1
|
Bourgeois NM, Wei L, Kaushansky A, Aitchison JD. Exploiting Host Kinases to Combat Dengue Virus Infection and Disease. Antiviral Res 2025:106172. [PMID: 40348023 DOI: 10.1016/j.antiviral.2025.106172] [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: 12/31/2024] [Revised: 04/03/2025] [Accepted: 04/23/2025] [Indexed: 05/14/2025]
Abstract
The burden of dengue on human health has dramatically increased in recent years, underscoring the urgent need for effective therapeutic interventions. Despite decades of research since the discovery of the dengue virus, no specific antiviral treatments are available and strategies to reliably prevent severe disease remain limited. Direct-acting antivirals against dengue are under active investigation but have shown limited efficacy to date. An underappreciated Achille's heal of the virus is its dependence on host factors for infection and pathogenesis, each of which presents a potential avenue for therapeutic intervention. We and others have demonstrated that dengue virus relies on multiple host kinases, some of which are already targeted by clinically approved inhibitors. These offer drug repurposing opportunities for host-directed dengue treatment. Here, we summarize findings on the role of kinases in dengue infection and disease and highlight potential kinase targets for the development of innovative host-directed therapeutics.
Collapse
Affiliation(s)
- Natasha M Bourgeois
- Department of Global Health, University of Washington, Seattle WA 98195, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle WA 98109, USA
| | - Ling Wei
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle WA 98109, USA
| | - Alexis Kaushansky
- Department of Global Health, University of Washington, Seattle WA 98195, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle WA 98109, USA.
| | - John D Aitchison
- Department of Global Health, University of Washington, Seattle WA 98195, USA; Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle WA 98109, USA.
| |
Collapse
|
2
|
Ye S, Liang Y, Chang Y, Lai B, Zhong J. Dengue Virus Replicative-Form dsRNA Is Recognized by Both RIG-I and MDA5 to Activate Innate Immunity. J Med Virol 2025; 97:e70194. [PMID: 39873327 DOI: 10.1002/jmv.70194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/30/2025]
Abstract
RIG-I like receptors (RLRs) are a family of cytosolic RNA sensors that sense RNA virus infection to activate innate immune response. It is generally believed that different RNA viruses are recognized by either RIG-I or MDA5, two important RLR members, depending on the nature of pathogen-associated molecular patterns (PAMPs) that are generated by RNA virus replication. Dengue virus (DENV) is an important RNA virus causing serious human diseases. Despite extensive investigations, the molecular basis of the DENV PAMP recognized by the host RLR has been poorly defined. Here, we demonstrated that the DENV infection-induced interferon response is dependent upon both RIG-I and MDA5, with RIG-I playing a predominant role. Next we purified the DENV PAMP RNA from the DENV-infected cells, and demonstrated that the purified DENV PAMP is viral full-length double-stranded RNA bearing 5'ppp modifications, likely representing the viral replicative-form RNA. Finally, we confirmed the nature of the DENV PAMP by reconstituting the viral replicative-form RNA from in vitro synthesized DENV genomic RNA. In conclusion, our work not only defined the molecular basis of the RLR-PAMP interaction during DENV infection, but also revealed the previously underappreciated recognition of a distinct moiety of the same PAMP by different RLRs in innate immunity against RNA viruses.
Collapse
Affiliation(s)
- Sichao Ye
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yisha Liang
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yu Chang
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Bailiang Lai
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Jin Zhong
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
3
|
Persaud AT, Khela J, Fernandes C, Chaphekar D, Burnie J, Tang VA, Colpitts CC, Guzzo C. Virion-incorporated CD14 enables HIV-1 to bind LPS and initiate TLR4 signaling in immune cells. J Virol 2024; 98:e0036324. [PMID: 38661384 PMCID: PMC11092368 DOI: 10.1128/jvi.00363-24] [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: 02/22/2024] [Accepted: 04/01/2024] [Indexed: 04/26/2024] Open
Abstract
HIV-1 has a broad range of nuanced interactions with the immune system, and the incorporation of cellular proteins by nascent virions continues to redefine our understanding of the virus-host relationship. Proteins located at the sites of viral egress can be selectively incorporated into the HIV-1 envelope, imparting new functions and phenotypes onto virions, and impacting viral spread and disease. Using virion capture assays and western blot, we show that HIV-1 can incorporate the myeloid antigen CD14 into its viral envelope. Virion-incorporated CD14 remained biologically active and able to bind its natural ligand, bacterial lipopolysaccharide (LPS), as demonstrated by flow virometry and immunoprecipitation assays. Using a Toll-like receptor 4 (TLR4) reporter cell line, we also demonstrated that virions with bound LPS can trigger TLR4 signaling to activate transcription factors that regulate inflammatory gene expression. Complementary assays with THP-1 monocytes demonstrated enhanced secretion of inflammatory cytokines like tumor necrosis factor alpha (TNF-α) and the C-C chemokine ligand 5 (CCL5), when exposed to LPS-loaded virus. These data highlight a new type of interplay between HIV-1 and the myeloid cell compartment, a previously well-established cellular contributor to HIV-1 pathogenesis and inflammation. Persistent gut inflammation is a hallmark of chronic HIV-1 infection, and contributing to this effect is the translocation of microbes across the gut epithelium. Our data herein provide proof of principle that virion-incorporated CD14 could be a novel mechanism through which HIV-1 can drive chronic inflammation, facilitated by HIV-1 particles binding bacterial LPS and initiating inflammatory signaling in TLR4-expressing cells.IMPORTANCEHIV-1 establishes a lifelong infection accompanied by numerous immunological changes. Inflammation of the gut epithelia, exacerbated by the loss of mucosal T cells and cytokine dysregulation, persists during HIV-1 infection. Feeding back into this loop of inflammation is the translocation of intestinal microbes across the gut epithelia, resulting in the systemic dissemination of bacterial antigens, like lipopolysaccharide (LPS). Our group previously demonstrated that the LPS receptor, CD14, can be readily incorporated by HIV-1 particles, supporting previous clinical observations of viruses derived from patient plasma. We now show that CD14 can be incorporated by several primary HIV-1 isolates and that this virion-incorporated CD14 can remain functional, enabling HIV-1 to bind to LPS. This subsequently allowed CD14+ virions to transfer LPS to monocytic cells, eliciting pro-inflammatory signaling and cytokine secretion. We posit here that virion-incorporated CD14 is a potential contributor to the dysregulated immune responses present in the setting of HIV-1 infection.
Collapse
Affiliation(s)
- Arvin T. Persaud
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jasmin Khela
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Claire Fernandes
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Deepa Chaphekar
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan Burnie
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Vera A. Tang
- Flow Cytometry and Virometry Core Facility, Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Che C. Colpitts
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Christina Guzzo
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Chen Z, Liu H, Zhu J, Duan X, Wang H, Li X, Zhou X, Zhao A, Yang S. Porcine promyelocytic leukemia protein isoforms suppress Japanese encephalitis virus replication in PK15 cells. Virol J 2023; 20:280. [PMID: 38031162 PMCID: PMC10687900 DOI: 10.1186/s12985-023-02212-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Promyelocytic leukemia protein (PML) is a primary component of PML nuclear bodies (PML-NBs). PML and PML-NBs play critical roles in processes like the cell cycle, DNA damage repair, apoptosis, and the antiviral immune response. Previously, we identified five porcine PML alternative splicing variants and observed an increase in the expression of these PML isoforms following Japanese encephalitis virus (JEV) infection. In this study, we examined the functional roles of these PML isoforms in JEV infection. METHODS PML isoforms were either knocked down or overexpressed in PK15 cells, after which they were infected with JEV. Subsequently, we analyzed the gene expression of PML isoforms, JEV, and the interferon (IFN)-β signaling pathway using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot. Viral titers were determined through 50% tissue culture infectious dose (TCID50) assays. RESULTS Our results demonstrated that the knockdown of endogenous PML promoted JEV replication, while the overexpression of PML isoforms 1, 3, 4, and 5 (PML1, PML3, PML4, and PML5) inhibited JEV replication. Further investigation revealed that PML1, PML3, PML4, and PML5 negatively regulated the expression of genes involved in the interferon (IFN)-β signaling pathway by inhibiting IFN regulatory factor 3 (IRF3) post-JEV infection. CONCLUSIONS These findings demonstrate that porcine PML isoforms PML1, PML3, PML4, and PML5 negatively regulate IFN-β and suppress viral replication during JEV infection. The results of this study provide insight into the functional roles of porcine PML isoforms in JEV infection and the regulation of the innate immune response.
Collapse
Affiliation(s)
- Zhenyu Chen
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Huaijin Liu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Jingjing Zhu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Xing Duan
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Han Wang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Xiangchen Li
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Xiaolong Zhou
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Ayong Zhao
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China.
| | - Songbai Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China.
| |
Collapse
|
5
|
Wu M, Pei Z, Long G, Chen H, Jia Z, Xia W. Mitochondrial antiviral signaling protein: a potential therapeutic target in renal disease. Front Immunol 2023; 14:1266461. [PMID: 37901251 PMCID: PMC10602740 DOI: 10.3389/fimmu.2023.1266461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/26/2023] [Indexed: 10/31/2023] Open
Abstract
Mitochondrial antiviral signaling protein (MAVS) is a key innate immune adaptor on the outer mitochondrial membrane that acts as a switch in the immune signal transduction response to viral infections. Some studies have reported that MAVS mediates NF-κB and type I interferon signaling during viral infection and is also required for optimal NLRP3 inflammasome activity. Recent studies have reported that MAVS is involved in various cancers, systemic lupus erythematosus, kidney diseases, and cardiovascular diseases. Herein, we summarize the structure, activation, pathophysiological roles, and MAVS-based therapies for renal diseases. This review provides novel insights into MAVS's role and therapeutic potential in the pathogenesis of renal diseases.
Collapse
Affiliation(s)
- Meng Wu
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiyin Pei
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Guangfeng Long
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Hongbing Chen
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Weiwei Xia
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
6
|
Bhattacharjee A, Naga R, Saha M, Karmakar S, Pal A, Roy S. Viral inhibitory potential of hyoscyamine in Japanese encephalitis virus-infected embryonated chicken eggs involving multiple signaling pathways. Arch Virol 2023; 168:264. [PMID: 37787913 DOI: 10.1007/s00705-023-05883-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/28/2023] [Indexed: 10/04/2023]
Abstract
Japanese encephalitis virus (JEV) is the leading cause of viral encephalitis worldwide. The emergence of new genotypes of the virus and a high rate of mutation make it necessary to develop alternative treatment strategies against this deadly pathogen. Although the antiviral properties of Atropa belladonna and some of its active components, such as atropine and scopolamine, have been studied, the effect of another important component, hyoscyamine, against JEV infection has not yet been investigated. In this study, we investigated the antiviral effect of hyoscyamine against JEV and its immunomodulatory activity in embryonated chicken eggs. Pretreatment with hyoscyamine sulphate resulted in a significant decrease in the viral load in both chorioallantoic membrane (CAM) and brain tissues at 48 and 96 hours postinfection. In silico studies showed stable binding and interaction between hyoscyamine and non-structural protein 5 (NS5), suggesting that this could be the basis of its antiviral effect. Embryonated eggs pretreated with hyoscyamine sulphate showed upregulation of Toll-like receptor 3 (TLR3), TLR7, TLR8, interleukin 4 (IL-4), and IL-10 as well as interferons and regulatory factors. Hyoscyamine sulphate was also found to cause significant downregulation of TLR4. The potential use of hyoscyamine for controlling JEV replication and its dissemination to the brain suggest that it may be a promising therapy option against JEV in the future.
Collapse
Affiliation(s)
- Arghyadeep Bhattacharjee
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal, India.
- Department of Microbiology, Kingston College of Science, Beruanpukuria, Malikapur, Kolkata-126, West Bengal, India.
| | - Rahul Naga
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal, India
| | - Manish Saha
- Department of Cardiology, R.G Kar Medical College and Hospital, Kolkata, West Bengal, India
| | - Srabani Karmakar
- Department of Microbiology, Kingston College of Science, Beruanpukuria, Malikapur, Kolkata-126, West Bengal, India
| | - Abhishek Pal
- Department of Microbiology, Ramkrishna Mission Vidyamandira, Belur, Howrah, West Bengal, India
| | - Souvik Roy
- Department of Biotechnology, St. Xavier's College, Kolkata, West Bengal, India
| |
Collapse
|
7
|
Guo Y, Pan L, Wang L, Wang S, Fu J, Luo W, Wang K, Li X, Huang C, Liu Y, Kang H, Zeng Q, Fu X, Huang Z, Li W, He Y, Li L, Peng T, Yang H, Li M, Xiao B, Cai M. Epstein-Barr Virus Envelope Glycoprotein gp110 Inhibits IKKi-Mediated Activation of NF-κB and Promotes the Degradation of β-Catenin. Microbiol Spectr 2023; 11:e0032623. [PMID: 37022262 PMCID: PMC10269791 DOI: 10.1128/spectrum.00326-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 03/10/2023] [Indexed: 04/07/2023] Open
Abstract
Epstein-Barr virus (EBV) infects host cells and establishes a latent infection that requires evasion of host innate immunity. A variety of EBV-encoded proteins that manipulate the innate immune system have been reported, but whether other EBV proteins participate in this process is unclear. EBV-encoded envelope glycoprotein gp110 is a late protein involved in virus entry into target cells and enhancement of infectivity. Here, we reported that gp110 inhibits RIG-I-like receptor pathway-mediated promoter activity of interferon-β (IFN-β) as well as the transcription of downstream antiviral genes to promote viral proliferation. Mechanistically, gp110 interacts with the inhibitor of NF-κB kinase (IKKi) and restrains its K63-linked polyubiquitination, leading to attenuation of IKKi-mediated activation of NF-κB and repression of the phosphorylation and nuclear translocation of p65. Additionally, gp110 interacts with an important regulator of the Wnt signaling pathway, β-catenin, and induces its K48-linked polyubiquitination degradation via the proteasome system, resulting in the suppression of β-catenin-mediated IFN-β production. Taken together, these results suggest that gp110 is a negative regulator of antiviral immunity, revealing a novel mechanism of EBV immune evasion during lytic infection. IMPORTANCE Epstein-Barr virus (EBV) is a ubiquitous pathogen that infects almost all human beings, and the persistence of EBV in the host is largely due to immune escape mediated by its encoded products. Thus, elucidation of EBV's immune escape mechanisms will provide a new direction for the design of novel antiviral strategies and vaccine development. Here, we report that EBV-encoded gp110 serves as a novel viral immune evasion factor, which inhibits RIG-I-like receptor pathway-mediated interferon-β (IFN-β) production. Furthermore, we found that gp110 targeted two key proteins, inhibitor of NF-κB kinase (IKKi) and β-catenin, which mediate antiviral activity and the production of IFN-β. gp110 inhibited K63-linked polyubiquitination of IKKi and induced β-catenin degradation via the proteasome, resulting in decreased IFN-β production. In summary, our data provide new insights into the EBV-mediated immune evasion surveillance strategy.
Collapse
Affiliation(s)
- Yingjie Guo
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
- Department of Clinical Laboratory, Fifth Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Lingxia Pan
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Liding Wang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Shuai Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Jiangqin Fu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Wenqi Luo
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Kezhen Wang
- School of Life Sciences, Anhui Medical University, Hefei, China
| | - Xiaoqing Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Chen Huang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Yintao Liu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Haoran Kang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Qiyuan Zeng
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Xiuxia Fu
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Zejin Huang
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Wanying Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Yingxin He
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Linhai Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Tao Peng
- State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
- Guangdong South China Vaccine, Guangzhou, China
| | - Haidi Yang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Institute of Hearing and Speech-Language Science, Guangzhou Xinhua University, Guangzhou, China
| | - Meili Li
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Bin Xiao
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
| | - Mingsheng Cai
- Department of Laboratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Qingyuan, China
- Department of Pathogenic Biology and Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
8
|
Bhattacharjee S, Ghosh D, Saha R, Sarkar R, Kumar S, Khokhar M, Pandey RK. Mechanism of Immune Evasion in Mosquito-Borne Diseases. Pathogens 2023; 12:635. [PMID: 37242305 PMCID: PMC10222277 DOI: 10.3390/pathogens12050635] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
In recent decades, mosquito-borne illnesses have emerged as a major health burden in many tropical regions. These diseases, such as malaria, dengue fever, chikungunya, yellow fever, Zika virus infection, Rift Valley fever, Japanese encephalitis, and West Nile virus infection, are transmitted through the bite of infected mosquitoes. These pathogens have been shown to interfere with the host's immune system through adaptive and innate immune mechanisms, as well as the human circulatory system. Crucial immune checkpoints such as antigen presentation, T cell activation, differentiation, and proinflammatory response play a vital role in the host cell's response to pathogenic infection. Furthermore, these immune evasions have the potential to stimulate the human immune system, resulting in other associated non-communicable diseases. This review aims to advance our understanding of mosquito-borne diseases and the immune evasion mechanisms by associated pathogens. Moreover, it highlights the adverse outcomes of mosquito-borne disease.
Collapse
Affiliation(s)
| | - Debanjan Ghosh
- Department of Biotechnology, Pondicherry University, Puducherry 605014, India
| | - Rounak Saha
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry 605014, India
| | - Rima Sarkar
- DBT Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Saurav Kumar
- DBT Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Manoj Khokhar
- Department of Biochemistry, AIIMS, Jodhpur 342005, India
| | - Rajan Kumar Pandey
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Solna, Sweden
| |
Collapse
|
9
|
Chan YT, Cheok YY, Cheong HC, Tang TF, Sulaiman S, Hassan J, Looi CY, Tan KK, AbuBakar S, Wong WF. Immune Recognition versus Immune Evasion Systems in Zika Virus Infection. Biomedicines 2023; 11:biomedicines11020642. [PMID: 36831177 PMCID: PMC9952926 DOI: 10.3390/biomedicines11020642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 02/22/2023] Open
Abstract
The reemergence of the Zika virus (ZIKV) infection in recent years has posed a serious threat to global health. Despite being asymptomatic or mildly symptomatic in a majority of infected individuals, ZIKV infection can result in severe manifestations including neurological complications in adults and congenital abnormalities in newborns. In a human host, ZIKV is primarily recognized by RIG-like receptors and Toll-like receptors that elicit anti-viral immunity through the secretion of type I interferon (IFN) to limit viral survival, replication, and pathogenesis. Intriguingly, ZIKV evades its host immune system through various immune evasion strategies, including suppressing the innate immune receptors and signaling pathways, mutation of viral structural and non-structural proteins, RNA modulation, or alteration of cellular pathways. Here, we present an overview of ZIKV recognition by the host immune system and the evasion strategies employed by ZIKV. Characterization of the host-viral interaction and viral disease mechanism provide a platform for the rational design of novel prophylactic and therapeutic strategies against ZIKV infection.
Collapse
Affiliation(s)
- Yee Teng Chan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Yi Ying Cheok
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Heng Choon Cheong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Ting Fang Tang
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Sofiah Sulaiman
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Jamiyah Hassan
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Chung Yeng Looi
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor’s University, 1, Jalan Taylors, Subang Jaya 47500, Malaysia
| | - Kim-Kee Tan
- Tropical Infectious Diseases Research and Education Centre (TIDREC), Higher Education Center of Excellence (HICoE), University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Sazaly AbuBakar
- Tropical Infectious Diseases Research and Education Centre (TIDREC), Higher Education Center of Excellence (HICoE), University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
- Correspondence: ; Tel.: +60-(3)-7967-6672
| |
Collapse
|
10
|
Wei KC, Wei WJ, Liao CL, Chang TH. Discrepant Activation Pattern of Inflammation and Pyroptosis Induced in Dermal Fibroblasts in Response to Dengue Virus Serotypes 1 and 2 and Nonstructural Protein 1. Microbiol Spectr 2023; 11:e0358622. [PMID: 36629424 PMCID: PMC9927091 DOI: 10.1128/spectrum.03586-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Four serotypes of dengue virus (DENV-1 to DENV-4) cause mild to severe disease in humans through infected mosquito bites. Dermal fibroblasts were found to be susceptible to DENV, and this may play a critical role in establishing the initial infection stage. However, the cellular response induced by the different DENV serotypes in dermal fibroblasts during the early stage of infection remains unclear. To determine this, normal human dermal fibroblast WS1 cells were infected with DENV-1 or DENV-2. Compared with the response elicited by DENV-1 infection, DENV-2 induced a stronger innate inflammatory response and cell death in the WS1 cells. However, DENV-1 activated a higher level of pyroptosis signaling than did DENV-2, which was associated with higher virion production. Caspase-1 inhibitor Ac-YVAD-cmk and imipramine, an antidepressant drug, reduced DENV-mediated caspase-1 and interleukin 1β (IL-β) cleavage in the pyroptosis pathway. Ac-YVAD-cmk and imipramine downregulated DENV virion production in WS1 cells. Furthermore, DENV-1 and DENV-2 NS1 proteins promoted diverse activation levels of cell death, inflammatory response, and activation of caspase-1 and IL-β in dermal fibroblasts at different time points. Collectively, these data suggest that DENV-1, DENV-2, and their nonstructural protein 1 (NS1) induce discrepant activation patterns of inflammation and pyroptosis in dermal fibroblasts. The pyroptosis caused by virus and NS1 may facilitate DENV replication in dermal fibroblasts. IMPORTANCE Skin fibroblasts are the primary cells of DENV infection through mosquito bites. Establishing a successful infection in dermal fibroblasts might be critical for dengue disease. However, the cellular response induced by DENV in dermal fibroblasts remains unclear. In this in vitro study, we found that DENV-2 and DENV-1 showed different time course patterns of virus replication and inflammation in dermal fibroblasts. We demonstrated that DENV-1 and DNEV-2 and their viral protein NS1 activate the cellular pyroptosis response to regulate virus replication in dermal fibroblasts. This finding suggests that pyroptosis activation in the DENV primary inoculation site plays a role in the establishment of a DENV infection.
Collapse
Affiliation(s)
- Kai-Che Wei
- Department of Dermatology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wan-Ju Wei
- Department of Dermatology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Tsung-Hsien Chang
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
11
|
Scrima N, Le Bars R, Nevers Q, Glon D, Chevreux G, Civas A, Blondel D, Lagaudrière-Gesbert C, Gaudin Y. Rabies virus P protein binds to TBK1 and interferes with the formation of innate immunity-related liquid condensates. Cell Rep 2023; 42:111949. [PMID: 36640307 DOI: 10.1016/j.celrep.2022.111949] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 07/27/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023] Open
Abstract
Viruses must overcome the interferon-mediated antiviral response to replicate and propagate into their host. Rabies virus (RABV) phosphoprotein P is known to inhibit interferon induction. Here, using a global mass spectrometry approach, we show that RABV P binds to TBK1, a kinase located at the crossroads of many interferon induction pathways, resulting in innate immunity inhibition. Mutations of TBK1 phosphorylation sites abolish P binding. Importantly, we demonstrate that upon RABV infection or detection of dsRNA by innate immunity sensors, TBK1 and its adaptor proteins NAP1 and SINTBAD form dynamic cytoplasmic condensates that have liquid properties. These condensates can form larger aggregates having ring-like structures in which NAP1 and TBK1 exhibit locally restricted movement. P binding to TBK1 interferes with the formation of these structures. This work demonstrates that proteins of the signaling pathway leading to interferon induction transiently form liquid organelles that can be targeted by viruses.
Collapse
Affiliation(s)
- Nathalie Scrima
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France
| | - Romain Le Bars
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France
| | - Quentin Nevers
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France
| | - Damien Glon
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France
| | | | - Ahmet Civas
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France
| | - Danielle Blondel
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France
| | - Cécile Lagaudrière-Gesbert
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France.
| | - Yves Gaudin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France.
| |
Collapse
|
12
|
Luria-Pérez R, Sánchez-Vargas LA, Muñoz-López P, Mellado-Sánchez G. Mucosal Vaccination: A Promising Alternative Against Flaviviruses. Front Cell Infect Microbiol 2022; 12:887729. [PMID: 35782117 PMCID: PMC9241634 DOI: 10.3389/fcimb.2022.887729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
The Flaviviridae are a family of positive-sense, single-stranded RNA enveloped viruses, and their members belong to a single genus, Flavivirus. Flaviviruses are found in mosquitoes and ticks; they are etiological agents of: dengue fever, Japanese encephalitis, West Nile virus infection, Zika virus infection, tick-borne encephalitis, and yellow fever, among others. Only a few flavivirus vaccines have been licensed for use in humans: yellow fever, dengue fever, Japanese encephalitis, tick-borne encephalitis, and Kyasanur forest disease. However, improvement is necessary in vaccination strategies and in understanding of the immunological mechanisms involved either in the infection or after vaccination. This is especially important in dengue, due to the immunological complexity of its four serotypes, cross-reactive responses, antibody-dependent enhancement, and immunological interference. In this context, mucosal vaccines represent a promising alternative against flaviviruses. Mucosal vaccination has several advantages, as inducing long-term protective immunity in both mucosal and parenteral tissues. It constitutes a friendly route of antigen administration because it is needle-free and allows for a variety of antigen delivery systems. This has promoted the development of several ways to stimulate immunity through the direct administration of antigens (e.g., inactivated virus, attenuated virus, subunits, and DNA), non-replicating vectors (e.g., nanoparticles, liposomes, bacterial ghosts, and defective-replication viral vectors), and replicating vectors (e.g., Salmonella enterica, Lactococcus lactis, Saccharomyces cerevisiae, and viral vectors). Because of these characteristics, mucosal vaccination has been explored for immunoprophylaxis against pathogens that enter the host through mucosae or parenteral areas. It is suitable against flaviviruses because this type of immunization can stimulate the parenteral responses required after bites from flavivirus-infected insects. This review focuses on the advantages of mucosal vaccine candidates against the most relevant flaviviruses in either humans or animals, providing supporting data on the feasibility of this administration route for future clinical trials.
Collapse
Affiliation(s)
- Rosendo Luria-Pérez
- Hospital Infantil de México Federico Gómez, Unidad de Investigación en Enfermedades Hemato-Oncológicas, Ciudad de México, Mexico
| | - Luis A. Sánchez-Vargas
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States
| | - Paola Muñoz-López
- Hospital Infantil de México Federico Gómez, Unidad de Investigación en Enfermedades Hemato-Oncológicas, Ciudad de México, Mexico
- Posgrado en Biomedicina y Biotecnología Molecular, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Gabriela Mellado-Sánchez
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
- Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Ciudad de México, Mexico
| |
Collapse
|
13
|
Zhu H, Zhang R, Yi L, Tang YD, Zheng C. UNC93B1 attenuates the cGAS-STING signaling pathway by targeting STING for autophagy-lysosome degradation. J Med Virol 2022; 94:4490-4501. [PMID: 35577759 DOI: 10.1002/jmv.27860] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 11/07/2022]
Abstract
STING (stimulator of interferon genes) is a pivotal innate immune adaptor, and its functions during DNA virus infections have been extensively documented. However, its homeostatic regulation is not well understood. Our study demonstrates that UNC93B1 is a crucial checker for STING to prevent hyperactivation. Ectopic expression of UNC93B1 attenuates IFN-β promoter activity and the transcriptions of IFN-β, ISG54, and ISG56 genes. Moreover, UNC93B1 also blocks the IRF3 nuclear translocation induced by ectopic expression of both cGAS and STING and reduces the stability of STING by facilitating its autophagy-lysosome degradation, which can be reversed by lysosome inhibitors. Mechanistically, UNC93B1 interacts with STING and suppresses STING-activated downstream signaling by delivering STING to the lysosomes for degradation depending on its trafficking capability. UNC93B1 knockout (KO) in human embryonic kidney 293T (HEK293T) cells facilitates IFN-β promoter activity, IFN-β, ISG54, and ISG56 transcriptions IRF3 nuclear translocation induced by ectopic expression of cGAS and STING. Infected with herpes simplex virus-1 (HSV-1), UNC93B1 knockdown BJ cells or primary peritoneal macrophages from Unc93b1-deficient (Unc93b1-/- ) mice show enhanced IFN-β, ISG54, and ISG56 transcriptions, TBK1 phosphorylation, and reduced STING degradation and viral replication. In addition, Unc93b1-/- mice exhibit higher IFN-β, ISG54, and ISG56 transcriptions and lower mortality upon HSV-1 infection in vivo. Collectively, these findings demonstrate that UNC93B1 attenuates the cGAS-STING signaling pathway by targeting STING for autophagy-lysosome degradation and provide novel insights into the function of UNC93B1 in antiviral innate immunity. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Huifang Zhu
- Neonatal/Pediatric Intensive Care Unit, Children's Medical Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Rongzhao Zhang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Li Yi
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yan-Dong Tang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
14
|
Rocha RF, Del Sarto JL, Gomes GF, Gonçalves MP, Rachid MA, Smetana JHC, Souza DG, Teixeira MM, Marques RE. Type I interferons are essential while type II interferon is dispensable for protection against St. Louis encephalitis virus infection in the mouse brain. Virulence 2021; 12:244-259. [PMID: 33410731 PMCID: PMC7808420 DOI: 10.1080/21505594.2020.1869392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/09/2020] [Accepted: 12/22/2020] [Indexed: 01/24/2023] Open
Abstract
St. Louis encephalitis virus (SLEV) is a neglected mosquito-borne flavivirus that causes severe neurological disease in humans. SLEV replication in the central nervous system (CNS) induces the local production of interferons (IFNs), which are attributed to host protection. The antiviral response to SLEV infection in the CNS is not completely understood, which led us to characterize the roles of IFNs using mouse models of St. Louis encephalitis. We infected mice deficient in type I IFN receptor (ABR-/-) or deficient in Type II IFN (IFNγ-/-) and assessed the contribution of each pathway to disease development. We found that type I and II IFNs play different roles in SLEV infection. Deficiency in type I IFN signaling was associated to an early and increased mortality, uncontrolled SLEV replication and impaired ISG expression, leading to increased proinflammatory cytokine production and brain pathology. Conversely, IFNγ-/- mice were moderately resistant to SLEV infection. IFNγ deficiency caused no changes to viral load or SLEV-induced encephalitis and did not change the expression of ISGs in the brain. We found that type I IFN is essential for the control of SLEV replication whereas type II IFN was not associated with protection in this model.
Collapse
Affiliation(s)
- Rebeca Froes Rocha
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
- Graduate Program in Genetics and Molecular Biology, State University of Campinas (UNICAMP), Campinas, Brazil
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Juliana L. Del Sarto
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Giovanni F. Gomes
- Laboratório de Neurofarmacologia, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Mariana P. Gonçalves
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
- Graduate Program in Genetics and Molecular Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Milene A. Rachid
- Laboratório de Apoptose, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Juliana H. C. Smetana
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Daniele G. Souza
- Laboratório de Interação Microrganismo-Hospedeiro, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Mauro Martins Teixeira
- Immunopharmacology Laboratory, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| |
Collapse
|
15
|
Harrison JJ, Hobson-Peters J, Bielefeldt-Ohmann H, Hall RA. Chimeric Vaccines Based on Novel Insect-Specific Flaviviruses. Vaccines (Basel) 2021; 9:1230. [PMID: 34835160 PMCID: PMC8623431 DOI: 10.3390/vaccines9111230] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/30/2022] Open
Abstract
Vector-borne flaviviruses are responsible for nearly half a billion human infections worldwide each year, resulting in millions of cases of debilitating and severe diseases and approximately 115,000 deaths. While approved vaccines are available for some of these viruses, the ongoing efficacy, safety and supply of these vaccines are still a significant problem. New technologies that address these issues and ideally allow for the safe and economical manufacture of vaccines in resource-poor countries where flavivirus vaccines are in most demand are urgently required. Preferably a new vaccine platform would be broadly applicable to all flavivirus diseases and provide new candidate vaccines for those diseases not yet covered, as well as the flexibility to rapidly pivot to respond to newly emerged flavivirus diseases. Here, we review studies conducted on novel chimeric vaccines derived from insect-specific flaviviruses that provide a potentially safe and simple system to produce highly effective vaccines against a broad spectrum of flavivirus diseases.
Collapse
Affiliation(s)
- Jessica J. Harrison
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; (J.H.-P.); (H.B.-O.); (R.A.H.)
| | - Jody Hobson-Peters
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; (J.H.-P.); (H.B.-O.); (R.A.H.)
| | - Helle Bielefeldt-Ohmann
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; (J.H.-P.); (H.B.-O.); (R.A.H.)
- School of Veterinary Science, University of Queensland, Gatton, QLD 4343, Australia
| | - Roy A. Hall
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Brisbane, QLD 4072, Australia; (J.H.-P.); (H.B.-O.); (R.A.H.)
| |
Collapse
|
16
|
Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Recent Insights Into the Molecular Mechanism of Toll-Like Receptor Response to Dengue Virus Infection. Front Microbiol 2021; 12:744233. [PMID: 34603272 PMCID: PMC8483762 DOI: 10.3389/fmicb.2021.744233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 08/23/2021] [Indexed: 12/15/2022] Open
Abstract
Dengue is the most prevalent and rapidly spreading mosquito-borne viral disease caused by dengue virus (DENV). Recently, DENV has been affecting humans within an expanding geographic range due to the warming of the earth. Innate immune responses play a significant role in antiviral defense, and Toll-like receptors (TLRs) are key regulators of innate immunity. Therefore, a detailed understanding of TLR and DENV interactions is important for devising therapeutic and preventive strategies. Several studies have indicated the ability of DENV to modulate the TLR signaling pathway and host immune response. Vaccination is considered one of the most successful medical interventions for preventing viral infections. However, only a partially protective dengue vaccine, the first licensed dengue vaccine CYD-TDV, is available in some dengue-endemic countries to protect against DENV infection. Therefore, the development of a fully protective, durable, and safe DENV vaccine is a priority for global health. Here, we demonstrate the progress made in our understanding of the host response to DENV infection, with a particular focus on TLR response and how DENV avoids the response toward establishing infection. We also discuss dengue vaccine candidates in late-stage development and the issues that must be overcome to enable their success.
Collapse
Affiliation(s)
- Mohammad Enamul Hoque Kayesh
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
- Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal, Bangladesh
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kyoko Tsukiyama-Kohara
- Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
17
|
Zheng Z, Li Y, Zhang M, Liu Y, Fu M, Gong S, Hu Q. Human Norovirus NTPase Antagonizes Interferon-β Production by Interacting With IkB Kinase ε. Front Microbiol 2021; 12:687933. [PMID: 34335514 PMCID: PMC8319745 DOI: 10.3389/fmicb.2021.687933] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/17/2021] [Indexed: 12/29/2022] Open
Abstract
Human norovirus (HuNoV) is the leading cause of epidemic acute gastroenteritis worldwide. Type I interferons (IFN)-α/β are highly potent cytokines that are initially identified for their essential roles in antiviral defense. It was reported that HuNoV infection did not induce IFN-β expression but was controlled in the presence of IFN-β in human intestinal enteroids and a gnotobiotic pig model, suggesting that HuNoV has likely developed evasion countermeasures. In this study, we found that a cDNA clone of GII.4 HuNoV, the predominantly circulating genotype worldwide, inhibits the production of IFN-β and identified the viral NTPase as a key component responsible for such inhibition. HuNoV NTPase not only inhibits the activity of IFN-β promoter but also the mRNA and protein production of IFN-β. Additional studies indicate that NTPase inhibits the phosphorylation and nuclear translocation of interferon-regulatory factor-3 (IRF-3), leading to the suppression of IFN-β promoter activation. Mechanistically, NTPase interacts with IkB kinase ε (IKKε), an important factor for IRF-3 phosphorylation, and such interaction blocks the association of IKKε with unanchored K48-linked polyubiquitin chains, resulting in the inhibition of IKKε phosphorylation. Further studies demonstrated that the 1-179 aa domain of NTPase which interacts with IKKε is critical for the suppression of IFN-β production. Our findings highlight the role of HuNoV NTPase in the inhibition of IFN-β production, providing insights into a novel mechanism underlying how HuNoV evades the host innate immunity.
Collapse
Affiliation(s)
- Zifeng Zheng
- The Joint Laboratory of Translational Precision Medicine, Guangzhou Women and Children's Medical Center, Guangzhou, China.,The Joint Laboratory of Translational Precision Medicine, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yuncheng Li
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Mudan Zhang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yalan Liu
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ming Fu
- The Joint Laboratory of Translational Precision Medicine, Guangzhou Women and Children's Medical Center, Guangzhou, China.,The Joint Laboratory of Translational Precision Medicine, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Sitang Gong
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qinxue Hu
- State Key Laboratory of Virology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,Institute for Infection and Immunity, St George's, University of London, London, United Kingdom
| |
Collapse
|
18
|
In Vitro Characterization of the Innate Immune Pathways Engaged by Live and Inactivated Tick-Borne Encephalitis Virus. Vaccines (Basel) 2021; 9:vaccines9060664. [PMID: 34204532 PMCID: PMC8234070 DOI: 10.3390/vaccines9060664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 12/28/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV) infection can lead to inflammation of the central nervous system. The disease can be effectively prevented by whole inactivated virus vaccines. Here, we investigated the innate immune profile induced in vitro by the antigen component of the vaccines, inactivated TBEV (I-TBEV), to gain insights into the mechanism of action of the TBE vaccine as compared to the live virus. To this end, we exposed human peripheral blood mononuclear cells (PBMCs) to inactivated and live TBEV and assessed cellular responses by RNA sequencing. Both inactivated and live TBEV significantly induced an interferon-dominated gene signature and an increased RIG-I-like receptor (RLR) expression. Using pathway-specific inhibitors, we assessed the involvement of pattern recognition receptors in the sensing of inactivated or live TBEV. Only RLR pathway inhibition significantly suppressed the downstream cascade induced by I-TBEV, while responses to the replicating virus were impacted by the inhibition of RIG-I-like, as well as Toll-like, receptors. Our results show that inactivated and live TBEV predominantly engaged an interferon response in our in vitro PBMC platform, and indicate RLRs as the main pattern recognition receptors involved in I-TBEV sensing.
Collapse
|
19
|
Zhang W, Zeng M, Jiang B, Lu T, Guo J, Hu T, Wang M, Jia R, Zhu D, Liu M, Zhao X, Yang Q, Wu Y, Zhang S, Ou X, Liu Y, Zhang L, Yu Y, Pan L, Cheng A, Chen S. Amelioration of Beta Interferon Inhibition by NS4B Contributes to Attenuating Tembusu Virus Virulence in Ducks. Front Immunol 2021; 12:671471. [PMID: 34079553 PMCID: PMC8165282 DOI: 10.3389/fimmu.2021.671471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/04/2021] [Indexed: 11/13/2022] Open
Abstract
Our previous studies reported that duck Tembusu virus nonstructural protein 2A (NS2A) is a major inhibitor of the IFNβ signaling pathway through competitively binding to STING with TBK1, leading to a reduction in TBK1 phosphorylation. Duck TMUV NS2B3 could cleave and bind STING to subvert the IFNβ signaling pathway. Here, we found that overexpression of duck TMUV NS4B could compete with TBK1 in binding to STING, reducing TBK1 phosphorylation and inhibiting the IFNβ signaling pathway by using the Dual-Glo® Luciferase Assay System and the NanoBiT protein-protein interaction (PPI) assay. We further identified the E2, M3, G4, W5, K10 and D34 residues in NS4B that were important for its interaction with STING and its inhibition of IFNβ induction, which were subsequently introduced into a duck TMUV replicon and an infectious cDNA clone. We found that the NS4B M3A mutant enhanced RNA replication and exhibited significantly higher titer levels than WT at 48-72 hpi but significantly decreased mortality (80%) in duck embryos compared to WT (100%); the NS4B G4A and R36A mutants slightly reduced RNA replication but exhibited the same titer levels as WT. However, the NS4B R36A mutant did not attenuate the virulence in duck embryos, whereas the G4A mutant significantly decreased the mortality (70%) of duck embryos. In addition, the NS4B W5A mutant did not affect viral replication, whereas the D34A mutant slightly reduced RNA replication, and both mutants exhibited significantly lower titer levels than the WT and significantly decreased mortality (90% and 70%, respectively) in duck embryos. Hence, our findings provide new insight into the development of attenuated flaviviruses by targeting the disabling viral strategies used to evade the innate defense mechanisms.
Collapse
Affiliation(s)
- Wei Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Miao Zeng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Bowen Jiang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Tong Lu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Jiaqi Guo
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Tao Hu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Mingshu Wang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Renyong Jia
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Dekang Zhu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Mafeng Liu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Xinxin Zhao
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Qiao Yang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Ying Wu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Shaqiu Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Xumin Ou
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Yunya Liu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Ling Zhang
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Yanling Yu
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Leichang Pan
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Anchun Cheng
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| | - Shun Chen
- Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
| |
Collapse
|
20
|
Rasi Bonab F, Baghbanzadeh A, Ghaseminia M, Bolandi N, Mokhtarzadeh A, Amini M, Dadashzadeh K, Hajiasgharzadeh K, Baradaran B, Bannazadeh Baghi H. Molecular pathways in the development of HPV-induced cervical cancer. EXCLI JOURNAL 2021; 20:320-337. [PMID: 33746665 PMCID: PMC7975633 DOI: 10.17179/excli2021-3365] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022]
Abstract
Recently, human papillomavirus (HPV) has gained considerable attention in cervical cancer research studies. It is one of the most important sexually transmitted diseases that can affect 160 to 289 out of 10000 persons every year. Due to the infectious nature of this virus, HPV can be considered a serious threat. The knowledge of viral structure, especially for viral oncoproteins like E6, E7, and their role in causing cancer is very important. This virus has different paths (PI3K/Akt, Wnt/β-catenin, ERK/MAPK, and JAK/STAT) that are involved in the transmission of signaling paths through active molecules like MEK (pMEK), ERK (pERK), and Akt (pAkt). It's eventually through these paths that cancer is developed. Precise knowledge of these paths and their signals give us the prognosis to adopt appropriate goals for prevention and control of these series of cancer.
Collapse
Affiliation(s)
- Farnaz Rasi Bonab
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Laboratory Sciences, Marand Branch, Islamic Azad University, Marand, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Moslem Ghaseminia
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nadia Bolandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kianoosh Dadashzadeh
- Department of Laboratory Sciences, Marand Branch, Islamic Azad University, Marand, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Kim H, Kim AY, Choi J, Park SY, Park SH, Kim JS, Lee SI, Park JH, Park CK, Ko YJ. Foot-and-Mouth Disease Virus Evades Innate Immune Response by 3C-Targeting of MDA5. Cells 2021; 10:271. [PMID: 33572945 PMCID: PMC7912020 DOI: 10.3390/cells10020271] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/18/2021] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
Foot-and-mouth disease (FMD) is a highly contagious disease caused by FMD virus (FMDV) in cloven-hoofed animals. Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are representative receptors in the cytoplasm for the detection of viral RNA and trigger antiviral responses, leading to the production of type I interferon. Although MDA5 is a crucial receptor for sensing picornavirus RNA, the interplay between MDA5 and FMDV is relatively unknown compared to the interplay between RIG-I and FMDV. Here, we observed that the FMDV infection inhibits MDA5 protein expression. Of the non-structural proteins, the Lb and 3C proteinases (Lbpro and 3Cpro) were identified to be primarily responsible for this inhibition. However, the inhibition by 3Cpro was independent of proteasome, lysosome and caspase-dependent pathway and was by 3C protease activity. A direct interaction between 3Cpro and MDA5 protein was observed. In conclusion, this is the first report that 3Cpro inhibits MDA5 protein expression as a mechanism to evade the innate immune response during FMDV infection. These results elucidate the pathogenesis of FMDV and provide fundamental insights for the development of a novel vaccine or therapeutic agent.
Collapse
Affiliation(s)
- Hyejin Kim
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Korea; (H.K.); (A.-Y.K.); (J.C.); (S.Y.P.); (S.H.P.); (J.-S.K.); (S.-I.L.); (J.-H.P.)
- College of Veterinary Medicine, Animal Disease Intervention Center, Kyungpook National University, Daegu 41566, Korea
| | - Ah-Young Kim
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Korea; (H.K.); (A.-Y.K.); (J.C.); (S.Y.P.); (S.H.P.); (J.-S.K.); (S.-I.L.); (J.-H.P.)
| | - Jieun Choi
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Korea; (H.K.); (A.-Y.K.); (J.C.); (S.Y.P.); (S.H.P.); (J.-S.K.); (S.-I.L.); (J.-H.P.)
| | - Sun Young Park
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Korea; (H.K.); (A.-Y.K.); (J.C.); (S.Y.P.); (S.H.P.); (J.-S.K.); (S.-I.L.); (J.-H.P.)
- College of Veterinary Medicine, Animal Disease Intervention Center, Kyungpook National University, Daegu 41566, Korea
| | - Sang Hyun Park
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Korea; (H.K.); (A.-Y.K.); (J.C.); (S.Y.P.); (S.H.P.); (J.-S.K.); (S.-I.L.); (J.-H.P.)
| | - Jae-Seok Kim
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Korea; (H.K.); (A.-Y.K.); (J.C.); (S.Y.P.); (S.H.P.); (J.-S.K.); (S.-I.L.); (J.-H.P.)
| | - Sim-In Lee
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Korea; (H.K.); (A.-Y.K.); (J.C.); (S.Y.P.); (S.H.P.); (J.-S.K.); (S.-I.L.); (J.-H.P.)
| | - Jong-Hyeon Park
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Korea; (H.K.); (A.-Y.K.); (J.C.); (S.Y.P.); (S.H.P.); (J.-S.K.); (S.-I.L.); (J.-H.P.)
| | - Choi-Kyu Park
- College of Veterinary Medicine, Animal Disease Intervention Center, Kyungpook National University, Daegu 41566, Korea
| | - Young-Joon Ko
- Animal and Plant Quarantine Agency, Gimcheon-si 39660, Korea; (H.K.); (A.-Y.K.); (J.C.); (S.Y.P.); (S.H.P.); (J.-S.K.); (S.-I.L.); (J.-H.P.)
| |
Collapse
|
22
|
Wang P, Deng Y, Guo Y, Xu Z, Li Y, Ou X, Xie L, Lu M, Zhong J, Li B, Hu L, Deng S, Peng T, Cai M, Li M. Epstein-Barr Virus Early Protein BFRF1 Suppresses IFN-β Activity by Inhibiting the Activation of IRF3. Front Immunol 2020; 11:513383. [PMID: 33391252 PMCID: PMC7774019 DOI: 10.3389/fimmu.2020.513383] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Epstein-Barr virus (EBV) is the causative agent of infectious mononucleosis that is closely associated with several human malignant diseases, while type I interferon (IFN-I) plays an important role against EBV infection. As we all know, EBV can encode some proteins to inhibit the production of IFN-I, but it’s not clear whether other proteins also take part in this progress. EBV early lytic protein BFRF1 is shown to be involved in viral maturation, however, whether BFRF1 participates in the host innate immune response is still not well known. In this study, we found BFRF1 could down-regulate sendai virus-induced IFN-β promoter activity and mRNA expression of IFN-β and ISG54 during BFRF1 plasmid transfection and EBV lytic infection, but BFRF1 could not affect the promoter activity of NF-κB or IRF7. Specifically, BFRF1 could co-localize and interact with IKKi. Although BFRF1 did not interfere the interaction between IKKi and IRF3, it could block the kinase activity of IKKi, which finally inhibited the phosphorylation, dimerization, and nuclear translocation of IRF3. Taken together, BFRF1 may play a critical role in disrupting the host innate immunity by suppressing IFN-β activity during EBV lytic cycle.
Collapse
Affiliation(s)
- Ping Wang
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Yangxi Deng
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Yingjie Guo
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Zuo Xu
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Yiwen Li
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Xiaowen Ou
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Li Xie
- Centralab, Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Manjiao Lu
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Jiayi Zhong
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Bolin Li
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Li Hu
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Shenyu Deng
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Tao Peng
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China.,South China Vaccine Corporation Limited, Guangzhou, China
| | - Mingsheng Cai
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| | - Meili Li
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
23
|
Onyango MG, Ciota AT, Kramer LD. The Vector - Host - Pathogen Interface: The Next Frontier in the Battle Against Mosquito-Borne Viral Diseases? Front Cell Infect Microbiol 2020; 10:564518. [PMID: 33178624 PMCID: PMC7596266 DOI: 10.3389/fcimb.2020.564518] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
An unprecedented spread of mosquito-borne viruses and increasing populations of mosquito vectors has led to an increase in the frequency of mosquito-borne virus disease outbreaks. Recent outbreaks of Zika virus (ZIKV) and yellow fever virus (YFV), among others have led to a concerted effort to understand the biology of mosquito-borne viruses and their interaction with their vector mosquito and vertebrate hosts. Recent studies have aimed to understand the vector-host-pathogen interface and how it influences infection, tropism and disease severity in the vertebrate host. The initial replication of the pathogen at the skin bite site is crucial in determining the progression of the infection in the vertebrate host. Delineating the role of the commensal microbes in the mosquito saliva as well as how they interact with the vertebrate host keratinocytes will improve our understanding of disease immunopathology and may lead to new therapeutics.
Collapse
Affiliation(s)
- Maria Gorreti Onyango
- New York State Department of Health, Wadsworth Center, Slingerlands, NY, United States
| | - Alexander T Ciota
- New York State Department of Health, Wadsworth Center, Slingerlands, NY, United States.,School of Public Health, State University of New York at Albany, Albany, NY, United States
| | - Laura D Kramer
- New York State Department of Health, Wadsworth Center, Slingerlands, NY, United States.,School of Public Health, State University of New York at Albany, Albany, NY, United States
| |
Collapse
|
24
|
Shi F, Li Q, Liu S, Liu F, Wang J, Cui D, Hou X, Zhou S, Zhang Y, Li H. Porcine circovirus type 2 upregulates endothelial-derived IL-8 production in porcine iliac artery endothelial cells via the RIG-I/MDA-5/MAVS/JNK signaling pathway. BMC Vet Res 2020; 16:265. [PMID: 32727484 PMCID: PMC7392700 DOI: 10.1186/s12917-020-02486-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
Background Dysfunction of endothelial cells and vascular system is one of the most important pathological changes of porcine circovirus disease (PCVD) caused by porcine circovirus type 2 (PCV2). PCV2-infected endothelial cells can upregulate the production of endothelial-derived IL-8, which can inhibit the maturation of dendritic cells. Endothelial-derived IL-8 has different structural and biological characteristics compared with monocyte-derived IL-8. However, the mechanism of endothelial-derived IL-8 production is still unclear. Results Key molecules of RIG-I-like signaling pathway RIG-I, MDA-5, MAVS and a key molecule of JNK signaling pathway c-Jun in PCV2-infected porcine iliac artery endothelial cells (PIECs) were upregulated significantly detected with quantitative PCR, Western blot and fluorescence confocal microscopy, while no significant changes were found in NF-κB signaling pathway. Meanwhile, the expression of endothelial-derived IL-8 was downregulated after RIG-I, MDA-5, or MAVS genes in PIECs were knocked down and PIECs were treated by JNK inhibitor. Conclusions PCV2 can activate RIG-I/MDA-5/MAVS/JNK signaling pathway to induce the production of endothelial-derived IL-8 in PIECs, which provides an insight into the further study of endothelial dysfunction and vascular system disorder caused by PCV2.
Collapse
Affiliation(s)
- Fengyang Shi
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, Changping District, China
| | - Qiuming Li
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, Changping District, China
| | - Shiyu Liu
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, Changping District, China
| | - Fengying Liu
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, Changping District, China
| | - Jianfang Wang
- Beijing Key Laboratory of TCVM, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, Changping District, China
| | - Defeng Cui
- Beijing Key Laboratory of TCVM, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, Changping District, China
| | - Xiaolin Hou
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, Changping District, China
| | - Shuanghai Zhou
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, Changping District, China
| | - Yonghong Zhang
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, Changping District, China.
| | - Huanrong Li
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, Changping District, China.
| |
Collapse
|
25
|
Human Type I Interferon Antiviral Effects in Respiratory and Reemerging Viral Infections. J Immunol Res 2020; 2020:1372494. [PMID: 32455136 PMCID: PMC7231083 DOI: 10.1155/2020/1372494] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/17/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
Type I interferons (IFN-I) are a group of related proteins that help regulate the activity of the immune system and play a key role in host defense against viral infections. Upon infection, the IFN-I are rapidly secreted and induce a wide range of effects that not only act upon innate immune cells but also modulate the adaptive immune system. While IFN-I and many IFN stimulated genes are well-known for their protective antiviral role, recent studies have associated them with potential pathogenic functions. In this review, we summarize the current knowledge regarding the complex effects of human IFN-I responses in respiratory as well as reemerging flavivirus infections of public health significance and the molecular mechanisms by which viral proteins antagonize the establishment of an antiviral host defense. Antiviral effects and immune modulation of IFN-stimulated genes is discussed in resisting and controlling pathogens. Understanding the mechanisms of these processes will be crucial in determining how viral replication can be effectively controlled and in developing safe and effective vaccines and novel therapeutic strategies.
Collapse
|
26
|
Wang Y, Wang X, Zhang K, Zhang X, Li S, Li Y, Fan W, Leng F, Yang M, Chen J. Extraction kinetics, thermodynamics, rheological properties and anti-BVDV activity of the hot water assisted extraction of Glycyrrhiza polysaccharide. Food Funct 2020; 11:4067-4080. [PMID: 32329761 DOI: 10.1039/d0fo00608d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The extraction kinetics and thermodynamic parameters of Glycyrrhiza polysaccharide (GP) were studied, and its rheological properties and antiviral activity were evaluated. The results showed that the extraction process could be fitted to Fick's second law of diffusion. The optimum concentration (97.62 mg mL-1) was obtained at a solid-liquid ratio of 1 : 15, (g mL-1), an extraction time of 120 min and an extraction temperature of 80 °C. The whole extraction process was spontaneous and endothermic. GP was shown to be an acid glycoprotein with a complex structure using high performance liquid chromatography (HPLC), circular dichroism (CD) and Fourier-transform infrared spectroscopy (FT-IR). A study of its rheological properties showed that GP has the characteristics of a typical non-Newtonian pseudoplastic fluid and that its viscosity could be significantly affected by temperature, pH and the presence of other ions. Branched and soft fiber structures with irregular molecular aggregation were observed by scanning electron microscopy (SEM) and atomic force microscopy (AFM). Moreover, GP showed good inhibitory activity against bovine viral diarrhea virus (BVDV) via the regulation of the relative expression levels of the IRF-1 and IRF-3 genes in MDBK cells. This activity was found to be dependent on the physicochemical and structural properties of GP. These findings imply that GP can be considered as a natural source of active material for the prevention of viral disease.
Collapse
Affiliation(s)
- Yonggang Wang
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Hingane S, Joshi N, Surjit M, Ranjith-Kumar CT. Hepatitis E Virus ORF2 Inhibits RIG-I Mediated Interferon Response. Front Microbiol 2020; 11:656. [PMID: 32351484 PMCID: PMC7174656 DOI: 10.3389/fmicb.2020.00656] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/23/2020] [Indexed: 01/18/2023] Open
Abstract
Understanding the dynamics of host innate immune responses against a pathogen marks the first step toward developing intervention strategies against the pathogen. The cytosolic pattern recognition receptor retinoic acid-inducible gene I (RIG-I) has been shown to be the major innate immune sensor for hepatitis E virus (HEV). Here, we show that HEV capsid protein (ORF2), a 660 amino acid long protein, interferes with the RIG-I signaling. Interestingly, only the full length ORF2 protein but not the 112-608 ORF2 protein inhibited RIG-I dependent interferon response. Both synthetic agonist and virus induced RIG-I activation was modulated by ORF2. Interference of interferon response was confirmed by reporter assays involving different interferon inducible promoters, qRT PCR, ELISA, and immunofluorescence microscopy. Neither glycosylation nor dimerization of the ORF2 protein had any effect on the observed inhibition. Further analyses revealed that the ORF2 protein antagonized Toll-like receptor (TLR) pathways as well. ORF2 inhibited signaling by RIG-I and TLR adapters, IPS-1, MyD88, and TRIF but was unable to inhibit activation by ectopically expressed IRF3 suggesting that it may be acting at a site upstream of IRF3 and downstream of adapter proteins. Our data uncover a new mechanism by which HEV may interfere with the host antiviral signaling.
Collapse
Affiliation(s)
- Smita Hingane
- Virology Laboratory, Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, Gurgaon, India
| | - Nishant Joshi
- Virology Laboratory, Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, Gurgaon, India
| | - Milan Surjit
- Virology Laboratory, Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, Gurgaon, India
| | - C T Ranjith-Kumar
- Virology Laboratory, Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, Gurgaon, India.,University School of Biotechnology, Guru Gobind Singh Indraprastha University, Dwarka, India
| |
Collapse
|
28
|
Wei KC, Wei WJ, Liu YS, Yen LC, Chang TH. Assessment of Prolonged Dengue Virus Infection in Dermal Fibroblasts and Hair-Follicle Dermal Papilla Cells. Viruses 2020; 12:v12030267. [PMID: 32121148 PMCID: PMC7150742 DOI: 10.3390/v12030267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/19/2020] [Accepted: 02/26/2020] [Indexed: 11/16/2022] Open
Abstract
Dengue virus (DENV)-mediated hair loss is one of the post-dengue fatigue syndromes and its pathophysiology remains unknown. Whether long-term or persistent infection with DENV in the scalp results in hair loss is unclear. In this study, we cultured human dermal fibroblasts (WS1 cells) and primary human hair-follicle dermal papilla cells (HFDPCs) in the long term with DENV-2 infection. The production of virion, the expression of inflammatory and anti-virus genes, and their signaling transduction activity in the infected cells were analyzed. DENV-2 NS3 protein and DENV-2 5′ UTR RNA were detected in fibroblasts and HFDPCs that were subjected to long-term infection with DENV-2 for 33 days. A significant amount of DENV-2 virion was produced by both WS1 cells and HFDPCs in the first two days of acute infection. The virion was also detected in WS1 cells that were infected in the long term, but HFDPCs failed to produce DENV-2 after long-term culture. Type I and type III interferons, and inflammatory cytokines were highly expressed in the acute phase of DENV infection in HFPDC and WS1 cells. However, in the long-term cultured cells, modest levels of anti-viral protein genes were expressed and we observed reduced signaling activity, which was correlated with the level of virus production changes. Long-term infection of DENV-2 downregulated the expression of hair growth regulatory factors, such as Rip1, Wnt1, and Wnt4. This in vitro study shows that the long-term infection with DENV-2 in dermal fibroblasts and dermal papilla cells may be involved with the prolonged-DENV-infection-mediated hair loss of post-dengue fatigue syndrome. However, direct evidence for viral replication in the human hair of a dengue victim or animal infection model is required.
Collapse
Affiliation(s)
- Kai-Che Wei
- Department of Dermatology, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan; (K.-C.W.); (W.-J.W.)
- Faculty of Yuh-Ing Junior College of Health Care and Management, Kaohsiung 80776, Taiwan
- National Yang Ming University, Taipei 11211, Taiwan
| | - Wan-Ju Wei
- Department of Dermatology, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan; (K.-C.W.); (W.-J.W.)
| | - Yi-Shan Liu
- Department of Dermatology, E-Da Hospital, I-Shou University, Kaohsiung 84001, Taiwan;
- Graduate Institute of Science Education and Environmental Education, National Kaohsiung Normal University, Kaohsiung 82446, Taiwan
| | - Li-Chen Yen
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Tsung-Hsien Chang
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 11490, Taiwan;
- Correspondence:
| |
Collapse
|
29
|
You H, Lin Y, Lin F, Yang M, Li J, Zhang R, Huang Z, Shen Q, Tang R, Zheng C. β-Catenin Is Required for the cGAS/STING Signaling Pathway but Antagonized by the Herpes Simplex Virus 1 US3 Protein. J Virol 2020; 94:e01847-19. [PMID: 31801859 PMCID: PMC7022340 DOI: 10.1128/jvi.01847-19] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022] Open
Abstract
The cGAS/STING-mediated DNA-sensing signaling pathway is crucial for interferon (IFN) production and host antiviral responses. Herpes simplex virus I (HSV-1) is a DNA virus that has evolved multiple strategies to evade host immune responses. Here, we demonstrate that the highly conserved β-catenin protein in the Wnt signaling pathway is an important factor to enhance the transcription of type I interferon (IFN-I) in the cGAS/STING signaling pathway, and the production of IFN-I mediated by β-catenin was antagonized by HSV-1 US3 protein via its kinase activity. Infection by US3-deficienct HSV-1 and its kinase-dead variants failed to downregulate IFN-I and IFN-stimulated gene (ISG) production induced by β-catenin. Consistent with this, absence of β-catenin enhanced the replication of US3-deficienct HSV-1, but not wild-type HSV-1. The underlying mechanism was the interaction of US3 with β-catenin and its hyperphosphorylation of β-catenin at Thr556 to block its nuclear translocation. For the first time, HSV-1 US3 has been shown to inhibit IFN-I production through hyperphosphorylation of β-catenin and to subvert host antiviral innate immunity.IMPORTANCE Although increasing evidence has demonstrated that HSV-1 subverts host immune responses and establishes lifelong latent infection, the molecular mechanisms by which HSV-1 interrupts antiviral innate immunity, especially the cGAS/STING-mediated cellular DNA-sensing signaling pathway, have not been fully explored. Here, we show that β-catenin promotes cGAS/STING-mediated activation of the IFN pathway, which is important for cellular innate immune responses and intrinsic resistance to DNA virus infection. The protein kinase US3 antagonizes the production of IFN by targeting β-catenin via its kinase activity. The findings in this study reveal a novel mechanism for HSV-1 to evade host antiviral immunity and add new knowledge to help in understanding the interaction between the host and HSV-1 infection.
Collapse
Affiliation(s)
- Hongjuan You
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yingying Lin
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Feng Lin
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Mingyue Yang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Jiahui Li
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Rongzhao Zhang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Zhiming Huang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Qingtang Shen
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
30
|
Xi S, Liu K, Xiao C, Hameed M, Ou A, Shao D, Li B, Wei J, Qiu Y, Miao D, Ma Z. Establishment and characterization of the pig tonsil epithelial (PT) cell line as a new model for persist infection of Japanese Encephalitis Virus. Vet Microbiol 2020; 242:108587. [PMID: 32122591 DOI: 10.1016/j.vetmic.2020.108587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 11/17/2022]
Abstract
Japanese encephalitis virus (JEV) causes a serious zoonotic disease worldwide, pig is the reservoir and amplifying host of JEV. JEV can persist infect tonsil in pig, but the relation between persist infection in tonsil and reservoir are not clear until now. A stable pig tonsil cell line is necessary for JEV persist infection research. In this study, we established a continuous epithelial cell line, named PT cell, from the pig tonsil. This cell is susceptible to JEV. We determined the growth characteristics, molecular properties, microstructure profiles of PT cell. JEV is easy to enter PT cell which may partly explain the reason of persist infection. We further determined that LMAN2L, a mannose lectin proteins, is the primary viral receptors for JEV entry in PT cell. IFITM3, an cellular surface antiviral factor, is underexpression in PT cell after JEV infection. All these results provide solid evidence that PT cell will promote additional research on JEV persist infection in pig tonsil.
Collapse
Affiliation(s)
- Shumin Xi
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Ke Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Changguang Xiao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Muddassar Hameed
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Anni Ou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Donghua Shao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Beibei Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Yafeng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China
| | - Denian Miao
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, PR China
| | - Zhiyong Ma
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, No. 518, Ziyue Road, Shanghai, 200241, PR China.
| |
Collapse
|
31
|
The Interplay between Dengue Virus and the Human Innate Immune System: A Game of Hide and Seek. Vaccines (Basel) 2019; 7:vaccines7040145. [PMID: 31658677 PMCID: PMC6963221 DOI: 10.3390/vaccines7040145] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022] Open
Abstract
With 40% of the world population at risk, infections with dengue virus (DENV) constitute a serious threat to public health. While there is no antiviral therapy available against this potentially lethal disease, the efficacy of the only approved vaccine is not optimal and its safety has been recently questioned. In order to develop better vaccines based on attenuated and/or chimeric viruses, one must consider how the human immune system is engaged during DENV infection. The activation of the innate immunity through the detection of viruses by cellular sensors is the first line of defence against those pathogens. This triggers a cascade of events which establishes an antiviral state at the cell level and leads to a global immunological response. However, DENV has evolved to interfere with the innate immune signalling at multiple levels, hence dampening antiviral responses and favouring viral replication and dissemination. This review elaborates on the interplay between DENV and the innate immune system. A special focus is given on the viral countermeasure mechanisms reported over the last decade which should be taken into consideration during vaccine development.
Collapse
|
32
|
Chang YT, Kung MH, Hsu TH, Hung WT, Chen YS, Yen LC, Chang TH. Aichi Virus Induces Antiviral Host Defense in Primary Murine Intestinal Epithelial Cells. Viruses 2019; 11:v11080763. [PMID: 31430947 PMCID: PMC6722774 DOI: 10.3390/v11080763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/09/2019] [Accepted: 08/15/2019] [Indexed: 12/30/2022] Open
Abstract
The picornavirus Aichi virus (AiV) is a non-enveloped RNA virus that causes acute gastroenteritis symptoms, such as diarrhea, abdominal pain, nausea, vomiting, and fever. Antiviral host defense involves the fast response of type I interferon (IFN) and the secretion of inflammatory cytokines against pathogens. However, the intestinal inflammatory and antiviral response to AiV infection is poorly understood. This study evaluated the antiviral activity of intestinal epithelial cells (IECs), which form a single-cell layer separating the bowel wall from pathogens. Isolated primary mouse IECs were subjected to AiV infection and virion production, inducing the mRNA expression of type I/type III IFNs and inflammatory cytokines. The mechanism involved induced the expression of phospho-IFN regulatory factor 3 and mitochondrial antiviral-signaling protein of type I IFN signaling. These findings were also observed in AiV-infected human colon carcinoma cells. In summary, a viral productive and pathogenic infection of AiV in primary murine IECs is validated.
Collapse
Affiliation(s)
- Yun-Te Chang
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
- Department of Physical Therapy, Shu-Zen Junior College of Medicine and Management, Kaohsiung 81362, Taiwan
| | - Ming-Hsiang Kung
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| | - Thung-Hsien Hsu
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| | - Wan-Ting Hung
- Department of Critical Care Center Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
| | - Yao-Shen Chen
- Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
- Department of Internal Medicine, National Yang-Ming University, Taipei 12221, Taiwan
| | - Li-Chen Yen
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Tsung-Hsien Chang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan.
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei 11490, Taiwan.
| |
Collapse
|
33
|
Rastogi M, Singh SK. Modulation of Type-I Interferon Response by hsa-miR-374b-5p During Japanese Encephalitis Virus Infection in Human Microglial Cells. Front Cell Infect Microbiol 2019; 9:291. [PMID: 31448245 PMCID: PMC6695837 DOI: 10.3389/fcimb.2019.00291] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/29/2019] [Indexed: 12/24/2022] Open
Abstract
Japanese Encephalitis virus (JEV) is a neurotropic ssRNA virus, belonging to the Flaviviridae family. JEV is one of the leading causes of the viral encephalitis in Southeast-Asian countries. JEV primarily infects neurons however, the microglial activation has been reported to further enhance the neuroinflammation and promote neuronal death. The PI3K/AKT pathway has been reported to play an important role in type-I interferon response via IRF3. Phosphatase and tensin homolog (PTEN), a negative regulator of PI3K/AKT pathway, participates in microglial polarization and neuroinflammation. The microRNAs are small non-coding endogenously expressed RNAs, which regulate the gene expression by binding at 3′ UTR of target gene. The human microglial cells were infected with JEV (JaOArS982 strain) and up-regulation of microRNA; hsa-miR-374b-5p was confirmed by qRT-PCR. The genes in PI3K/AKT pathway, over-expression and knock-down studies of hsa-miR-374b-5p with and without JEV infection were analyzed through immuno blotting. The regulatory role of hsa-miR-374b-5p on the expression of type-I interferon was determined by luciferase assays. JEV infection modulated the expression of hsa-miR-374b-5p and PI3K/AKT pathway via PTEN. The over-expression of hsa-miR-374b-5p suppressed the PTEN while up-regulated the AKT and IRF3 proteins, whereas, the knockdown rescued the PTEN expression and suppressed the AKT and IRF3 proteins. The modulation of hsa-miR-374b-5p regulated the type-I interferon response during JEV infection. In present study, we have shown the modulation of PTEN by hsa-miR-374b-5p, which regulated the PI3K/AKT/IRF3 axis in JEV infected microglial cells.
Collapse
Affiliation(s)
- Meghana Rastogi
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sunit K Singh
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
34
|
Esser-Nobis K, Aarreberg LD, Roby JA, Fairgrieve MR, Green R, Gale M. Comparative Analysis of African and Asian Lineage-Derived Zika Virus Strains Reveals Differences in Activation of and Sensitivity to Antiviral Innate Immunity. J Virol 2019; 93:e00640-19. [PMID: 31019057 PMCID: PMC6580957 DOI: 10.1128/jvi.00640-19] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/24/2022] Open
Abstract
In recent years, Asian lineage Zika virus (ZIKV) strains emerged to cause pandemic outbreaks associated with a high rate of congenital ZIKV syndrome (CZVS). The reasons for the enhanced spread and severe disease caused by newly emerging strains are not fully understood. Here we compared viral sequences, viral replication, and innate immune signaling induction of three different ZIKV strains derived from African and Asian lineages and West Nile virus, another flavivirus. We found pronounced differences in activation of innate immune signaling and inhibition of viral replication across ZIKV strains. The newly emerged Asian ZIKV strain Brazil Fortaleza 2015, which is associated with a higher rate of neurodevelopmental disorders like microcephaly, induced much weaker and delayed innate immune signaling in infected cells. However, superinfection studies to assess control of innate immune signaling induced by Sendai virus argue against an active block of IRF3 activation by the Brazilian strain of ZIKV and rather suggest an evasion of detection by host cell pattern recognition receptors. Compared to the Asian strain FSS13025 isolated in Cambodia, both ZIKV Uganda MR766 and ZIKV Brazil Fortaleza appear less sensitive to the interferon-induced antiviral response. ZIKV infection studies of cells lacking the different RIG-I-like receptors identified RIG-I as the major cytosolic pattern recognition receptor for detection of ZIKV.IMPORTANCE Zika Virus (ZIKV), discovered in 1947, is divided into African and Asian lineages. Pandemic outbreaks caused by currently emerging Asian lineage strains are accompanied by high rates of neurological disorders and exemplify the global health burden associated with this virus. Here we compared virological and innate immunological aspects of two ZIKV strains from the Asian lineage, an emerging Brazilian strain and a less-pathogenic Cambodian strain, and the prototypic African lineage ZIKV strain from Uganda. Compared to the replication of other ZIKV strains, the replication of ZIKV Brazil was less sensitive to the antiviral actions of interferon (IFN), while infection with this strain induced weaker and delayed innate immune responses in vitro Our data suggest that ZIKV Brazil directs a passive strategy of innate immune evasion that is reminiscent of a stealth virus. Such strain-specific properties likely contribute to differential pathogenesis and should be taken into consideration when choosing virus strains for future molecular studies.
Collapse
Affiliation(s)
- Katharina Esser-Nobis
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Lauren D Aarreberg
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Justin A Roby
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Marian R Fairgrieve
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Richard Green
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
35
|
You H, Zheng S, Huang Z, Lin Y, Shen Q, Zheng C. Herpes Simplex Virus 1 Tegument Protein UL46 Inhibits TANK-Binding Kinase 1-Mediated Signaling. mBio 2019; 10:e00919-19. [PMID: 31113902 PMCID: PMC6529639 DOI: 10.1128/mbio.00919-19] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 12/15/2022] Open
Abstract
TANK-binding kinase 1 (TBK1) is a key component of the antiviral immunity signaling pathway. It activates downstream interferon regulatory factor 3 (IRF3) and subsequent type I interferon (IFN-I) production. Herpes simplex virus type 1 (HSV-1) can antagonize host antiviral immune responses and lead to latent infection. Here, HSV-1 tegument protein UL46 was demonstrated to downregulate TBK1-dependent antiviral innate immunity. UL46 interacted with TBK1 and reduced TBK1 activation and its downstream signaling. Our results showed that UL46 impaired the interaction of TBK1 and IRF3 and downregulated the activation of IRF3 by inhibiting the dimerization of TBK1 to reduce the IFN-I production induced by TBK1 and immunostimulatory DNA. The IFN-I and its downstream antiviral genes induced by UL46-deficient HSV-1 (ΔUL46 HSV-1) were higher than those of wild-type HSV-1 (WT HSV-1). In addition, the stable knockdown of TBK1 facilitated the replication of ΔUL46 HSV-1, but not WT HSV-1. Together, these findings reveal a novel mechanism of immune evasion by HSV-1.IMPORTANCE HSV-1 has evolved multiple strategies to evade host antiviral responses and establish a lifelong latent infection, but the molecular mechanisms by which HSV-1 interrupts antiviral innate immunity are not completely understood. As TBK1 is very critical for antiviral innate immunity, it is of great interest to reveal the immune evasion mechanism of HSV-1 by targeting TBK1. In the present study, HSV-1 UL46 was found to inhibit the activation of IFN-I by targeting TBK1, suggesting that the evasion of TBK1 mediated antiviral innate immunity by HSV-1 UL46. Findings in this study will provide new insights into the host-virus interaction and help develop new approaches against HSV-1 infection.
Collapse
Affiliation(s)
- Hongjuan You
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Sisilia Zheng
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
| | - Zhiming Huang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yingying Lin
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Qingtang Shen
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
36
|
Davi C, Pastor A, Oliveira T, Neto FBDL, Braga-Neto U, Bigham AW, Bamshad M, Marques ETA, Acioli-Santos B. Severe Dengue Prognosis Using Human Genome Data and Machine Learning. IEEE Trans Biomed Eng 2019; 66:2861-2868. [PMID: 30716030 DOI: 10.1109/tbme.2019.2897285] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Dengue has become one of the most important worldwide arthropod-borne diseases. Dengue phenotypes are based on laboratorial and clinical exams, which are known to be inaccurate. OBJECTIVE We present a machine learning approach for the prediction of dengue fever severity based solely on human genome data. METHODS One hundred and two Brazilian dengue patients and controls were genotyped for 322 innate immunity single nucleotide polymorphisms (SNPs). Our model uses a support vector machine algorithm to find the optimal loci classification subset and then an artificial neural network (ANN) is used to classify patients into dengue fever or severe dengue. RESULTS The ANN trained on 13 key immune SNPs selected under dominant or recessive models produced median values of accuracy greater than 86%, and sensitivity and specificity over 98% and 51%, respectively. CONCLUSION The proposed classification method, using only genome markers, can be used to identify individuals at high risk for developing the severe dengue phenotype even in uninfected conditions. SIGNIFICANCE Our results suggest that the genetic context is a key element in phenotype definition in dengue. The methodology proposed here is extendable to other Mendelian based and genetically influenced diseases.
Collapse
|
37
|
Suppression of Type I Interferon Signaling by Flavivirus NS5. Viruses 2018; 10:v10120712. [PMID: 30558110 PMCID: PMC6316265 DOI: 10.3390/v10120712] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/08/2018] [Accepted: 12/09/2018] [Indexed: 01/02/2023] Open
Abstract
Type I interferon (IFN-I) is the first line of mammalian host defense against viral infection. To counteract this, the flaviviruses, like other viruses, have encoded a variety of antagonists, and use a multi-layered molecular defense strategy to establish their infections. Among the most potent antagonists is non-structural protein 5 (NS5), which has been shown for all disease-causing flaviviruses to target different steps and players of the type I IFN signaling pathway. Here, we summarize the type I IFN antagonist mechanisms used by flaviviruses with a focus on the role of NS5 in regulating one key regulator of type I IFN, signal transducer and activator of transcription 2 (STAT2).
Collapse
|
38
|
Kao YT, Lai MMC, Yu CY. How Dengue Virus Circumvents Innate Immunity. Front Immunol 2018; 9:2860. [PMID: 30564245 PMCID: PMC6288372 DOI: 10.3389/fimmu.2018.02860] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/20/2018] [Indexed: 12/24/2022] Open
Abstract
In the battle between a virus and its host, innate immunity serves as the first line of defense protecting the host against pathogens. The antiviral actions start with the recognition of pathogen-associated molecular patterns derived from the virus, then ultimately turning on particular transcription factors to generate antiviral interferons (IFNs) or proinflammatory cytokines via fine-tuned signaling cascades. With dengue virus (DENV) infection, its viral RNA is recognized by the host RNA sensors, mainly retinoic acid inducible gene-I (RIG-I)-like receptors (RLRs) and toll-like receptors. DENV infection also activates the cyclic GMP-AMP synthase–stimulator of interferon genes (cGAS–STING)-mediated DNA-sensing pathway despite the absence of a DNA stage in the DENV lifecycle. In the last decade, DENV has been considered a weak IFN-inducing pathogen with the evidence that DENV has evolved multiple strategies antagonizing the host IFN system. DENV passively escapes from innate immunity surveillance and also actively subverts the innate immune system at multiple steps. DENV targets both RNA-triggered RLR–mitochondrial antiviral signaling protein (RLR–MAVS) and DNA-triggered cGAS–STING signaling to reduce IFN production in infected cells. It also blocks IFN action by inhibiting IFN regulatory factor- and signal transducer and activator of transcription-mediated signaling. This review explores the current understanding of how DENV escapes the control of the innate immune system by modifying viral RNA and viral protein and by post-translational modification of cellular factors. The roles of the DNA-sensing pathway in DENV infection, and how mitochondrial dynamics participates in innate immunity are also discussed.
Collapse
Affiliation(s)
- Yu-Ting Kao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| | - Michael M C Lai
- Research Center for Emerging Viruses, China Medical University Hospital, Taichung, Taiwan.,Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chia-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| |
Collapse
|
39
|
Wei KC, Huang MS, Chang TH. Dengue Virus Infects Primary Human Hair Follicle Dermal Papilla Cells. Front Cell Infect Microbiol 2018; 8:268. [PMID: 30186771 PMCID: PMC6110916 DOI: 10.3389/fcimb.2018.00268] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022] Open
Abstract
During the epidemic of the dengue virus (DENV) infection in Taiwan in 2014 and 2015, we observed an abnormally high frequency of increased scalp hair shedding in infected individuals that could not be explained by telogen effluvium. In this study, the mechanism of hair loss caused by DENV was explored. Human hair follicle dermal papilla cells (HFDPCs) are essential for hair follicle morphogenesis and cycling. Thus, we established an in vitro DENV infection model in HFDPCs. On immunofluorescence analysis, HFDPCs that were susceptible to DENV infection responded to type I interferon (IFN) treatment, and the cells showed antibody-dependent enhancement (ADE) effect. The expression of the pro-inflammatory cytokines, interleukin 6 (IL-6), and tumor necrosis factor-alpha (TNF-α), revealed an inflammatory response in DENV-infected HFDPCs. In particular, DENV infection impaired cell viability, and it activated caspase-associated cell death signaling in HFDPCs. In conclusion, our data indicate that direct infection with DENV causes inflammation and cell death in HFDPCs, which is involved in the mechanisms of hair loss after DENV infection. The knowledge of DENV infection in an immune-privileged tissue, such as hair follicles, may suggest their use for further studies on post-dengue fatigue syndrome (PDFS).
Collapse
Affiliation(s)
- Kai-Che Wei
- Department of Dermatology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Faculty of Yuh-Ing Junior College of Health Care and Management, Kaohsiung, Taiwan
| | - Mei-Shu Huang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Tsung-Hsien Chang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, Taiwan
| |
Collapse
|
40
|
Cuartas-López AM, Hernández-Cuellar CE, Gallego-Gómez JC. Disentangling the role of PI3K/Akt, Rho GTPase and the actin cytoskeleton on dengue virus infection. Virus Res 2018; 256:153-165. [DOI: 10.1016/j.virusres.2018.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/30/2018] [Accepted: 08/14/2018] [Indexed: 12/22/2022]
|
41
|
Huang J, You H, Su C, Li Y, Chen S, Zheng C. Herpes Simplex Virus 1 Tegument Protein VP22 Abrogates cGAS/STING-Mediated Antiviral Innate Immunity. J Virol 2018; 92:e00841-18. [PMID: 29793952 PMCID: PMC6052299 DOI: 10.1128/jvi.00841-18] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 05/16/2018] [Indexed: 11/20/2022] Open
Abstract
Cytosolic DNA arising from intracellular pathogens is sensed by cyclic GMP-AMP synthase (cGAS) and triggers a powerful innate immune response. However, herpes simplex virus 1 (HSV-1), a double-stranded DNA virus, has developed multiple mechanisms to attenuate host antiviral machinery and facilitate viral infection and replication. In the present study, we found that HSV-1 tegument protein VP22 acts as an inhibitor of cGAS/stimulator of interferon genes (cGAS/STING)-mediated production of interferon (IFN) and its downstream antiviral genes. Our results showed that ectopic expression of VP22 decreased cGAS/STING-mediated IFN-β promoter activation and IFN-β production. Infection with wild-type (WT) HSV-1, but not VP22-deficient virus (ΔVP22), inhibited immunostimulatory DNA (ISD)-induced activation of the IFN signaling pathway. Further study showed that VP22 interacted with cGAS and inhibited the enzymatic activity of cGAS. In addition, stable knockdown of cGAS facilitated the replication of ΔVP22 virus but not the WT. In summary, our findings indicate that HSV-1 VP22 acts as an antagonist of IFN signaling to persistently evade host innate antiviral responses.IMPORTANCE cGAS is very important for host defense against viral infection, and many viruses have evolved ways to target cGAS and successfully evade the attack by the immune system of their susceptible host. This study demonstrated that HSV-1 tegument protein VP22 counteracts the cGAS/STING-mediated DNA-sensing antiviral innate immunity signaling pathway by inhibiting the enzymatic activity of cGAS. The findings in this study will expand our understanding of the interaction between HSV-1 replication and the host DNA-sensing signaling pathway.
Collapse
Affiliation(s)
- Jian Huang
- Soochow University, Institutes of Biology and Medical Sciences, Suzhou, China
| | - Hongjuan You
- Soochow University, Institutes of Biology and Medical Sciences, Suzhou, China
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chenhe Su
- Lady Davis Institute-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Yangxin Li
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shunhua Chen
- Microbiology and Immunology College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Minhou, Fuzhou, China
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
42
|
Chazal M, Beauclair G, Gracias S, Najburg V, Simon-Lorière E, Tangy F, Komarova AV, Jouvenet N. RIG-I Recognizes the 5' Region of Dengue and Zika Virus Genomes. Cell Rep 2018; 24:320-328. [PMID: 29996094 DOI: 10.1016/j.celrep.2018.06.047] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 05/21/2018] [Accepted: 06/08/2018] [Indexed: 12/24/2022] Open
Abstract
The flavivirus genus comprises major human pathogens, such as Dengue (DENV) and Zika (ZIKV) viruses. RIG-I and MDA5 are key cytoplasmic pathogen recognition receptors that are implicated in detecting viral RNAs. Here, we show that RNAs that co-purified with RIG-I during DENV infection are immuno-stimulatory, whereas RNAs bound to MDA5 are not. An affinity purification method combined with next-generation sequencing (NGS) revealed that the 5' region of the DENV genome is recognized by RIG-I. No DENV RNA was bound to MDA5. In vitro production of fragments of the DENV genome confirmed the NGS data and revealed that the 5' end of the genome, when bearing 5'-triphosphates, is the RIG-I ligand. The 5' region of the ZIKV genome is also a RIG-I agonist. We propose that RIG-I binds to the highly structured and conserved 5' region of flavivirus nascent transcripts before capping and that this mechanism leads to interferon secretion by infected cells.
Collapse
Affiliation(s)
- Maxime Chazal
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France; Unité de Génétique Fonctionnelle des Maladies Infectieuses, Institut Pasteur, Paris 75015, France; CNRS UMR2000 Génomique Évolutive, Modélisation et Santé, Institut Pasteur, Paris 75015, France
| | - Guillaume Beauclair
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France
| | - Ségolène Gracias
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France
| | - Valérie Najburg
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France
| | - Etienne Simon-Lorière
- Unité de Génétique Fonctionnelle des Maladies Infectieuses, Institut Pasteur, Paris 75015, France; CNRS UMR2000 Génomique Évolutive, Modélisation et Santé, Institut Pasteur, Paris 75015, France
| | - Frédéric Tangy
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France
| | - Anastassia V Komarova
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France.
| | - Nolwenn Jouvenet
- Unité Génomique Virale et Vaccination, Département de Virologie, CNRS UMR 3569, Institut Pasteur, Paris 75015, France.
| |
Collapse
|
43
|
Zhang Y, Dittmer DP, Mieczkowski PA, Host KM, Fusco WG, Duncan JA, Damania B. RIG-I Detects Kaposi's Sarcoma-Associated Herpesvirus Transcripts in a RNA Polymerase III-Independent Manner. mBio 2018; 9:e00823-18. [PMID: 29970461 PMCID: PMC6030556 DOI: 10.1128/mbio.00823-18] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 12/24/2022] Open
Abstract
Retinoic acid-inducible gene I (RIG-I) is a cytosolic pathogen recognition receptor that initiates the innate immune response against many RNA viruses. We previously showed that RIG-I restricts Kaposi's sarcoma-associated herpesvirus (KSHV) reactivation (J. A. West et al., J Virol 88:5778-5787, 2014, https://doi.org/10.1128/JVI.03226-13). In this study, we report that KSHV stimulates the RIG-I signaling pathway in a RNA polymerase (Pol) III-independent manner and subsequently induces type I interferon (IFN) responses. Knockdown or inhibition of RNA Pol III had no effect on beta interferon (IFN-β) induction by KSHV. By using high-throughput sequencing of RNA isolated by cross-linking immunoprecipitation (HITS-CLIP) approach, we identified multiple KSHV regions that give rise to RNA fragments binding to RIG-I, such as ORF810420-10496, Repeat region (LIR1)119059-119204, and ORF2543561-43650 The sequence dissimilarity between these fragments suggests that RIG-I detects a particular structure rather than a specific sequence motif. Synthesized ORF810420-10496 RNA stimulated RIG-I-dependent but RNA Pol III-independent IFN-β signaling. In summary, several KSHV RNAs are sensed by RIG-I in a RNA Pol III-independent manner.IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. Innate immune responses against viral infections, especially the induction of type I interferon, are critical for limiting the replication of viruses. Retinoic acid-inducible gene I (RIG-I), a cytosolic RNA helicase sensor, plays a significant role in the induction of type I interferon responses following viral infection. Here, we identified multiple RNA regions in KSHV as potential virus ligands that bind to RIG-I and stimulate RIG-I-dependent but RNA Pol III-independent IFN-β signaling. Our results expand the role of RIG-I by providing an example of a DNA virus activating a canonical RNA-sensing pathway.
Collapse
Affiliation(s)
- Yugen Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Dirk P Dittmer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Piotr A Mieczkowski
- High Throughput Sequencing Facility, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kurtis M Host
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - William G Fusco
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joseph A Duncan
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Blossom Damania
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
44
|
Zhang B, He Y, Xu Y, Mo F, Mi T, Shen QS, Li C, Li Y, Liu J, Wu Y, Chen G, Zhu W, Qin C, Hu B, Zhou G. Differential antiviral immunity to Japanese encephalitis virus in developing cortical organoids. Cell Death Dis 2018; 9:719. [PMID: 29915260 PMCID: PMC6006338 DOI: 10.1038/s41419-018-0763-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/26/2018] [Accepted: 05/03/2018] [Indexed: 12/23/2022]
Abstract
Japanese encephalitis (JE) caused by Japanese encephalitis virus (JEV) poses a serious threat to the world’s public health yet without a cure. Certain JEV-infected neural cells express a subset of previously identified intrinsic antiviral interferon stimulated genes (ISGs), indicating brain cells retain autonomous antiviral immunity. However, whether this happens in composited brain remains unclear. Human pluripotent stem cell (hPSC)-derived organoids can model disorders caused by human endemic pathogens such as Zika virus, which may potentially address this question and facilitate the discovery of a cure for JE. We thus generated telencephalon organoid and infected them with JEV. We found JEV infection caused significant decline of cell proliferation and increase of cell death in brain organoid, resulting in smaller organoid spheres. JEV tended to infect astrocytes and neural progenitors, especially the population representing outer radial glial cells (oRGCs) of developing human brain. In addition, we revealed variable antiviral immunity in brain organoids of different stages of culture. In organoids of longer culture (older than 8 weeks), but not of early ones (less than 4 weeks), JEV infection caused typical activation of interferon signaling pathway. Preferential infection of oRGCs and differential antiviral response at various stages might explain the much more severe outcomes of JEV infection in the younger, which also provide clues to develop effective therapeutics of such diseases.
Collapse
Affiliation(s)
- Boya Zhang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China.,Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, 200032, PR China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Yangzhige He
- School of Life Sciences, Tsinghua University, Beijing, 100084, PR China
| | - Yanpeng Xu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, PR China
| | - Fan Mo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Tingwei Mi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Qing Sunny Shen
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, 100871, PR China
| | - Chunfeng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, PR China
| | - Yali Li
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, 100871, PR China
| | - Jing Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Yihui Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Guilai Chen
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Wenliang Zhu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Chengfeng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, 100071, PR China.
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China.
| | - Guomin Zhou
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China. .,Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, 200032, PR China.
| |
Collapse
|
45
|
Chan YL, Liao CL, Lin YL. Human Kinase/Phosphatase-Wide RNAi Screening Identified Checkpoint Kinase 2 as a Cellular Factor Facilitating Japanese Encephalitis Virus Infection. Front Cell Infect Microbiol 2018; 8:142. [PMID: 29868498 PMCID: PMC5966567 DOI: 10.3389/fcimb.2018.00142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/20/2018] [Indexed: 11/15/2022] Open
Abstract
Japanese encephalitis virus (JEV), a mosquito-borne flavivirus, causes acute encephalitis in humans with high mortality. Not much is known about the interactions between viral and cellular factors that regulate JEV infection. By using a kinase/phosphatase-wide RNAi screening approach, we identified a cell cycle-regulating molecule, checkpoint kinase 2 (CHK2), that plays a role in regulating JEV replication. JEV infection induced G1 arrest and activated CHK2. Inactivation of CHK2 and its upstream ataxia-telangiectasia mutated kinase in JEV-infected cells by using inhibitors reduced virus replication. Likewise, JEV replication was significantly decreased by knockdown of CHK2 expression with shRNA-producing lentiviral transduction. We identified CHK2 as a cellular factor participating in JEV replication, for a new strategy in addressing JEV infection.
Collapse
Affiliation(s)
- Yi-Lin Chan
- Department of Life Science, Chinese Culture University, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ching-Len Liao
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan.,National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Yi-Ling Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
46
|
Toll-Like Receptors and RIG-I-Like Receptors Play Important Roles in Resisting Flavivirus. J Immunol Res 2018; 2018:6106582. [PMID: 29888293 PMCID: PMC5977009 DOI: 10.1155/2018/6106582] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/02/2018] [Accepted: 03/29/2018] [Indexed: 01/08/2023] Open
Abstract
Flaviviridae family is a class of single-stranded RNA virus, which is fatal to human and animals and mainly prevalent in subtropic and tropic countries. Even though people and animals are barraged with flavivirus infection every year, we have not invented either vaccines or antiviral for most flavivirus infections yet. Innate immunity is the first line of defense in resisting pathogen invasion, serving an important role in a resisting virus. Toll-like receptors (TLRs) and retinoic acid-inducible gene I- (RIG-I-) like receptors (RLRs) are crucial pattern recognition receptors (PRRs) that play essential roles in recognizing and clearing pathogens, including resisting flavivirus. In the present review, we provide a significant reference for further research on the function of innate immunity in resisting flavivirus.
Collapse
|
47
|
Zhang H, Yao X, Ding Y, Xu Z, Liang R, Zhang Y, Wu Y, Li B, Guan B. PI3K signaling pathways modulated white spot syndrome virus (WSSV) replication in Procambarus clarkii. FISH & SHELLFISH IMMUNOLOGY 2018; 76:279-286. [PMID: 29496475 DOI: 10.1016/j.fsi.2018.02.045] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/19/2018] [Accepted: 02/24/2018] [Indexed: 06/08/2023]
Abstract
The PI3K/AKT signaling pathway is commonly exploited to regulate viral replication and affect the fate of infected cells. In the present study, a PI3K-specific inhibitor (LY294002) was employed to pretreat crayfish to evaluate the effects of PI3K/AKT signaling pathway in WSSV replication. The results showed that the WSSV copy numbers in crayfish pretreated with LY294002 were significantly lower than those in Tris-HCl pretreatment crayfish on the sixth and tenth day after WSSV infection. In semigranular cells, the apoptosis rates were up-regulated on the third day post-WSSV infection, and a significantly lower proportion of apoptosis cells were observed in LY294002-pretreatment group. The expression level of Bax, Bax inhibitor-1 and lectin mRNA in haemocytes of crayfish were increased after WSSV infection. After the secondary stimulation with Tris-HCl, the Bax expression level in LY294002-pretreatment crayfish was significantly higher than that of crayfish pretreated with Tris-HCl on the third or sixth day, but the Toll and lectin mRNA expression decreased significantly on the third, sixth and tenth day. The Bax mRNA expression levels in LY294002-WSSV group were significantly higher than those in Tris-HCl-WSSV group on the third and tenth day. The Bax inhibitor-1 mRNA expression levels in LY294002-WSSV group were significantly lower than those in Tris-HCl-WSSV crayfish on the third day. These results together indicated that the hosts PI3K/AKT signaling pathway play positive roles in WSSV replication through the balance between host cell apoptois and innate immune responses. This information is helpful to further understand the role of PI3K/AKT signaling pathway on WSSV replication in Decapoda crustaceans.
Collapse
Affiliation(s)
- Huijing Zhang
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, Shandong, China; State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, Hainan, China
| | - Xuemei Yao
- State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou, 570228, Hainan, China
| | - Yunfei Ding
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Zheng Xu
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Rongning Liang
- Key Laboratory of Coastal Zone Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Yantai, 264003, Shandong, China
| | - Ying Zhang
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, Shandong, China.
| | - Yulong Wu
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Boqing Li
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Bo Guan
- Key Laboratory of Coastal Zone Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research (YIC), Chinese Academy of Sciences (CAS), Yantai, 264003, Shandong, China.
| |
Collapse
|
48
|
Qian L, Zuo Y, Deng W, Miao Y, Liu J, Yuan Y, Guo T, Zhang L, Jin J, Wang J, Zheng H. MCPIP1 is a positive regulator of type I interferons antiviral activity. Biochem Biophys Res Commun 2018; 498:891-897. [PMID: 29545178 DOI: 10.1016/j.bbrc.2018.03.076] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 03/09/2018] [Indexed: 11/18/2022]
Abstract
Type-I interferons (IFN-I) are widely used for antiviral immunotherapy in clinic. Therefore, identification of the regulators of IFN-I antiviral activity is important for developing novel targets for IFN-based antiviral therapy. Monocyte chemoattractant protein 1-induced protein 1 (MCPIP1) is critical for cellular inflammatory responses. However, the roles of MCPIP1 in interferons (IFNs)-mediated antiviral immunity are unexplored. In this study, we demonstrate for the first time that MCPIP1 is an important positive regulator of IFNs antiviral activity. We found that MCPIP1 can promote innate antiviral immunity independently of both its RNase and deubiquitinase activity. Furthermore, we reveal that MCPIP1 is an IFN-induced positive feedback signal molecule which promotes IFN-I-mediated antiviral efficacy. Mechanistically, MCPIP1 does not affect the activation of JAK/STAT upstream of IFN-I signaling, but significantly promotes IFN-I signaling by enhancing ISRE promoter activity and expression of interferon-stimulated genes (ISGs). And MCPIP1-mediated activation of IFN-I signaling is independently of its RNase and deubiquitinase activity. These findings uncover a novel innate antiviral mechanism mediated by the IFN-MCPIP1 axis, and may provide potential targets for enhancing IFNs antiviral therapy.
Collapse
Affiliation(s)
- Liping Qian
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Yibo Zuo
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Wenjun Deng
- Department of Intensive Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Ying Miao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Jin Liu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Yukang Yuan
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Tingting Guo
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Liting Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Jun Jin
- Department of Intensive Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Jun Wang
- Department of Intensive Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215123, China.
| | - Hui Zheng
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province, 215123, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, Jiangsu Province, 215123, China.
| |
Collapse
|
49
|
Abstract
Innate immunity is traditionally thought of as the first line of defense against pathogens that enter the body. It is typically characterized as a rather weak defense mechanism, designed to restrict pathogen replication until the adaptive immune response generates a tailored response and eliminates the infectious agent. However, intensive research in recent years has resulted in better understanding of innate immunity as well as the discovery of many effector proteins, revealing its numerous powerful mechanisms to defend the host. Furthermore, this research has demonstrated that it is simplistic to strictly separate adaptive and innate immune functions since these two systems often work synergistically rather than sequentially. Here, we provide a broad overview of innate pattern recognition receptors in antiviral defense, with a focus on the TRIM family, and discuss their signaling pathways and mechanisms of action with special emphasis on the intracellular antibody receptor TRIM21.
Collapse
Affiliation(s)
| | - Leo C James
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom.
| |
Collapse
|
50
|
Cedillo-Barrón L, García-Cordero J, Shrivastava G, Carrillo-Halfon S, León-Juárez M, Bustos Arriaga J, León Valenzuela P, Gutiérrez Castañeda B. The Role of Flaviviral Proteins in the Induction of Innate Immunity. Subcell Biochem 2018; 88:407-442. [PMID: 29900506 DOI: 10.1007/978-981-10-8456-0_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Flaviviruses are positive, single-stranded, enveloped cytoplasmic sense RNA viruses that cause a variety of important diseases worldwide. Among them, Zika virus, West Nile virus, Japanese encephalitis virus, and Dengue virus have the potential to cause severe disease. Extensive studies have been performed to elucidate the structure and replication strategies of flaviviruses, and current studies are aiming to unravel the complex molecular interactions between the virus and host during the very early stages of infection. The outcomes of viral infection and rapid establishment of the antiviral state, depends on viral detection by pathogen recognition receptors and rapid initiation of signalling cascades to induce an effective innate immune response. Extracellular and intracellular pathogen recognition receptors play a crucial role in detecting flavivirus infection and inducing a robust antiviral response. One of the main hallmarks of flaviviral nonstructural proteins is their multiple strategies to antagonise the interferon system. In this chapter, we summarize the molecular characteristics of flaviviral proteins and discuss how viral proteins target different components of the interferon signalling pathway by blocking phosphorylation, enhancing degradation, and downregulating the expression of major components of the Janus kinase/signal transducer and activator of transcription pathway. We also discuss how the interactions of viral proteins with host proteins facilitate viral pathogenesis. Due to the lack of antivirals or prophylactic treatments for many flaviviral infections, it is necessary to fully elucidate how these viruses disrupt cellular processes to influence pathogenesis and disease outcomes.
Collapse
Affiliation(s)
- L Cedillo-Barrón
- Departamento de Biomedicina Molecular, CINVESTAV IPN, México, D.F, Mexico.
| | - J García-Cordero
- Departamento de Biomedicina Molecular, CINVESTAV IPN, México, D.F, Mexico
| | - G Shrivastava
- Departamento de Biomedicina Molecular, CINVESTAV IPN, México, D.F, Mexico
| | - S Carrillo-Halfon
- Departamento de Biomedicina Molecular, CINVESTAV IPN, México, D.F, Mexico
| | - M León-Juárez
- Department of Immunobiochemistry, National Institute of Perinatology, México City, Mexico
| | - J Bustos Arriaga
- Unidad de Biomedicina. Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autonoma de México, Edo. de México, Mexico
| | - Pc León Valenzuela
- Departamento de Biomedicina Molecular, CINVESTAV IPN, México, D.F, Mexico
| | - B Gutiérrez Castañeda
- Immunology Department UMF Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autonoma de México, Edo. de México, Mexico
| |
Collapse
|