapse
|
34
|
Hepatitis A virus-induced hsa-miR-146a-5p attenuates IFN-β signaling by targeting adaptor protein TRAF6. Arch Virol 2021; 166:789-799. [PMID: 33459883 DOI: 10.1007/s00705-021-04952-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/26/2020] [Indexed: 12/21/2022]
Abstract
Hepatitis A virus (HAV), a unique hepatotropic human picornavirus, is the causative agent of acute hepatitis A in humans. Some studies have shown that HAV antagonizes the innate immune response by disrupting interferon-beta (IFN-β) signaling by viral proteins. However, whether microRNAs (miRNAs), a class of non-coding RNAs, are involved in the antagonism of IFN-β induction upon HAV infection is still unclear. In this study, we investigated the effects and mechanisms by which HAV-induced miRNAs antagonize IFN-β signaling. A variety of analytical methods, including miRNA microarray, RT-qPCR, dual-luciferase reporter assay, and Western blotting, were performed using HAV-infected cells. The results indicated that HAV infection upregulates the expression of hsa-miR-146a-5p, which in turn partially suppresses the induction of IFN-β synthesis, thereby promoting viral replication. Mechanistically, TRAF6 (TNF receptor-associated factor 6), a key adaptor protein in the RIG-I/MDA5-mediated IFN-I signaling pathway, is targeted and degraded by hsa-miR-146a-5p. As TRAF6 is necessary for IFN-β induction, inhibition of this protein attenuates IFN-β signaling. Taken together, the results from this study indicated that HAV disrupts RIG-I/MDA5-mediated IFN-I signaling partially through the cleavage of the essential adaptor molecule TRAF6 via hsa-miR-146a-5p.
Collapse
|
35
|
Maurya DK, Sharma D. Evaluation of traditional ayurvedic Kadha for prevention and management of the novel Coronavirus (SARS-CoV-2) using in silico approach. J Biomol Struct Dyn 2020; 40:3949-3964. [PMID: 33251972 PMCID: PMC7754934 DOI: 10.1080/07391102.2020.1852119] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since the emergence of novel Coronavirus (SARS-CoV-2) infection in Wuhan, China in December 2019, it has now spread to over 205 countries. The ever-growing list of globally spread corona virus-19 disease (COVID-19) patients has demonstrated the high transmission rate among the human population. Currently, there are no FDA approved drugs or vaccines to prevent and treat the infection of the SARS-CoV-2. Considering the current state of affairs, there is an urgent unmet medical need to identify novel and effective approaches for the prevention and treatment of COVID-19 by re-evaluating the knowledge of traditional medicines and repurposing of drugs. Here, we used molecular docking and molecular dynamics simulation approach to explore the beneficial roles of phytochemicals and active pharmacological agents present in the Indian herbs which are widely used in the preparation of Ayurvedic medicines in the form of Kadha to control various respiratory disorders such as cough, cold and flu. Our study has identified an array of phytochemicals present in these herbs which have significant docking scores and potential to inhibit different stages of SARS-CoV-2 infection as well as other Coronavirus target proteins. The phytochemicals present in these herbs possess significant anti-inflammatory property. Apart from this, based on their pharmaceutical characteristics, we have also performed in-silico drug-likeness and predicted pharmacokinetics of the selected phytochemicals found in the Kadha. Overall our study provides scientific justification in terms of binding of active ingredients present in different plants used in Kadha preparation with viral proteins and target proteins for prevention and treatment of the COVID-19. Communicated by Ramaswamy H. Sarma
Collapse
Affiliation(s)
- Dharmendra Kumar Maurya
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India.,Homi Bhabha National Institute, Anushaktinagar, India
| |
Collapse
|
36
|
Rasaei R, Sarodaya N, Kim KS, Ramakrishna S, Hong SH. Importance of Deubiquitination in Macrophage-Mediated Viral Response and Inflammation. Int J Mol Sci 2020; 21:ijms21218090. [PMID: 33138315 PMCID: PMC7662591 DOI: 10.3390/ijms21218090] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 01/14/2023] Open
Abstract
Ubiquitination and deubiquitination play a fundamental role in the signaling pathways associated with innate and adaptive immune responses. Macrophages are key sentinels for the host defense, triggering antiviral and inflammatory responses against various invading pathogens. Macrophages recognize the genetic material of these pathogens as pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) through the activation of its pattern recognition receptors (PRRs), initiating the cascade of immune signaling, which leads to the production of pro- and anti-inflammatory cytokines that initiates the appropriate immune response. Macrophage-mediated immune response is highly regulated and tightly controlled by the ubiquitin system since its abnormal activation or dysregulation may result in the severe pathogenesis of numerous inflammatory and autoimmune diseases. Deubiquitinating enzymes (DUBs) play a crucial role in reversing the ubiquitination and controlling the magnitude of the immune response. During infection, pathogens manipulate the host defense system by regulating DUBs to obtain nutrients and increase proliferation. Indeed, the regulation of DUBs by small molecule inhibitors has been proposed as an excellent way to control aberrant activation of immune signaling molecules. This review is focused on the complex role of DUBs in macrophage-mediated immune response, exploring the potential use of DUBs as therapeutic targets in autoimmune and inflammatory diseases by virtue of small molecule DUB inhibitors.
Collapse
Affiliation(s)
- Roya Rasaei
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea;
| | - Neha Sarodaya
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (N.S.); (K.-S.K.)
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (N.S.); (K.-S.K.)
- College of Medicine, Hanyang University, Seoul 04763, Korea
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea; (N.S.); (K.-S.K.)
- College of Medicine, Hanyang University, Seoul 04763, Korea
- Correspondence: or (S.R.); or (S.-H.H.)
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea;
- Correspondence: or (S.R.); or (S.-H.H.)
| |
Collapse
|
37
|
Ibi D, Nakasai G, Koide N, Sawahata M, Kohno T, Takaba R, Nagai T, Hattori M, Nabeshima T, Yamada K, Hiramatsu M. Reelin Supplementation Into the Hippocampus Rescues Abnormal Behavior in a Mouse Model of Neurodevelopmental Disorders. Front Cell Neurosci 2020; 14:285. [PMID: 32982694 PMCID: PMC7492784 DOI: 10.3389/fncel.2020.00285] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022] Open
Abstract
In the majority of schizophrenia patients, chronic atypical antipsychotic administration produces a significant reduction in or even complete remission of psychotic symptoms such as hallucinations and delusions. However, these drugs are not effective in improving cognitive and emotional deficits in patients with schizophrenia. Atypical antipsychotic drugs have a high affinity for the dopamine D2 receptor, and a modest affinity for the serotonin 5-HT2A receptor. The cognitive and emotional deficits in schizophrenia are thought to involve neural networks beyond the classical dopaminergic mesolimbic pathway, however, including serotonergic systems. For example, mutations in the RELN gene, which encodes Reelin, an extracellular matrix protein involved in neural development and synaptic plasticity, are associated with neurodevelopmental disorders such as schizophrenia and autism spectrum disorder. Furthermore, hippocampal Reelin levels are down-regulated in the brains of both schizophrenic patients and in rodent models of schizophrenia. In the present study, we investigated the effect of Reelin microinjection into the mouse hippocampus on behavioral phenotypes to evaluate the role of Reelin in neurodevelopmental disorders and to test a therapeutic approach that extends beyond classical monoamine targets. To model the cognitive and emotional deficits, as well as histological decreases in Reelin-positive cell numbers and hippocampal synaptoporin distribution, a synaptic vesicle protein, offspring that were prenatally exposed to maternal immune activation were used. Microinjections of recombinant Reelin protein into the hippocampus rescued impairments in object memory and anxiety-like behavior and recruited synaptoporin in the hippocampus in offspring exposed to antenatal inflammation. These results suggest that Reelin supplementation has the potential to treat cognitive and emotional impairments, as well as synaptic disturbances, in patients with neurodevelopmental disorders such as schizophrenia.
Collapse
Affiliation(s)
- Daisuke Ibi
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Genki Nakasai
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Nayu Koide
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Masahito Sawahata
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takao Kohno
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Rika Takaba
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Project Office for Neuropsychological Research Center, Fujita Health University, Toyoake, Japan
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University, Graduate School of Health Sciences, Toyoake, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masayuki Hiramatsu
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| |
Collapse
|
38
|
Yan J, Zhang Y, Tan Y, Dai Y, Wei J, Cao Y, Feng H. Black carp TRAFD1 restrains MAVS-mediated antiviral signaling during the innate immune activation. FISH & SHELLFISH IMMUNOLOGY 2020; 103:66-72. [PMID: 32334128 DOI: 10.1016/j.fsi.2020.04.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/05/2020] [Accepted: 04/19/2020] [Indexed: 06/11/2023]
Abstract
TRAFD1 negatively regulates TLR and RLR signaling in human and mammal; however, its role in teleost fish remains unknown. In this paper, the TRAFD1 homologue has been cloned and characterized from black carp (Mylopharyngodon piceus). Black carp TRAFD1 (bcTRAFD1) consists of 567 amino acids and shows low similarity to that of mammalian TRAFD1, which has been identified as a cytosolic protein through immunofluorescence staining. When co-expressed with bcTRAFD1, the IFN promoter-inducing ability of black carp MAVS (bcMAVS) was obviously dampened in the luciferase reporter assay. Accordingly, bcMAVS-mediated antiviral activity against grass carp reovirus (GCRV) and spring viremia of carp virus (SVCV) was potently repressed by bcTRAFD1 in plaque assay. And the co-immunoprecipitation assay between bcTRAFD1 and bcMAVS has identified the association between these two molecules. Thus, our data supports the conclusion that bcTRAFD1 interacts with bcMAVS and negatively regulates bcMAVS-mediated antiviral signaling during the innate immune activation, which sheds a light on the regulation of MAVS in teleost.
Collapse
Affiliation(s)
- Jun Yan
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yinyin Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yaqi Tan
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yuhan Dai
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jing Wei
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yingyi Cao
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Hao Feng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
39
|
Kolodny O, Berger M, Feldman MW, Ram Y. A new perspective for mitigation of SARS-CoV-2 infection: priming the innate immune system for viral attack. Open Biol 2020; 10:200138. [PMID: 36416599 PMCID: PMC7574546 DOI: 10.1098/rsob.200138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/11/2020] [Indexed: 12/14/2022] Open
Abstract
The course of infection by SARS-CoV-2 frequently includes a long asymptomatic period, followed in some individuals by an immune dysregulation period that may lead to complications and immunopathology-induced death. This course of disease suggests that the virus often evades detection by the innate immune system. We suggest a novel therapeutic approach to mitigate the infection's severity, probability of complications and duration. We propose that priming an individual's innate immune system for viral attack shortly before it is expected to occur may allow pre-activation of the preferable trajectory of immune response, leading to early detection of the virus. Priming can be carried out, for example, by administering a standard vaccine or another reagent that elicits a broad anti-viral innate immune response. By the time that the expected SARS-CoV-2 infection occurs, activation cascades will have been put in motion and levels of immune factors needed to combat the infection will have been elevated. The infection would thus be cleared faster and with less complication than otherwise, alleviating adverse clinical outcomes at the individual level. Moreover, priming may also mitigate population-level risk by reducing need for hospitalizations and decreasing the infectious period of individuals, thus slowing the spread and reducing the impact of the epidemic. In view of the latter consideration, our proposal may have a significant epidemiological impact even if applied primarily to low-risk individuals, such as young adults, who often show mild symptoms or none, by shortening the period during which they unknowingly infect others. The proposed view is, at this time, an unproven hypothesis. Although supported by robust bio-medical reasoning and multiple lines of evidence, carefully designed clinical trials are necessary.
Collapse
Affiliation(s)
- Oren Kolodny
- Department of Ecology, Evolution and Behavior, Alexander Silberman, Institute of Life Sciences, The Hebrew University of Jerusalem, 9190401 Jerusalem, Israel
| | - Michael Berger
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem–Hadassah Medical School, Israel
| | | | - Yoav Ram
- School of Computer Science, Interdisciplinary Center Herzliya, Israel
| |
Collapse
|
40
|
Ben-Yehuda H, Matcovitch-Natan O, Kertser A, Spinrad A, Prinz M, Amit I, Schwartz M. Maternal Type-I interferon signaling adversely affects the microglia and the behavior of the offspring accompanied by increased sensitivity to stress. Mol Psychiatry 2020; 25:1050-1067. [PMID: 31772304 PMCID: PMC7192855 DOI: 10.1038/s41380-019-0604-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 11/10/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
Abstract
Viral infection during pregnancy is often associated with neuropsychiatric conditions. In mice, exposure of pregnant dams to the viral mimetic poly(I:C), serves as a model that simulates such pathology in the offspring, through a process known as Maternal Immune Activation (MIA). To investigate the mechanism of such effect, we hypothesized that maternal upregulation of Type-I interferon (IFN-I), as part of the dam's antiviral response, might contribute to the damage imposed on the offspring. Using mRNA sequencing and flow cytometry analyses we found that poly(I:C) treatment during pregnancy caused reduced expression of genes related to proliferation and cell cycle in the offspring's microglia relative to controls. This was found to be associated with an IFN-I signature in the embryonic yolk sac, the origin of microglia in development. Neutralizing IFN-I signaling in dams attenuated the effect of MIA on the newborn's microglia, while systemic maternal administration of IFNβ was sufficient to mimic the effect of poly(I:C), and led to increased vulnerability of offspring's microglia to subsequent stress. Furthermore, maternal elevation of IFNβ resulted in behavioral manifestations reminiscent of neuropsychiatric disorders. In addition, by adopting a "two-hit" experimental paradigm, we show a higher sensitivity of the offspring to postnatal stress subsequent to the maternal IFNβ elevation, demonstrated by behavioral irregularities. Our results suggest that maternal upregulation of IFN-I, in response to MIA, interferes with the offspring's programmed microglial developmental cascade, increases their susceptibility to postnatal stress, and leads to behavioral abnormalities.
Collapse
Affiliation(s)
- Hila Ben-Yehuda
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Orit Matcovitch-Natan
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Kertser
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Amit Spinrad
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
41
|
Zhang C, Lu LF, Li ZC, Zhou XY, Zhou Y, Chen DD, Li S, Zhang YA. Grass carp reovirus VP56 represses interferon production by degrading phosphorylated IRF7. FISH & SHELLFISH IMMUNOLOGY 2020; 99:99-106. [PMID: 32032764 PMCID: PMC7111710 DOI: 10.1016/j.fsi.2020.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/16/2020] [Accepted: 02/03/2020] [Indexed: 05/14/2023]
Abstract
Grass carp reovirus (GCRV) is an efficient pathogen causing high mortality in grass carp, meanwhile, fish interferon (IFN) is a powerful cytokine enabling host cells to establish an antiviral state; therefore, the strategies used by GCRV to escape the cellular IFN response need to be investigated. Here, we report that GCRV VP56 inhibits host IFN production by degrading the transcription factor IFN regulatory factor 7 (IRF7). First, overexpression of VP56 inhibited the IFN production induced by the polyinosinic-polycytidylic acid (poly I:C) and mitochondrial antiviral signaling protein (MAVS), while the capacity of IRF7 on IFN induction was unaffected. Second, VP56 interacted with RLRs but did not affect the stabilization of the proteins in the normal state, while the phosphorylated IRF7 activated by TBK1 was degraded by VP56 through K48-linked ubiquitination. Finally, overexpression of VP56 remarkably reduced the host cellular ifn transcription and facilitated viral proliferation. Taken together, our results demonstrate that GCRV VP56 suppresses the host IFN response by targeting phosphorylated IRF7 for ubiquitination and degradation.
Collapse
Affiliation(s)
- Can Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Long-Feng Lu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Zhuo-Cong Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xiao-Yu Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yu Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Dan-Dan Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Shun Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
42
|
Yu S, Mao H, Jin M, Lin X. Transcriptomic Analysis of the Chicken MDA5 Response Genes. Genes (Basel) 2020; 11:E308. [PMID: 32183248 PMCID: PMC7140832 DOI: 10.3390/genes11030308] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 11/29/2022] Open
Abstract
RIG-I and MDA5 are two key pattern recognition receptors that sense RNA virus invasion, but RIG-I is absent in chickens. Although chickens have intact MDA5, the genes downstream of chicken MDA5 (chMDA5) that may mediate antiviral response are not well studied. We compared the transcriptional profile of chicken embryonic fibroblasts (DF1) transfected with chMDA5, and poly(I:C), using RNA-seq. Transfected chMDA5 and poly(I:C) in DF1 cells were associated with the marked induction of many antiviral innate immune genes compared with control. Interestingly, nine interferon-stimulated genes (ISGs) were listed in the top 15 upregulated genes by chMDA5 and poly(I:C) transfection. We used real-time PCR to confirm the upregulation of the nine ISGs, namely, MX1, IFI6, IFIT5, RSAD2, OASL, CMPK2, HELZ2, EPSTI1, and OLFML1, by chMDA5 and poly(I:C) transfection in DF1 cells. However, avian influenza virus H5N6 infection only increased MX1, IFI6, IFIT5, RSAD2, and OASL expression levels. Further study showed that the overexpression of these five genes could significantly inhibit H5N6 virus replication. These results provide some insights into the gene expression pattern induced by chMDA5, which would be beneficial for understanding and identifying innate immune genes of chicken that may lead to new antiviral therapies.
Collapse
Affiliation(s)
- Shiman Yu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (S.Y.); (H.M.); (M.J.)
- Department of Preventive Veterinary Medicine, College of Animal Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Haiying Mao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (S.Y.); (H.M.); (M.J.)
- Department of Preventive Veterinary Medicine, College of Animal Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Meilin Jin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (S.Y.); (H.M.); (M.J.)
- Department of Preventive Veterinary Medicine, College of Animal Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Xian Lin
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (S.Y.); (H.M.); (M.J.)
- Department of Preventive Veterinary Medicine, College of Animal Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Department of Biotechnology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
43
|
Wang ZX, Zhou Y, Lu LF, Lu XB, Ni B, Liu MX, Guan HX, Li S, Zhang YA, Ouyang S. Infectious hematopoietic necrosis virus N protein suppresses fish IFN1 production by targeting the MITA. FISH & SHELLFISH IMMUNOLOGY 2020; 97:523-530. [PMID: 31881328 DOI: 10.1016/j.fsi.2019.12.075] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 06/10/2023]
Abstract
Interferon (IFN) is a vital antiviral factor in host in the early stages after the viral invasion. Meanwhile, viruses have to survive by taking advantage of the cellular machinery and complete their replication. As a result, viruses evolved several immune escape mechanisms to inhibit host IFN expression. However, the mechanisms used to escape the host's IFN system are still unclear for infectious hematopoietic necrosis virus (IHNV). In this study, we report that the N protein of IHNV inhibits IFN1 production in rainbow trout by degrading the MITA. Firstly, the upregulation of IFN1 promoter activity stimulated by poly I:C was suppressed by IHNV infection. Consistent with this result, the overexpression of the N protein of IHNV blocked the IFN1 transcription that was activated by poly I:C and MITA. Secondly, MITA was remarkably decreased by the overexpression of N protein at the protein level. Further analysis demonstrated that the N protein targeted MITA and promoted the ubiquitination of MITA. Taken together, these data suggested that the production of rainbow trout IFN1 could be suppressed by the N protein of IHNV via degrading MITA.
Collapse
Affiliation(s)
- Zhao-Xi Wang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, The Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, China; University of Chinese Academy of Science, Beijing, China
| | - Yu Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Science, Beijing, China
| | - Long-Feng Lu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Xiao-Bing Lu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Bo Ni
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, The Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, China; The Public Service Platform for Industrialization Development Technology of Marine Biological Medicine and Product of State Oceanic Administration, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, China
| | - Meng-Xi Liu
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, The Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, China; Fujian Key Laboratory of Special Marine Bio-resources Sustainable Utilization, Fujian Normal University, Fuzhou, 350117, China
| | - Hong-Xin Guan
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, The Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, China; The Public Service Platform for Industrialization Development Technology of Marine Biological Medicine and Product of State Oceanic Administration, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, China; Fujian Key Laboratory of Special Marine Bio-resources Sustainable Utilization, Fujian Normal University, Fuzhou, 350117, China
| | - Shun Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), 266337, China; State Key Laboratory of Aquaculture Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, China.
| | - Songying Ouyang
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), 266337, China; Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, The Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, Key Laboratory of OptoElectronic Science and Technology for Medicine of the Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, China; The Public Service Platform for Industrialization Development Technology of Marine Biological Medicine and Product of State Oceanic Administration, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, China; Fujian Key Laboratory of Special Marine Bio-resources Sustainable Utilization, Fujian Normal University, Fuzhou, 350117, China
| |
Collapse
|
44
|
Frascaroli G, Rossini G, Maltoni V, Bartoletti M, Ortolani P, Gredmark-Russ S, Gelsomino F, Moroni A, Silenzi S, Castellani G, Sambri V, Mastroianni A, Brune W, Varani S. Genetic and Functional Characterization of Toll-Like Receptor Responses in Immunocompetent Patients With CMV Mononucleosis. Front Cell Infect Microbiol 2020; 10:386. [PMID: 32850485 PMCID: PMC7426556 DOI: 10.3389/fcimb.2020.00386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/24/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Human cytomegalovirus (CMV) modulates both innate and adaptive immune responses. However, limited data are available on the role of receptors of innate immunity, such as toll-like receptors (TLRs) in contributing to antiviral responses and inflammation. Objectives: The aim of this translational study was to characterize TLR responses in immunocompetent patients with primary and symptomatic CMV infection. Study Design: The study population consisted of 40 patients suffering from CMV mononucleosis and 124 blood donors included as controls. We evaluated the association between TLR2, 3, 4, 7 and 9 gene single nucleotide polymorphism (SNP) and susceptibility to symptomatic CMV infection in immunocompetent adults. Additionally, functional TLR-mediated cytokine responses in supernatants of short-term cultures of whole blood from patients with CMV mononucleosis and blood donors were evaluated. Results: TLR2 and TLR7/8 responses were altered in CMV infected patients as compared to healthy donors and were associated with the release of higher levels of the pro-inflammatory cytokines IL-6 and TNF-α, but not of the anti-inflammatory mediator IL-10. The analysis on the TLR SNPs indicated no difference between patients with CMV infection and the control group. Conclusions: No variation in the TLR2,3,4,7 and 9 genes was associated to the development of symptomatic CMV infection in immunocompetent adults. Nevertheless, TLR-mediated responses in CMV-infected patients appeared to be skewed toward a pro-inflammatory profile, which may contribute to the development of inflammatory symptoms during the CMV mononucleotic syndrome.
Collapse
Affiliation(s)
- Giada Frascaroli
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
- *Correspondence: Giada Frascaroli
| | - Giada Rossini
- Unit of Microbiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Virginia Maltoni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Michele Bartoletti
- Infectious Diseases Unit, Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | | | - Sara Gredmark-Russ
- Center for Infectious Medicine, ANA Futura, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Francesco Gelsomino
- Unit of Microbiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Alessandra Moroni
- Unit of Microbiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Silvia Silenzi
- Unit of Microbiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Gastone Castellani
- Department of Physics and Astronomy and Galvani Center for Biocomplexity, University of Bologna, Bologna, Italy
| | - Vittorio Sambri
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- Unit of Microbiology, The Romagna Hub Laboratory, Pievesestina, Italy
| | - Antonio Mastroianni
- Unit of Infectious and Tropical Diseases, St. Annunziata Hospital, Cosenza, Italy
- Unit of Infectious Diseases, G.B. Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Wolfram Brune
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Stefania Varani
- Unit of Microbiology, St. Orsola-Malpighi University Hospital, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| |
Collapse
|
45
|
Hassan E, Baldridge MT. Norovirus encounters in the gut: multifaceted interactions and disease outcomes. Mucosal Immunol 2019; 12:1259-1267. [PMID: 31501514 PMCID: PMC7479810 DOI: 10.1038/s41385-019-0199-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/20/2019] [Indexed: 02/04/2023]
Abstract
Noroviruses are major causes of gastroenteritis, with epidemic outbreaks occurring frequently. They are an important global health concern, especially for pediatric and immunocompromised populations, and are challenging pathogens to target immunologically due to their rapid rates of genetic and antigenic evolution and failure to stimulate durable protective immunity. In this Review, we summarize our current understanding of norovirus pathogenesis, noting the prominent role of murine norovirus as a small animal model for norovirus research. We highlight intriguing data supporting the possible involvement of norovirus in sequelae including irritable bowel syndrome and inflammatory bowel diseases, and describe the innate and adaptive immune mechanisms involved in control of both human and murine norovirus infection. Furthermore, we discuss the potential implications of recent discoveries regarding norovirus interactions with the gut microbiota, and briefly detail current understanding of noroviral evolution and its influence on viral pathogenesis. Our mechanistic understanding of norovirus pathogenesis continues to improve with increasing availability of powerful model systems, which will ultimately facilitate development of effective preventive and therapeutic approaches for this pathogen.
Collapse
Affiliation(s)
- Ebrahim Hassan
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Megan T. Baldridge
- Division of Infectious Diseases, Department of Medicine, Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.,Correspondence: Megan T. Baldridge, MD, PhD, Phone: 314-273-1212, Fax: 314-362-2156,
| |
Collapse
|
46
|
Bull JJ, Nuismer SL, Antia R. Recombinant vector vaccine evolution. PLoS Comput Biol 2019; 15:e1006857. [PMID: 31323032 PMCID: PMC6668849 DOI: 10.1371/journal.pcbi.1006857] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 07/31/2019] [Accepted: 06/07/2019] [Indexed: 01/01/2023] Open
Abstract
Replicating recombinant vector vaccines consist of a fully competent viral vector backbone engineered to express an antigen from a foreign transgene. From the perspective of viral replication, the transgene is not only dispensable but may even be detrimental. Thus vaccine revertants that delete or inactivate the transgene may evolve to dominate the vaccine virus population both during the process of manufacture of the vaccine as well as during the course of host infection. A particular concern is that this vaccine evolution could reduce its antigenicity—the immunity elicited to the transgene. We use mathematical and computational models to study vaccine evolution and immunity. These models include evolution arising during the process of manufacture, the dynamics of vaccine and revertant growth, plus innate and adaptive immunity elicited during the course of infection. Although the selective basis of vaccine evolution is easy to comprehend, the immunological consequences are not. One complication is that the opportunity for vaccine evolution is limited by the short period of within-host growth before the viral population is cleared. Even less obvious, revertant growth may only weakly interfere with vaccine growth in the host and thus have a limited effect on immunity to vaccine. Overall, we find that within-host vaccine evolution can sometimes compromise vaccine immunity, but only when the extent of evolution during vaccine manufacture is severe, and this evolution can be easily avoided or mitigated. Recombinant vector vaccines are live replicating viruses that are engineered to carry extra genes derived from a pathogen—and these extra genes produce proteins against which we want to generate immunity. These vaccine genomes may evolve to lose the extra genes during the process of manufacture of the vaccine or during replication within an individual, and there is a concern that this evolution might severely limit the vaccine’s efficacy. The dynamics of this process are studied here with mathematical models. The potential for vaccine evolution within the host is somewhat limited by the short-term growth of the vaccine population before it is suppressed by the immune response. We find that evolution is a problem only when the process of manufacture results in the majority of the vaccine virus being revertant. We show that increasing the vaccine inoculum size or reducing the level of revertant in the vaccine inoculum can largely avoid the loss of immunity arising from evolution.
Collapse
Affiliation(s)
- James J. Bull
- Department Integrative Biology, University of Texas, Austin, Texas, United States of America
- * E-mail:
| | - Scott L. Nuismer
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, United States of America
| | - Rustom Antia
- Department of Biology, Emory University, Altanta, Georgia, United States of America
| |
Collapse
|
47
|
Freer G, Maggi F, Pistello M. Virome and Inflammasomes, a Finely Tuned Balance with Important Consequences for the Host Health. Curr Med Chem 2019; 26:1027-1044. [PMID: 28982318 DOI: 10.2174/0929867324666171005112921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 07/06/2017] [Accepted: 07/27/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND The virome is a network of viruses normally inhabiting humans. It forms a conspicuous portion of the so-called microbiome, once generically referred to as resident flora. Indeed, viruses infecting humans without leading to clinical disease are increasingly recognized as part of the microbiome and have an impact on the development of our immune system. In addition, they activate inflammasomes, multiprotein complexes that assemble in cells and that are responsible for the downstream effects of sensing pathogens. OBJECTIVE This review aims at summarizing the evidence on the role of the virome in modulating inflammation and emphasizes evidence for Anelloviruses as useful molecular markers to monitor inflammatory processes and immune system competence. METHOD We carried out a review of the literature published in the last 5 years and summarized older literature to take into account ground-breaking discoveries concerning inflammasome assembly and virome. RESULTS A massive amount of data recently emerging demonstrate that the microbiome closely reflects what we eat, and many other unexpected variables. Composition, location, and amount of the microbiome have an impact on innate and adaptive immune defences. Viruses making up the virome contribute to shaping the immune system. Anelloviruses, the best known of such viruses, are present in most human beings, persistently without causing apparent disease. Depending on their interplay with such viruses, inflammasomes instruct host defences to tolerate or forfeit a specific microorganism. CONCLUSION The virome plays an important role in shaping human immune defences and contributes to inflammatory processes by quenching or increasing them.
Collapse
Affiliation(s)
- Giulia Freer
- Retrovirus Center and Virology Section, Department of Translational Research, University of Pisa, Pisa, Italy
| | | | - Mauro Pistello
- Retrovirus Center and Virology Section, Department of Translational Research, University of Pisa, Pisa, Italy.,Virology Unit, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
48
|
Krishnan R, Kurcheti PP, Mushtaq Z, K J, Naik T V. Interferon-regulatory factors, IRF3 and IRF7 in Asian seabass, Lates calcarifer: Characterization, ontogeny and transcriptional modulation upon challenge with nervous necrosis virus. FISH & SHELLFISH IMMUNOLOGY 2019; 89:468-476. [PMID: 30940578 DOI: 10.1016/j.fsi.2019.03.073] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/11/2019] [Accepted: 03/28/2019] [Indexed: 06/09/2023]
Abstract
Interferon regulatory factor (IRF) 3 and IRF7 are key regulators of type I interferon (IFN) gene expression for the antiviral immune response. In the present study, interferon regulatory factor 3 and 7 from Asian seabass, namely AsIRF3 and AsIRF7 were cloned and characterized. The full-length cDNA sequence of IRF3 and IRF7 consisted of 2965 and 2343 bp respectively. AsIRF3 and AsIRF7 were true orthologes of vertebrate IRF3/7 and showed similar domain organization, with an N-terminal DBD which consisted five tryptophan residues in IRF3 and four in IRF7, a C-terminal IRF3 domain and a serine rich region. Both IRF3 and 7 constitutively expressed during the ontogenesis and in all tissues of healthy fish. The expression of both genes was up-regulated following NNV challenge with obvious transcript abundance in brain heart and kidney. Ectopic expression of AsIRF3 and AsIRF7 displayed activation of ISRE/NF-κB promoters and modulation of interferon, ISGs and pro-inflammatory cytokine gene expression. These observations indicated that IRF3 and IRF7 play an important role in Asian seabass's antiviral defense and the RIG-IRF-IFN axis is conserved in the species.
Collapse
Affiliation(s)
- Rahul Krishnan
- Aquatic Environment and Health Management Division, ICAR- Central Institute of Fisheries Education, Mumbai, 400061, India; Present Address: Department of Aqualife Medicine, Chonnam National University, Republic of Korea
| | - Pani Prasad Kurcheti
- Aquatic Environment and Health Management Division, ICAR- Central Institute of Fisheries Education, Mumbai, 400061, India.
| | - Zahoor Mushtaq
- Aquatic Environment and Health Management Division, ICAR- Central Institute of Fisheries Education, Mumbai, 400061, India
| | - Jeena K
- Aquatic Environment and Health Management Division, ICAR- Central Institute of Fisheries Education, Mumbai, 400061, India
| | - Vismai Naik T
- Aquatic Environment and Health Management Division, ICAR- Central Institute of Fisheries Education, Mumbai, 400061, India
| |
Collapse
|
49
|
Activated dendritic cells and monocytes in HIV immunological nonresponders: HIV-induced interferon-inducible protein-10 correlates with low future CD4+ recovery. AIDS 2019; 33:1117-1129. [PMID: 30789356 PMCID: PMC6511429 DOI: 10.1097/qad.0000000000002173] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Supplemental Digital Content is available in the text Objective: To explore monocyte and dendritic cell immune responses, and their association with future CD4+ gain in treated HIV patients with suboptimal CD4+ recovery. Design: A cross-sectional study of HIV-infected, virally suppressed individuals on antiretroviral therapy for at least 24 months; 41 immunological nonresponders (INRs) (CD4+ cell count <400 cells/μl) and 26 immunological responders (CD4+ cell count >600 cells/μl). Ten HIV-infected antiretroviral therapy-naive and 10 HIV-negative healthy persons served as controls. CD4+ cell counts were registered after median 2.4 and 4.7 years. Methods: Monocyte, dendritic-cell and T-cell activation and regulatory T cells (Tregs) were analyzed by flow cytometry. In INR and immunological responder subgroups matched on age and nadir CD4+ cell count, upregulation of interferon-inducible protein-10 (IP-10) and indoleamine 2,3-dioxygenase in monocytes and dendritic cells and cytokines in cell supernatants were measured in vitro in peripheral blood mononuclear cells stimulated with aldrithiol-2-inactivated HIV-1. Results: The INR group displayed higher spontaneous activation of both monocytes (HLA-DR+) and myeloid and plasmacytoid dendritic cells (HLA-DR+, CD83+ and CD86+) compared with immunological responders, and this was associated with increased T-cell activation (CD38+HLA-DR+), an effector memory T-cell phenotype and activated Tregs. The IP-10 response in monocytes after in-vitro HIV stimulation was negatively associated with prospective CD4+ gain. IP-10, indoleamine 2,3-dioxygenase and cytokines levels were comparable between the groups, but inversely correlated with activated Tregs in INRs. Conclusion: HIV-infected individuals with suboptimal immune recovery demonstrated more activated monocytes and in particular dendritic cells, compared with patients with acceptable CD4+ gain. A high level of HIV-specific IP-10 expression in monocytes may be predictive of future CD4+ recovery.
Collapse
|
50
|
Li S, Lu LF, Li ZC, Zhang C, Zhou XY, Zhou Y, Zhang YA. Zebrafish MVP Recruits and Degrades TBK1 To Suppress IFN Production. THE JOURNAL OF IMMUNOLOGY 2018; 202:559-566. [PMID: 30530482 DOI: 10.4049/jimmunol.1801325] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/13/2018] [Indexed: 11/19/2022]
Abstract
IFN production is crucial for hosts to defend against viral infection, yet it must be tightly controlled to maintain immune homeostasis. TANK-binding kinase 1 (TBK1) is a pivotal kinase in the IFN induction signaling pathway, but it is negatively regulated by multiple molecules to avoid the excessive expression of IFN in mammals. However, the identified TBK1 suppressors and the mechanisms are rare in fish. In this study, we show that zebrafish major vault protein (MVP) recruits and degrades TBK1 in a lysosome-dependent manner to inhibit IFN production. Through viral infection, polyinosinic:polycytidylic acid and RIG-I-like receptor factor stimulation upregulated IFN expression, but overexpression of MVP significantly subverted these inductions. On the protein level, MVP interacted with TBK1, and interestingly, MVP recruited TBK1 from a uniformly distributed state in the cytoplasm to an aggregated state. Finally, MVP mediated the lysosome-dependent degradation of TBK1 and decreased the IFN response and IFN-stimulated genes expression. Our findings reveal that zebrafish MVP is a negative regulator of IFN production by restricting the activation of TBK1, supplying evidence of the balanced mechanisms of IFN expression in lower vertebrates.
Collapse
Affiliation(s)
- Shun Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, 430072 Wuhan, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 266071 Qingdao, China
| | - Long-Feng Lu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, 430072 Wuhan, China
| | - Zhuo-Cong Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, 430072 Wuhan, China.,University of Chinese Academy of Sciences, 100049 Beijing, China; and
| | - Can Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, 430072 Wuhan, China.,University of Chinese Academy of Sciences, 100049 Beijing, China; and
| | - Xiao-Yu Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, 430072 Wuhan, China.,University of Chinese Academy of Sciences, 100049 Beijing, China; and
| | - Yu Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, 430072 Wuhan, China.,University of Chinese Academy of Sciences, 100049 Beijing, China; and
| | - Yong-An Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, 430072 Wuhan, China; .,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 266071 Qingdao, China.,State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, 43007 Wuhan, China
| |
Collapse
|