1
|
Yang Z, Xiang Q, Nicholas J. Direct and biologically significant interactions of human herpesvirus 8 interferon regulatory factor 1 with STAT3 and Janus kinase TYK2. PLoS Pathog 2023; 19:e1011806. [PMID: 37983265 PMCID: PMC10695398 DOI: 10.1371/journal.ppat.1011806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 12/04/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
Human herpesvirus 8 (HHV-8) encodes four viral interferon regulatory factors (vIRFs) that target cellular IRFs and/or other innate-immune and stress signaling regulators and suppress the cellular response to viral infection and replication. For vIRF-1, cellular protein targets include IRFs, p53, p53-activating ATM kinase, BH3-only proteins, and antiviral signaling effectors MAVS and STING; vIRF-1 inhibits each, with demonstrated or likely promotion of HHV-8 de novo infection and productive replication. Here, we identify direct interactions of vIRF-1 with STAT3 and STAT-activating Janus kinase TYK2 (the latter reported previously by us to be inhibited by vIRF-1) and suppression by vIRF-1 of cytokine-induced STAT3 activation. Suppression of active, phosphorylated STAT3 (pSTAT3) by vIRF-1 was evident in transfected cells and vIRF-1 ablation in lytically-reactivated recombinant-HHV-8-infected cells led to increased levels of pSTAT3. Using a panel of vIRF-1 deletion variants, regions of vIRF-1 required for interactions with STAT3 and TYK2 were identified, which enabled correlation of STAT3 signaling inhibition by vIRF-1 with TYK2 binding, independently of STAT3 interaction. A viral mutant expressing vIRF-1 deletion-variant Δ198-222 refractory for TYK2 interaction and pSTAT3 suppression was severely compromised for productive replication. Conversely, expression of phosphatase-resistant, protractedly-active STAT3 led to impaired HHV-8 replication. Cells infected with HHV-8 mutants expressing STAT3-refractory vIRF-1 deletion variants or depleted of STAT3 displayed reduced vIRF-1 expression, while custom-peptide-promoted STAT3 interaction could effect increased vIRF-1 expression and enhanced virus replication. Taken together, our data identify vIRF-1 targeting and inhibition of TYK2 as a mechanism of STAT3-signaling suppression and critical for HHV-8 productive replication, the importance of specific pSTAT3 levels for replication, positive roles of STAT3 and vIRF-1-STAT3 interaction in vIRF-1 expression, and significant contributions to lytic replication of STAT3 targeting by vIRF-1.
Collapse
Affiliation(s)
- Zunlin Yang
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Qiwang Xiang
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - John Nicholas
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
2
|
Zhang MQ, Jia X, Cheng CQ, Wang YX, Li YY, Kong LD, Li QQ, Xie F, Yu YL, He YT, Dong QT, Jia ZH, Wang Y, Xu AL. Capsaicin functions as a selective degrader of STAT3 to enhance host resistance to viral infection. Acta Pharmacol Sin 2023; 44:2253-2264. [PMID: 37311796 PMCID: PMC10618195 DOI: 10.1038/s41401-023-01111-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/13/2023] [Indexed: 06/15/2023] Open
Abstract
Although STAT3 has been reported as a negative regulator of type I interferon (IFN) signaling, the effects of pharmacologically inhibiting STAT3 on innate antiviral immunity are not well known. Capsaicin, approved for the treatment of postherpetic neuralgia and diabetic peripheral nerve pain, is an agonist of transient receptor potential vanilloid subtype 1 (TRPV1), with additional recognized potencies in anticancer, anti-inflammatory, and metabolic diseases. We investigated the effects of capsaicin on viral replication and innate antiviral immune response and discovered that capsaicin dose-dependently inhibited the replication of VSV, EMCV, and H1N1. In VSV-infected mice, pretreatment with capsaicin improved the survival rate and suppressed inflammatory responses accompanied by attenuated VSV replication in the liver, lung, and spleen. The inhibition of viral replication by capsaicin was independent of TRPV1 and occurred mainly at postviral entry steps. We further revealed that capsaicin directly bound to STAT3 protein and selectively promoted its lysosomal degradation. As a result, the negative regulation of STAT3 on the type I IFN response was attenuated, and host resistance to viral infection was enhanced. Our results suggest that capsaicin is a promising small-molecule drug candidate, and offer a feasible pharmacological strategy for strengthening host resistance to viral infection.
Collapse
Affiliation(s)
- Mei-Qi Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xin Jia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cui-Qin Cheng
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu-Xi Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yi-Ying Li
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ling-Dong Kong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qi-Qi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Fang Xie
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yan-Li Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu-Ting He
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qiu-Tong Dong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhan-Hong Jia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yao Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China.
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - An-Long Xu
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China.
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
3
|
Kwon G, Yu J, Kim KH. Identifying transcription factors associated with Fusarium graminearum virus 2 accumulation in Fusarium graminearum by phenome-based investigation. Virus Res 2023; 326:199061. [PMID: 36738934 DOI: 10.1016/j.virusres.2023.199061] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/27/2023] [Accepted: 02/01/2023] [Indexed: 02/06/2023]
Abstract
Fusarium graminearum virus 2 (FgV2) infection induces phenotypic changes like reduction of growth rate and virulence with an alteration of the transcriptome, including various transcription factor (TFs) gene transcripts in Fusarium graminearum. Transcription factors are the primary regulator in many cellular processes and are significant in virus-host interactions. However, a detailed study about specific TFs to understand interactions between FgV2 and F. graminearum has yet to be conducted. We transferred FgV2 to a F. graminearum TF gene deletion mutant library to identify host TFs related to FgV2 infection. FgV2-infected TF mutants were classified into three groups depending on colony growth. The FgV2 accumulation level was generally higher in TF mutants showing more reduced growth. Among these FgV2-infected TF mutants, we found several possible TFs that might be involved in FgV2 accumulation, generation of defective interfering RNAs, and transcriptional regulation of FgDICER-2 and FgAGO-1 in response to virus infection. We also investigated the relation between FgV2 accumulation and production of reactive oxygen species (ROS) and DNA damage in fungal host cells by using DNA damage- or ROS-responsive TF deletion mutants. Our studies provide insights into the host factors related to FgV2 infection and bases for further investigation to understand interactions between FgV2 and F. graminearum.
Collapse
Affiliation(s)
- Gudam Kwon
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - Jisuk Yu
- Plant Genomics and Breeding Institute, Seoul National University, Seoul 08826, South Korea.
| | - Kook-Hyung Kim
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea; Plant Genomics and Breeding Institute, Seoul National University, Seoul 08826, South Korea; Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
4
|
Wang Y, Huang H, Li D, Zhao C, Li S, Qin P, Li Y, Yang X, Du W, Li W, Li Y. Identification of niclosamide as a novel antiviral agent against porcine epidemic diarrhea virus infection by targeting viral internalization. Virol Sin 2023; 38:296-308. [PMID: 36702255 PMCID: PMC10176444 DOI: 10.1016/j.virs.2023.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/16/2023] [Indexed: 01/24/2023] Open
Abstract
Porcine epidemic diarrhea virus (PEDV), an enteropathogenic coronavirus, has catastrophic impacts on the global pig industry. However, there remain no effective drugs against PEDV infection. In this study, we utilized a recombinant PEDV expressing renilla luciferase (PEDV-Rluc) to screen potential anti-PEDV agents from an FDA-approved drug library in Vero cells. Four compounds were identified that significantly decreased luciferase activity of PEDV-Rluc. Among them, niclosamide was further characterized because it exhibited the most potent antiviral activity with the highest selectivity index. It can efficiently inhibit viral RNA synthesis, protein expression and viral progeny production of classical and variant PEDV strains in a dose-dependent manner. Time of addition assay showed that niclosamide exhibited potent anti-PEDV activity when added simultaneously with or after virus infection. Furthermore, niclosamide significantly inhibited the entry stage of PEDV infection by affecting viral internalization rather than viral attachment to cells. In addition, a combination with other small molecule inhibitors of endosomal acidification enhanced the anti-PEDV effect of niclosamide in vitro. Taken together, these findings suggested that niclosamide is a novel antiviral agent that might provide a basis for the development of novel drug therapies against PEDV and other related pathogenic coronavirus infections.
Collapse
Affiliation(s)
- Yue Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Huimin Huang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Dongliang Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Chenxu Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Shuai Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Panpan Qin
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Yaqin Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xia Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China
| | - Wenjuan Du
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CL, the Netherlands
| | - Wentao Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Wuhan, 430070, China.
| | - Yongtao Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, China; Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CL, the Netherlands.
| |
Collapse
|
5
|
Srivastava S, Chaudhary N, Dhembla C, Sundd M, Gupta S, Patel AK. STAT3 inhibition mediated upregulation of multiple immune response pathways in dengue infection. Virology 2023; 578:81-91. [PMID: 36473280 DOI: 10.1016/j.virol.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 11/05/2022] [Accepted: 11/21/2022] [Indexed: 11/26/2022]
Abstract
Dengue infection is a world-wide public health threat infecting millions of people annually. Till date no specific antiviral or vaccine is available against dengue virus. Recent evidence indicates that targeting host STAT3 could prove to be an effective antiviral therapy against dengue infection. To explore the potential of STAT3 inhibition as an antiviral strategy, we utilized a STAT3 inhibitor stattic as antiviral agent and performed whole proteome analysis of mammalian cells by mass spectrometry. Differentially expressed proteins among the infected and stattic treated groups were sorted based on their fold change expression and their functional annotation studies were carried out to establish their biological networks. The results presented in the current study indicated that treatment with stattic induces several antiviral pathways to counteract dengue infection. Together with this, we also observed that treatment with stattic downregulates pathways involved in viral transcription and translation thus establishing STAT3 as a suitable target for the development of antiviral interventions. This study establishes the role of STAT3 inhibition as an alternative strategy to counteract DENV pathogenesis. Targeting STAT3 by stattic or similar molecules may help in identifying novel therapeutic interventions against DENV and probably other flaviviruses.
Collapse
Affiliation(s)
- Shikha Srivastava
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Nidhi Chaudhary
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Chetna Dhembla
- Department of Biochemistry, University of Delhi, South Campus, Benito Juarez Marg, New Delhi, 110021, India
| | - Monica Sundd
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110 067, India
| | - Sunny Gupta
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Ashok Kumar Patel
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
6
|
Wang Q, Xin Q, Shang W, Wan W, Xiao G, Zhang LK. Activation of the STAT3 Signaling Pathway by the RNA-Dependent RNA Polymerase Protein of Arenavirus. Viruses 2021; 13:v13060976. [PMID: 34070281 PMCID: PMC8225222 DOI: 10.3390/v13060976] [Citation(s) in RCA: 1] [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: 04/25/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022] Open
Abstract
Arenaviruses cause chronic and asymptomatic infections in their natural host, rodents, and several arenaviruses cause severe hemorrhagic fever that has a high mortality in infected humans, seriously threatening public health. There are currently no FDA-licensed drugs available against arenaviruses; therefore, it is important to develop novel antiviral strategies to combat them, which would be facilitated by a detailed understanding of the interactions between the viruses and their hosts. To this end, we performed a transcriptomic analysis on cells infected with arenavirus lymphocytic choriomeningitis virus (LCMV), a neglected human pathogen with clinical significance, and found that the signal transducer and activator of transcription 3 (STAT3) signaling pathway was activated. A further investigation indicated that STAT3 could be activated by the RNA-dependent RNA polymerase L protein (Lp) of LCMV. Our functional analysis found that STAT3 cannot affect LCMV multiplication in A549 cells. We also found that STAT3 was activated by the Lp of Mopeia virus and Junin virus, suggesting that this activation may be conserved across certain arenaviruses. Our study explored the interactions between arenaviruses and STAT3, which may help us to better understand the molecular and cell biology of arenaviruses.
Collapse
Affiliation(s)
- Qingxing Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China; (Q.W.); (W.S.); (W.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qilin Xin
- UMR754, Viral Infections and Comparative Pathology, 50 Avenue Tony Garnier, CEDEX 07, 69366 Lyon, France;
| | - Weijuan Shang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China; (Q.W.); (W.S.); (W.W.)
| | - Weiwei Wan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China; (Q.W.); (W.S.); (W.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China; (Q.W.); (W.S.); (W.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence: (G.X.); (L.-K.Z.)
| | - Lei-Ke Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, China; (Q.W.); (W.S.); (W.W.)
- University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence: (G.X.); (L.-K.Z.)
| |
Collapse
|
7
|
Srivastava S, Chaudhary N, Ojha A, Guchhait P, Patel AK. Signal transducer and activator of transcription 3 (STAT3) acts as a proviral factor for dengue virus propagation. Virus Res 2021; 300:198436. [PMID: 33901593 DOI: 10.1016/j.virusres.2021.198436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/31/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022]
Abstract
Dengue fever is a significant mosquito-borne viral disease that affects millions of people every year. As a co-existing mechanism, DENV has evolved to evade elimination by the host antiviral immune system. DENV is reported to modulate host interferon response either by attenuating the factors that mediate interferon response like STAT1 and STAT2 or inhibiting the activation of STAT1 or by STAT2 degradation. Through this study we aim to understand how DENV modulates STAT3 mediated interferon response to its own advantage. We employed various techniques like Western blot, Confocal microscopy, RT-PCR to show that STAT3 acts as a pro-viral factor for DV-2 propagation. As per result of the present study STAT3 is upregulated as well as activated by phosphorylation in DV-2 infected A549 cells. Additionally, STAT3 knockdown led to a significant decrease in expression of viral proteins as well as viral replication. We show that DV-2 strategically tweaks STAT3 which is a negative regulator of Type I IFN signaling, in order to evade host Type I and Type III interferon response by upregulating its expression and activation. Our results demonstrate the proviral role of STAT3 for DV-2 propagation which is correlated to activation by tyrosine phosphorylation. Furthermore, since STAT3 is critical factor for DV-2 propagation, its modulation can facilitate targeted development of antivirals against Dengue.
Collapse
Affiliation(s)
- Shikha Srivastava
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Nidhi Chaudhary
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Amrita Ojha
- Disease Biology Laboratory, Regional Centre for Biotechnology, National Capital Region, Biotech Science Cluster, Faridabad, India; Department of Immunology and Microbial Science, The Scripps Research Institute, Jupiter, FL, 33458, USA
| | - Prasenjit Guchhait
- Disease Biology Laboratory, Regional Centre for Biotechnology, National Capital Region, Biotech Science Cluster, Faridabad, India
| | - Ashok Kumar Patel
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
8
|
Yu TM, Li CY, Chuang YW, Chen CH, Lee BK, Chung MC, Chiu HF, Lin MC, Wu MJ, Kao CH. Risk of severe herpes zoster infection in patients with polycystic kidney disease: A nation-wide cohort study with propensity score matching analysis. Int J Clin Pract 2021; 75:e13675. [PMID: 32798268 DOI: 10.1111/ijcp.13675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/01/2020] [Accepted: 08/10/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Polycystic kidney disease (PKD) is suggested to be likely associated with underlying immunological dysregulation. This lymphopenia poses a risk of viral infection. Data to elucidate the herpes virus infection risk in patients with PKD are lacking; therefore, we conducted a national-wide population-based cohort study to investigate the herpes virus risk in PKD patients. METHODS From the Taiwan National Health Insurance Research Database (NHIRD), patients who were hospitalised with a diagnosis of polycystic kidney disease were defined as case group of PKD patients; patients without any diagnosis of PKD during the study period were grouped into the non-PKD cohort. The index date was set as the date when the patients were newly diagnosed with PKD. All study patients were followed up until the occurrence of herpes zoster infection, death, withdrawal from the NHIRD for other reasons, or until December 31, 2013. RESULTS We included 4366 PKD patients and 4366 non-PKD patients. The incidence rate and the risk of developing herpes zoster infection were estimated using multivariate stratified analyses. PKD patients had a 1.97-fold risk of herpes zoster virus infection (aHR = 1.97, 95% CI 1.17-3.31) compared with the non-PKD cohort. On multilayer stratification, PKD patients without any comorbidities had a significantly increased risk of herpes zoster infection (aHR = 3.10, 95% CI 1.37-7.00). CONCLUSION This is the first study to reveal a high risk of severe herpes zoster infection in patients with PKD. High index suspicion of severe herpes zoster infection should be maintained in clinical professionals.
Collapse
Affiliation(s)
- Tung-Min Yu
- Graduate Institute of Biomedical Sciences and School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
- Division of Nephrology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chi-Yuan Li
- Graduate Institute of Biomedical Sciences and School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Ya-Wen Chuang
- Graduate Institute of Biomedical Sciences and School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
- Division of Nephrology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Cheng-Hsu Chen
- Division of Nephrology, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Life Science, Tunghai University, Taichung, Taiwan
| | - Brian K Lee
- Division of Nephrology, Department of Medicine, UCSF Medical Center, San Francisco, CA, USA
| | - Mu-Chi Chung
- Division of Nephrology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hsien-Fu Chiu
- Division of Nephrology, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Life Science, Tunghai University, Taichung, Taiwan
| | - Mei-Chen Lin
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Ming-Ju Wu
- Graduate Institute of Biomedical Sciences and School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
- Division of Nephrology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chia-Hung Kao
- Graduate Institute of Biomedical Sciences and School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
- Department of Nuclear Medicine and PET Center, China Medical University Hospital, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
- Center of Augmented Intelligence in Healthcare, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
9
|
Pant A, Dsouza L, Cao S, Peng C, Yang Z. Viral growth factor- and STAT3 signaling-dependent elevation of the TCA cycle intermediate levels during vaccinia virus infection. PLoS Pathog 2021; 17:e1009303. [PMID: 33529218 PMCID: PMC7880457 DOI: 10.1371/journal.ppat.1009303] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 02/12/2021] [Accepted: 01/11/2021] [Indexed: 12/25/2022] Open
Abstract
Metabolism is a crucial frontier of host-virus interaction as viruses rely on their host cells to provide nutrients and energy for propagation. Vaccinia virus (VACV) is the prototype poxvirus. It makes intensive demands for energy and macromolecules in order to build hundreds and thousands of viral particles in a single cell within hours of infection. Our comprehensive metabolic profiling reveals profound reprogramming of cellular metabolism by VACV infection, including increased levels of the intermediates of the tri-carboxylic acid (TCA) cycle independent of glutaminolysis. By investigating the level of citrate, the first metabolite of the TCA cycle, we demonstrate that the elevation of citrate depends on VACV-encoded viral growth factor (VGF), a viral homolog of cellular epidermal growth factor. Further, the upregulation of citrate is dependent on STAT3 signaling, which is activated non-canonically at the serine727 upon VACV infection. The STAT3 activation is dependent on VGF, and VGF-dependent EGFR and MAPK signaling. Together, our study reveals a novel mechanism by which VACV manipulates cellular metabolism through a specific viral factor and by selectively activating a series of cellular signaling pathways. Vaccinia virus (VACV) is a large DNA virus with an acute and increasing demand for energy and macromolecules to build hundreds and thousands of viral particles in a single cell within hours of infection. The demand postulates reprogramming of the TCA cycle, as it is the central metabolic hub of a cell that generates metabolites for energy production and macromolecule synthesis. We show that VACV infection reprograms cellular metabolism globally, elevating the TCA cycle intermediate levels and modulating related cell metabolism. The elevation of the TCA cycle intermediates depends on the virus-encoded growth factor that stimulates non-canonical STAT3 signaling during VACV infection. Our results provide the metabolic foundation of viral growth factor to boost VACV infection.
Collapse
Affiliation(s)
- Anil Pant
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - Lara Dsouza
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - Shuai Cao
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - Chen Peng
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - Zhilong Yang
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
- * E-mail:
| |
Collapse
|
10
|
Thakuri BKC, Zhang J, Zhao J, Nguyen LN, Nguyen LNT, Schank M, Khanal S, Dang X, Cao D, Lu Z, Wu XY, Jiang Y, El Gazzar M, Ning S, Wang L, Moorman JP, Yao ZQ. HCV-Associated Exosomes Upregulate RUNXOR and RUNX1 Expressions to Promote MDSC Expansion and Suppressive Functions through STAT3-miR124 Axis. Cells 2020; 9:cells9122715. [PMID: 33353065 PMCID: PMC7766103 DOI: 10.3390/cells9122715] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/09/2020] [Accepted: 12/17/2020] [Indexed: 12/17/2022] Open
Abstract
RUNX1 overlapping RNA (RUNXOR) is a long non-coding RNA and plays a pivotal role in the differentiation of myeloid cells via targeting runt-related transcription factor 1 (RUNX1). We and others have previously reported that myeloid-derived suppressor cells (MDSCs) expand and inhibit host immune responses during chronic viral infections; however, the mechanisms responsible for MDSC differentiation and suppressive functions, in particular the role of RUNXOR–RUNX1, remain unclear. Here, we demonstrated that RUNXOR and RUNX1 expressions are significantly upregulated and associated with elevated levels of immunosuppressive molecules, such as arginase 1 (Arg1), inducible nitric oxide synthase (iNOS), signal transducer and activator of transcription 3 (STAT3), and reactive oxygen species (ROS) in MDSCs during chronic hepatitis C virus (HCV) infection. Mechanistically, we discovered that HCV-associated exosomes (HCV-Exo) can induce the expressions of RUNXOR and RUNX1, which in turn regulates miR-124 expression via STAT3 signaling, thereby promoting MDSC differentiation and suppressive functions. Importantly, overexpression of RUNXOR in healthy CD33+ myeloid cells promoted differentiation and suppressive functions of MDSCs. Conversely, silencing RUNXOR or RUNX1 expression in HCV-derived CD33+ myeloid cells significantly inhibited their differentiation and expressions of suppressive molecules and improved the function of co-cultured autologous CD4 T cells. Taken together, these results indicate that the RUNXOR–RUNX1–STAT3–miR124 axis enhances the differentiation and suppressive functions of MDSCs and could be a potential target for immunomodulation in conjunction with antiviral therapy during chronic HCV infection.
Collapse
Affiliation(s)
- Bal Krishna Chand Thakuri
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Jinyu Zhang
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Juan Zhao
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Lam N. Nguyen
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Lam N. T. Nguyen
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Madison Schank
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Sushant Khanal
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Xindi Dang
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Dechao Cao
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Zeyuan Lu
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Xiao Y. Wu
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Yong Jiang
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
| | - Mohamed El Gazzar
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
| | - Shunbin Ning
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Ling Wang
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
| | - Jonathan P. Moorman
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
- Hepatitis (HCV/HBV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN 37614, USA
| | - Zhi Q. Yao
- Center of Excellence for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA; (B.K.C.T.); (J.Z.); (J.Z.); (L.N.N.); (L.N.T.N.); (M.S.); (S.K.); (X.D.); (D.C.); (Z.L.); (X.Y.W.); (Y.J.); (M.E.G.); (S.N.); (L.W.); (J.P.M.)
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN 37614, USA
- Hepatitis (HCV/HBV/HIV) Program, James H. Quillen VA Medical Center, Department of Veterans Affairs, Johnson City, TN 37614, USA
- Correspondence: ; Tel.: +1-423-439-8029; Fax: +1-423-439-7010
| |
Collapse
|
11
|
Abstract
Viruses commonly antagonize the antiviral type I interferon response by targeting signal transducer and activator of transcription 1 (STAT1) and STAT2, key mediators of interferon signaling. Other STAT family members mediate signaling by diverse cytokines important to infection, but their relationship with viruses is more complex. Importantly, virus-STAT interaction can be antagonistic or stimulatory depending on diverse viral and cellular factors. While STAT antagonism can suppress immune pathways, many viruses promote activation of specific STATs to support viral gene expression and/or produce cellular conditions conducive to infection. It is also becoming increasingly clear that viruses can hijack noncanonical STAT functions to benefit infection. For a number of viruses, STAT function is dynamically modulated through infection as requirements for replication change. Given the critical role of STATs in infection by diverse viruses, the virus-STAT interface is an attractive target for the development of antivirals and live-attenuated viral vaccines. Here, we review current understanding of the complex and dynamic virus-STAT interface and discuss how this relationship might be harnessed for medical applications.
Collapse
|
12
|
Wang J, Prinz RA, Liu X, Xu X. In Vitro and In Vivo Antiviral Activity of Gingerenone A on Influenza A Virus Is Mediated by Targeting Janus Kinase 2. Viruses 2020; 12:v12101141. [PMID: 33050000 PMCID: PMC7650803 DOI: 10.3390/v12101141] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Janus kinase (JAK) inhibitors have been developed as novel immunomodulatory drugs and primarily used for treating rheumatoid arthritis and other inflammatory diseases. Recent studies have suggested that this category of anti-inflammatory drugs could be potentially useful for the control of inflammation "storms" in respiratory virus infections. In addition to their role in regulating immune cell functions, JAK1 and JAK2 have been recently identified as crucial cellular factors involved in influenza A virus (IAV) replication and could be potentially targeted for antiviral therapy. Gingerenone A (Gin A) is a compound derived from ginger roots and a dual inhibitor of JAK2 and p70 S6 kinase (S6K1). Our present study aimed to determine the antiviral activity of Gin A on influenza A virus (IAV) and to understand its mechanisms of action. Here, we reported that Gin A suppressed the replication of three IAV subtypes (H1N1, H5N1, H9N2) in four cell lines. IAV replication was also inhibited by Ruxolitinib (Rux), a JAK inhibitor, but not by PF-4708671, an S6K1 inhibitor. JAK2 overexpression enhanced H5N1 virus replication and attenuated Gin A-mediated antiviral activity. In vivo experiments revealed that Gin A treatment suppressed IAV replication in the lungs of H5N1 virus-infected mice, alleviated their body weight loss, and prolonged their survival. Our study suggests that Gin A restricts IAV replication by inhibiting JAK2 activity; Gin A could be potentially useful for the control of influenza virus infections.
Collapse
Affiliation(s)
- Jiongjiong Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China;
| | - Richard A. Prinz
- Department of Surgery, Northshore University HealthSystem, Evanston, IL 60201, USA;
| | - Xiufan Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China;
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiulong Xu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China;
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou 225009, China;
- Institutes of Agricultural Science and Technology Development, Yangzhou University Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Correspondence: ; Tel.: +86-514-8797-7382
| |
Collapse
|
13
|
Goto K, Roca Suarez AA, Wrensch F, Baumert TF, Lupberger J. Hepatitis C Virus and Hepatocellular Carcinoma: When the Host Loses Its Grip. Int J Mol Sci 2020; 21:ijms21093057. [PMID: 32357520 PMCID: PMC7246584 DOI: 10.3390/ijms21093057] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/20/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic infection with hepatitis C virus (HCV) is a major cause of hepatocellular carcinoma (HCC). Novel treatments with direct-acting antivirals achieve high rates of sustained virologic response; however, the HCC risk remains elevated in cured patients, especially those with advanced liver disease. Long-term HCV infection causes a persistent and accumulating damage of the liver due to a combination of direct and indirect pro-oncogenic mechanisms. This review describes the processes involved in virus-induced disease progression by viral proteins, derailed signaling, immunity, and persistent epigenetic deregulation, which may be instrumental to develop urgently needed prognostic biomarkers and as targets for novel chemopreventive therapies.
Collapse
Affiliation(s)
- Kaku Goto
- Université de Strasbourg, F-67000 Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg (IVH), F-67000 Strasbourg, France
| | - Armando Andres Roca Suarez
- Université de Strasbourg, F-67000 Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg (IVH), F-67000 Strasbourg, France
| | - Florian Wrensch
- Université de Strasbourg, F-67000 Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg (IVH), F-67000 Strasbourg, France
| | - Thomas F. Baumert
- Université de Strasbourg, F-67000 Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg (IVH), F-67000 Strasbourg, France
- Pôle Hépato-digestif, Institut Hopitalo-Universitaire, F-67000 Strasbourg, France
- Institut Universitaire de France, F-75231 Paris, France
- Correspondence: (T.F.B.); (J.L.); Tel.: +33-3-68-85-37-03 (T.F.B. & J.L.); Fax: +33-3-68-85-37-24 (T.F.B. & J.L.)
| | - Joachim Lupberger
- Université de Strasbourg, F-67000 Strasbourg, France
- Institut National de la Santé et de la Recherche Médicale, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg (IVH), F-67000 Strasbourg, France
- Correspondence: (T.F.B.); (J.L.); Tel.: +33-3-68-85-37-03 (T.F.B. & J.L.); Fax: +33-3-68-85-37-24 (T.F.B. & J.L.)
| |
Collapse
|
14
|
Wong RR, Abd-Aziz N, Affendi S, Poh CL. Role of microRNAs in antiviral responses to dengue infection. J Biomed Sci 2020; 27:4. [PMID: 31898495 PMCID: PMC6941309 DOI: 10.1186/s12929-019-0614-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/29/2019] [Indexed: 12/13/2022] Open
Abstract
Dengue virus (DENV) is the etiological agent of dengue fever. Severe dengue could be fatal and there is currently no effective antiviral agent or vaccine. The only licensed vaccine, Dengvaxia, has low efficacy against serotypes 1 and 2. Cellular miRNAs are post-transcriptional regulators that could play a role in direct regulation of viral genes. Host miRNA expressions could either promote or repress viral replications. Induction of some cellular miRNAs could help the virus to evade the host immune response by suppressing the IFN-α/β signaling pathway while others could upregulate IFN-α/β production and inhibit the viral infection. Understanding miRNA expressions and functions during dengue infections would provide insights into the development of miRNA-based therapeutics which could be strategized to act either as miRNA antagonists or miRNA mimics. The known mechanisms of how miRNAs impact DENV replication are diverse. They could suppress DENV multiplication by directly binding to the viral genome, resulting in translational repression. Other miRNA actions include modulation of host factors. In addition, miRNAs that could modulate immunopathogenesis are discussed. Major hurdles lie in the development of chemical modifications and delivery systems for in vivo delivery. Nevertheless, advancement in miRNA formulations and delivery systems hold great promise for the therapeutic potential of miRNA-based therapy, as supported by Miravirsen for treatment of Hepatitis C infection which has successfully completed phase II clinical trial.
Collapse
Affiliation(s)
- Rui Rui Wong
- Centre for Virus and Vaccine Research (CVVR), Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Noraini Abd-Aziz
- Centre for Virus and Vaccine Research (CVVR), Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Sarah Affendi
- Centre for Virus and Vaccine Research (CVVR), Sunway University, 47500, Subang Jaya, Selangor, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research (CVVR), Sunway University, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
15
|
Virzì A, Roca Suarez AA, Baumert TF, Lupberger J. Rewiring Host Signaling: Hepatitis C Virus in Liver Pathogenesis. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a037366. [PMID: 31501266 DOI: 10.1101/cshperspect.a037366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatitis C virus (HCV) is a major cause of liver disease including metabolic disease, fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). HCV induces and promotes liver disease progression by perturbing a range of survival, proliferative, and metabolic pathways within the proinflammatory cellular microenvironment. The recent breakthrough in antiviral therapy using direct-acting antivirals (DAAs) can cure >90% of HCV patients. However, viral cure cannot fully eliminate the HCC risk, especially in patients with advanced liver disease or comorbidities. HCV induces an epigenetic viral footprint that promotes a pro-oncogenic hepatic signature, which persists after DAA cure. In this review, we summarize the main signaling pathways deregulated by HCV infection, with potential impact on liver pathogenesis. HCV-induced persistent signaling patterns may serve as biomarkers for the stratification of HCV-cured patients at high risk of developing HCC. Moreover, these signaling pathways are potential targets for novel chemopreventive strategies.
Collapse
Affiliation(s)
- Alessia Virzì
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France.,Université de Strasbourg, 67000 Strasbourg, France
| | - Armando Andres Roca Suarez
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France.,Université de Strasbourg, 67000 Strasbourg, France
| | - Thomas F Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France.,Université de Strasbourg, 67000 Strasbourg, France.,Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, 67000 Strasbourg, France.,Institut Universitaire de France (IUF), 75231 Paris, France
| | - Joachim Lupberger
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France.,Université de Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
16
|
Wang H, Yuan M, Wang S, Zhang L, Zhang R, Zou X, Wang X, Chen D, Wu Z. STAT3 Regulates the Type I IFN-Mediated Antiviral Response by Interfering with the Nuclear Entry of STAT1. Int J Mol Sci 2019; 20:ijms20194870. [PMID: 31575039 PMCID: PMC6801597 DOI: 10.3390/ijms20194870] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/24/2019] [Accepted: 09/28/2019] [Indexed: 12/15/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a multifunctional factor that regulates inflammation and immunity. Knowledge of its regulatory mechanisms is very limited. Here, we showed that enterovirus 71 (EV71) infection induced the phosphorylation of STAT3 and the expression of its downstream inflammatory regulators. Knockdown of STAT3 with siRNAs significantly restricted viral RNA and protein levels, and also reduced viral titers. With further investigation, we found that importin α family member Karyopherin-α1 (KPNA1) was employed by both STAT1 and STAT3 for their nuclear import. The phosphorylated and un-phosphorylated STAT3 competed with STAT1 for binding to the decreased KPNA1 post infection and repressed downstream ISG expression. STAT3 knockdown alleviated the repressed type I IFN-mediated antiviral response upon infection and led to decreased viral replication. Taken together, our data suggested the role of STAT3 in maintaining the balance of inflammation and antiviral responses in the central nervous system (CNS) upon infection.
Collapse
Affiliation(s)
- Huanru Wang
- Center for Public Health Research, Medical School, Nanjing University, Nanjing 210093, China.
| | - Meng Yuan
- Center for Public Health Research, Medical School, Nanjing University, Nanjing 210093, China.
| | - Shuaibo Wang
- Jinling College, Nanjing University, Nanjing 210089, China.
| | - Li Zhang
- Center for Public Health Research, Medical School, Nanjing University, Nanjing 210093, China.
| | - Rui Zhang
- Center for Public Health Research, Medical School, Nanjing University, Nanjing 210093, China.
| | - Xue Zou
- Center for Public Health Research, Medical School, Nanjing University, Nanjing 210093, China.
| | - Xiaohui Wang
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China.
| | - Deyan Chen
- Center for Public Health Research, Medical School, Nanjing University, Nanjing 210093, China.
| | - Zhiwei Wu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing 210093, China.
- State Key Lab of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210023, China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
17
|
Pentagalloylglucose Inhibits the Replication of Rabies Virus via Mediation of the miR-455/SOCS3/STAT3/IL-6 Pathway. J Virol 2019; 93:JVI.00539-19. [PMID: 31243136 DOI: 10.1128/jvi.00539-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/23/2019] [Indexed: 02/07/2023] Open
Abstract
Our previous study showed that pentagalloylglucose (PGG), a naturally occurring hydrolyzable phenolic tannin, possesses significant anti-rabies virus (RABV) activity. In BHK-21 cells, RABV induced the overactivation of signal transducer and activator of transcription 3 (STAT3) by suppressing the expression of suppressor of cytokine signaling 3 (SOCS3). Inhibition of STAT3 by niclosamide, small interfering RNA, or exogenous expression of SOCS3 all significantly suppressed the replication of RABV. Additionally, RABV-induced upregulation of microRNA 455-5p (miR-455-5p) downregulated SOCS3 by directly binding to the 3' untranslated region (UTR) of SOCS3. Importantly, PGG effectively reversed the expression of miR-455-5p and its following SOCS3/STAT3 signaling pathway. Finally, activated STAT3 elicited the expression of interleukin-6 (IL-6), thereby contributing to RABV-associated encephalomyelitis; however, PGG restored the level of IL-6 in vitro and in vivo in a SOCS3/STAT3-dependent manner. Altogether, these data identify a new miR-455-5p/SOCS3/STAT3 signaling pathway that contributes to viral replication and IL-6 production in RABV-infected cells, with PGG exerting its antiviral effect by inhibiting the production of miR-455-5p and the activation of STAT3.IMPORTANCE Rabies virus causes lethal encephalitis in mammals and poses a serious public health threat in many parts of the world. Numerous strategies have been explored to combat rabies; however, their efficacy has always been unsatisfactory. We previously reported a new drug, PGG, which possesses a potent inhibitory activity on RABV replication. Herein, we describe the underlying mechanisms by which PGG exerts its anti-RABV activity. Our results show that RABV induces overactivation of STAT3 in BHK-21 cells, which facilitates viral replication. Importantly, PGG effectively inhibits the activity of STAT3 by disrupting the expression of miR-455-5p and increases the level of SOCS3 by directly targeting the 3' UTR of SOCS3. Furthermore, the downregulated STAT3 inhibits the production of IL-6, thereby contributing to a reduction in the inflammatory response in vivo Our study indicates that PGG effectively inhibits the replication of RABV by the miR-455-5p/SOCS3/STAT3/IL-6-dependent pathway.
Collapse
|
18
|
Convery O, Gargan S, Kickham M, Schroder M, O'Farrelly C, Stevenson NJ. The hepatitis C virus (HCV) protein, p7, suppresses inflammatory responses to tumor necrosis factor (TNF)-α via signal transducer and activator of transcription (STAT)3 and extracellular signal-regulated kinase (ERK)-mediated induction of suppressor of cytokine signaling (SOCS)3. FASEB J 2019; 33:8732-8744. [PMID: 31163989 DOI: 10.1096/fj.201800629rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Viruses use a spectrum of immune evasion strategies that enable infection and replication. The acute phase of hepatitis C virus (HCV) infection is characterized by nonspecific and often mild clinical symptoms, suggesting an immunosuppressive mechanism that, unless symptomatic liver disease presents, allows the virus to remain largely undetected. We previously reported that HCV induced the regulatory protein suppressor of cytokine signaling (SOCS)3, which inhibited TNF-α-mediated inflammatory responses. However, the mechanism by which HCV up-regulates SOCS3 remains unknown. Here we show that the HCV protein, p7, enhances both SOCS3 mRNA and protein expression. A p7 inhibitor reduced SOCS3 induction, indicating that p7's ion channel activity was required for optimal up-regulation of SOCS3. Short hairpin RNA and chemical inhibition revealed that both the Janus kinase-signal transducer and activator of transcription (JAK-STAT) and MAPK pathways were required for p7-mediated induction of SOCS3. HCV-p7 expression suppressed TNF-α-mediated IκB-α degradation and subsequent NF-κB promoter activity, revealing a new and functional, anti-inflammatory effect of p7. Together, these findings identify a molecular mechanism by which HCV-p7 induces SOCS3 through STAT3 and ERK activation and demonstrate that p7 suppresses proinflammatory responses to TNF-α, possibly explaining the lack of inflammatory symptoms observed during early HCV infection.-Convery, O., Gargan, S., Kickham, M., Schroder, M., O'Farrelly, C., Stevenson, N. J. The hepatitis C virus (HCV) protein, p7, suppresses inflammatory responses to tumor necrosis factor (TNF)-α via signal transducer and activator of transcription (STAT)3 and extracellular signal-regulated kinase (ERK)-mediated induction of suppressor of cytokine signaling (SOCS)3.
Collapse
Affiliation(s)
- Orla Convery
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Siobhan Gargan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Nigel J Stevenson
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
19
|
|
20
|
Virzì A, Roca Suarez AA, Baumert TF, Lupberger J. Oncogenic Signaling Induced by HCV Infection. Viruses 2018; 10:v10100538. [PMID: 30279347 PMCID: PMC6212953 DOI: 10.3390/v10100538] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/29/2018] [Accepted: 09/30/2018] [Indexed: 02/07/2023] Open
Abstract
The liver is frequently exposed to toxins, metabolites, and oxidative stress, which can challenge organ function and genomic stability. Liver regeneration is therefore a highly regulated process involving several sequential signaling events. It is thus not surprising that individual oncogenic mutations in hepatocytes do not necessarily lead to cancer and that the genetic profiles of hepatocellular carcinomas (HCCs) are highly heterogeneous. Long-term infection with hepatitis C virus (HCV) creates an oncogenic environment by a combination of viral protein expression, persistent liver inflammation, oxidative stress, and chronically deregulated signaling events that cumulate as a tipping point for genetic stability. Although novel direct-acting antivirals (DAA)-based treatments efficiently eradicate HCV, the associated HCC risk cannot be fully eliminated by viral cure in patients with advanced liver disease. This suggests that HCV may persistently deregulate signaling pathways beyond viral cure and thereby continue to perturb cancer-relevant gene function. In this review, we summarize the current knowledge about oncogenic signaling pathways derailed by chronic HCV infection. This will not only help to understand the mechanisms of hepatocarcinogenesis but will also highlight potential chemopreventive strategies to help patients with a high-risk profile of developing HCC.
Collapse
Affiliation(s)
- Alessia Virzì
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France.
- Université de Strasbourg, 67000 Strasbourg, France.
| | - Armando Andres Roca Suarez
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France.
- Université de Strasbourg, 67000 Strasbourg, France.
| | - Thomas F Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France.
- Université de Strasbourg, 67000 Strasbourg, France.
- Pôle Hépato-digestif, Institut Hospitalo-universitaire, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France.
| | - Joachim Lupberger
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France.
- Université de Strasbourg, 67000 Strasbourg, France.
| |
Collapse
|
21
|
Li CW, Jheng BR, Chen BS. Investigating genetic-and-epigenetic networks, and the cellular mechanisms occurring in Epstein-Barr virus-infected human B lymphocytes via big data mining and genome-wide two-sided NGS data identification. PLoS One 2018; 13:e0202537. [PMID: 30133498 PMCID: PMC6105016 DOI: 10.1371/journal.pone.0202537] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/03/2018] [Indexed: 12/17/2022] Open
Abstract
Epstein-Barr virus (EBV), also known as human herpesvirus 4, is prevalent in all human populations. EBV mainly infects human B lymphocytes and epithelial cells, and is therefore associated with their various malignancies. To unravel the cellular mechanisms during the infection, we constructed interspecies networks to investigate the molecular cross-talk mechanisms between human B cells and EBV at the first (0-24 hours) and second (8-72 hours) stages of EBV infection. We first constructed a candidate genome-wide interspecies genetic-and-epigenetic network (the candidate GIGEN) by big database mining. We then pruned false positives in the candidate GIGEN to obtain the real GIGENs at the first and second infection stages in the lytic phase by their corresponding next-generation sequencing data through dynamic interaction models, the system identification approach, and the system order detection method. The real GIGENs are very complex and comprise protein-protein interaction networks, gene/microRNA (miRNA)/long non-coding RNA regulation networks, and host-virus cross-talk networks. To understand the molecular cross-talk mechanisms underlying EBV infection, we extracted the core GIGENs including host-virus core networks and host-virus core pathways from the real GIGENs using the principal network projection method. According to the results, we found that the activities of epigenetics-associated human proteins or genes were initially inhibited by viral proteins and miRNAs, and human immune responses were then dysregulated by epigenetic modification. We suggested that EBV exploits viral proteins and miRNAs, such as EBNA1, BPLF1, BALF3, BVRF1 and miR-BART14, to develop its defensive mechanism to defeat multiple immune attacks by the human immune system, promotes virion production, and facilitates the transportation of viral particles by activating the human genes NRP1 and CLIC5. Ultimately, we propose a therapeutic intervention comprising thymoquinone, valpromide, and zebularine to act as inhibitors of EBV-associated malignancies.
Collapse
Affiliation(s)
- Cheng-Wei Li
- Laboratory of Control and Systems Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Bo-Ren Jheng
- Laboratory of Control and Systems Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Bor-Sen Chen
- Laboratory of Control and Systems Biology, National Tsing Hua University, Hsinchu, Taiwan
- * E-mail:
| |
Collapse
|
22
|
Lu NT, Liu NM, Patel D, Vu JQ, Liu L, Kim CY, Cho P, Khachatoorian R, Patel N, Magyar CE, Ganapathy E, Arumugaswami V, Dasgupta A, French SW. Oncoprotein Stathmin Modulates Sensitivity to Apoptosis in Hepatocellular Carcinoma Cells During Hepatitis C Viral Replication. J Cell Death 2018; 11:1179066018785141. [PMID: 30034249 PMCID: PMC6047100 DOI: 10.1177/1179066018785141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/28/2018] [Indexed: 01/28/2023] Open
Abstract
Patients with chronic hepatitis C virus (HCV) infection risk complications of
cirrhosis, liver failure, and hepatocellular carcinoma (HCC). Previously, our
proteomic examination of hepatocytes carrying a HCV-replicon revealed that
deregulation of cytoskeletal dynamics may be a potential mechanism of
viral-induced HCC growth. Here, we demonstrate the effect of HCV replication on
the microtubule regulator stathmin (STMN1) in HCC cells. We further explore how
the altered activity or synthesis of stathmin affects cellular proliferation and
sensitivity to apoptosis in control HCC cells (Huh7.5) and experimental
HCV-replicon harboring HCC cells (R-Huh7.5). The HCV-replicon harboring HCC
cells (R-Huh 7.5) lack viral structural genes/proteins for acute infectivity and
thus is the standard model for in vitro chronic infection study. Knockdown of
endogenous stathmin reduced sensitivity to apoptosis in replicon cells.
Meanwhile, constitutively active stathmin increased sensitivity to apoptosis in
replicon cells. In addition, overexpression of constitutively active stathmin
reduced cell proliferation in both control and replicon cells. These findings
implicate, for the first time, a novel role for stathmin in viral
replication–related apoptosis. Stathmin’s potential role in HCV replication and
HCC make it a candidate for the future study of viral-induced malignancies.
Collapse
Affiliation(s)
- Nu T Lu
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.,Department of Hematology and Oncology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Natalie M Liu
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Darshil Patel
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - James Q Vu
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Lisa Liu
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Chae Yeon Kim
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Peter Cho
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Ronik Khachatoorian
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Nikita Patel
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Clara E Magyar
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Ekambaram Ganapathy
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Vaithilingaraja Arumugaswami
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.,Department of Surgery and Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Asim Dasgupta
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Samuel Wheeler French
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.,UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| |
Collapse
|
23
|
Chang Z, Wang Y, Zhou X, Long JE. STAT3 roles in viral infection: antiviral or proviral? Future Virol 2018; 13:557-574. [PMID: 32201498 PMCID: PMC7079998 DOI: 10.2217/fvl-2018-0033] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a transcription factor which can be activated by cytokines, growth factor receptors, and nonreceptor-like tyrosine kinase. An activated STAT3 translocates into the nucleus and combines with DNA to regulate the expression of target genes involved in cell proliferation, differentiation, apoptosis and metastasis. Recent studies have shown that STAT3 plays important roles in viral infection and pathogenesis. STAT3 exhibits a proviral function in several viral infections, including those of HBV, HCV, HSV-1, varicella zoster virus, human CMV and measles virus. However, in some circumstances, STAT3 has an antiviral function in other viral infections, such as enterovirus 71, severe acute respiratory syndrome coronavirus and human metapneumovirus. This review summarizes the roles of STAT3 in viral infection and pathogenesis, and briefly discusses the molecular mechanisms involved in these processes.
Collapse
Affiliation(s)
- Zhangmei Chang
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Kunshan Center For Disease Control & Prevention, 458 Tongfengxi Road, Kunshan, Jiangsu, 215301, PR China.,Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Kunshan Center For Disease Control & Prevention, 458 Tongfengxi Road, Kunshan, Jiangsu, 215301, PR China
| | - Yan Wang
- Department of Medical Microbiology & Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan R., Shanghai 200032, PR China.,Department of Medical Microbiology & Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan R., Shanghai 200032, PR China
| | - Xin Zhou
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | - Jian-Er Long
- Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Department of Medical Microbiology & Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan R., Shanghai 200032, PR China.,Key Laboratory of Medical Molecular Virology of Ministries of Education & Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Department of Medical Microbiology & Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan R., Shanghai 200032, PR China
| |
Collapse
|
24
|
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a key regulator of numerous physiological functions, including the immune response. As pathogens elicit an acute phase response with concerted activation of STAT3, they are confronted with two evolutionary options: either curtail it or employ it. This has important consequences for the host, since abnormal STAT3 function is associated with cancer development and other diseases. This review provides a comprehensive outline of how human viruses cope with STAT3-mediated inflammation and how this affects the host. Finally, we discuss STAT3 as a potential target for antiviral therapy.
Collapse
Affiliation(s)
- Armando Andres Roca Suarez
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Nicolaas Van Renne
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Thomas F. Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Pôle Hépato-digestif, Institut Hospitalo-universitaire, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Joachim Lupberger
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- * E-mail:
| |
Collapse
|
25
|
Wang Y, Li Y. MiR-29c inhibits HCV replication via activation of type I IFN response by targeting STAT3 in JFH-1-infected Huh7 cells. RSC Adv 2018; 8:8164-8172. [PMID: 35542013 PMCID: PMC9078521 DOI: 10.1039/c7ra12815k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/13/2018] [Indexed: 01/05/2023] Open
Abstract
Background: MiR-29c, a member of the miR-29 family, has been recognized to play an important role in hepatitis C virus (HCV) infection. However, the underlying molecular mechanism of miR-29c involved in HCV replication is not fully understood. Methods: RT-qPCR assay was used to detect the expression pattern of miR-29c and signal transducer and activator of transcription 3 (STAT3) mRNA in JFH-1-infected Huh7 cells. HCV replication was evaluated by the expression of HCV RNA, non-structural protein 5A (NS5A) and non-structural protein 3 (NS3). Dual-Luciferase Reporter assay was applied to search for the candidate target mRNAs of miR-29c. Western blot assay was performed to detect the protein level of double-stranded RNA-dependent protein kinase R (PKR), (2'-5')-oligoadenylate synthetase (OAS) and interferon regulatory transcription factor 1 (IRF1). Results: miR-29c expression was down-regulated, and STAT3 mRNA and protein expressions were up-regulated in JFH-1-infected Huh7 cells. MiR-29c overexpression or STAT3 knockdown repressed HCV replication, while miR-29c depletion or STAT3 upregulation promoted HCV replication. Additionally, STAT3 was a direct target of miR-29c, and miR-29c suppressed STAT3 protein expression in Huh7 cells. Moreover, STAT3 overexpression reversed miR-29c-mediated suppression on HCV replication. Furthermore, the anti-miR-29c-mediated inhibitory effect on type I IFN response was abated following STAT3 knockdown. Conclusions: miR-29c might repress HCV infection via promoting type I IFN response by targeting STAT3 in JFH-1-infected Huh7 cells, offering a promising avenue for HCV treatment.
Collapse
Affiliation(s)
- Yanjing Wang
- Department of Infectious Disease, Huaihe Hospital of Henan University No. 115 West Road, Gulou District Kaifeng 475000 China +86-13633784192
| | - Yuanyuan Li
- Department of Infectious Disease, Huaihe Hospital of Henan University No. 115 West Road, Gulou District Kaifeng 475000 China +86-13633784192
| |
Collapse
|
26
|
Dai CY, Tsai YS, Chou WW, Liu T, Huang CF, Wang SC, Tsai PC, Yeh ML, Hsieh MY, Huang CI, Vanson Liu SY, Huang JF, Chuang WL, Yu ML. The IL-6/STAT3 pathway upregulates microRNA-125b expression in hepatitis C virus infection. Oncotarget 2018; 9:11291-11302. [PMID: 29541414 PMCID: PMC5834265 DOI: 10.18632/oncotarget.24129] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/01/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND/AIMS MicroRNA-125b (miR-125b) has been found to regulate inflammation and acts as an oncogene in many cancers. The mechanisms of miR-125b expression during hepatitis C virus (HCV) infection remain to be clarified. The present study aims to identify the factors that might regulate miR-125b expression in HCV infection. RESULTS High expression of miR-125b was found to correlate with HCV infection in replicon cells and in sera from HCV-infected patients, whereas the miR-125b inhibitor reduced HCV gene expression. The interleukin 6 (IL-6)/signal transducer and activator of transcription 3 (STAT3) pathway plays an inducible effect on miR-125b gene expression. STAT3 siRNA or inhibitor could reduce HCV replication. MATERIALS AND METHODS HCV replicon cells Con1 (type 1b) and Huh7/Ava5 (type 1b) were treated with 17-hydroxy-jolkinolide B (HJB) or STAT3 siRNA. Cell viability assay and Renilla Luciferase Assay were used. Fragments of the miR-125b-1 promoter were constructed for the luciferase reporter assay. PSMB8, PSMB9, miR-125b-1, and miR-125b-2 expression was determined using TaqMan® Gene Expression Assays. Western blot analysis was performed to assess protein abundance. CONCLUSIONS This study elucidates a novel pathway for miR-125b in the pathogenesis of chronic HCV infection and suggests it as a possible target for treating HCV infection.
Collapse
Affiliation(s)
- Chia-Yen Dai
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Occupational and Environmental Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Health Management Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Faculty of Internal Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Shan Tsai
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Wen-Wen Chou
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Tawei Liu
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chung-Feng Huang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Occupational and Environmental Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Faculty of Internal Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Chi Wang
- Health Management Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Pei-Chien Tsai
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ming-Lun Yeh
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Faculty of Internal Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Yen Hsieh
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ching-I Huang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shang-Yin Vanson Liu
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Jee-Fu Huang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Faculty of Internal Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Long Chuang
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Faculty of Internal Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Lung Yu
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Occupational and Environmental Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Health Management Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Faculty of Internal Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| |
Collapse
|
27
|
Valadkhan S, Fortes P. Regulation of the Interferon Response by lncRNAs in HCV Infection. Front Microbiol 2018; 9:181. [PMID: 29503633 PMCID: PMC5820368 DOI: 10.3389/fmicb.2018.00181] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/26/2018] [Indexed: 12/24/2022] Open
Affiliation(s)
- Saba Valadkhan
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- *Correspondence: Saba Valadkhan, Puri Fortes,
| | - Puri Fortes
- Center for Applied Medical Research, Department of Gene Therapy and Hepatology, Navarra Institute for Health Research (IdiSNA), University of Navarra, Pamplona, Spain
- *Correspondence: Saba Valadkhan, Puri Fortes,
| |
Collapse
|
28
|
Chang Z, Wang Y, Bian L, Liu Q, Long JE. Enterovirus 71 antagonizes the antiviral activity of host STAT3 and IL-6R with partial dependence on virus-induced miR-124. J Gen Virol 2017; 98:3008-3025. [PMID: 29120300 DOI: 10.1099/jgv.0.000967] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Enterovirus 71 (EV71) has caused major outbreaks of hand, foot and mouth disease. EV71 infections increase the production of many host cytokines and pro-inflammatory factors, including interleukin (IL)-6, IL-10 and COX-2. Some of these molecules could stimulate the signal transducer and activator of transcription 3 (STAT3), which plays a key role in regulating host immune responses and several viral diseases. However, the role of STAT3 in EV71 infection remains unknown. This study found that the phosphorylation levels of STAT3 (pY705-STAT3) are closely related to EV71 infection. Further experiments revealed that STAT3 exerts an anti-EV71 activity. However, the antiviral activity of STAT3 is partially antagonized by EV71-induced miR-124, which directly targets STAT3 mRNA. Similarly, IL-6R, the α-subunit of the IL-6 receptor complex, exhibits anti-EV71 activity and is directly targeted by the virus-induced miR-124. These results indicate that EV71 can evade host IL-6R- and STAT3-mediated antiviral activities by EV71-induced miR-124. This suggests that controlling miR-124 and the downstream targets, IL-6R and STAT3, might benefit the antiviral treatment of EV71 infection.
Collapse
Affiliation(s)
- Zhangmei Chang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | - Yan Wang
- Department of Medical Microbiology and Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Liang Bian
- Department of Medical Microbiology and Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| | - Qingqing Liu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | - Jian-Er Long
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College of Fudan University, Shanghai 200032, PR China.,Department of Medical Microbiology and Parasitology, Laboratory of Medical Microbiology, Shanghai Medical College of Fudan University, 138 Yixueyuan Road, Shanghai 200032, PR China
| |
Collapse
|
29
|
Amano R, Yamashita A, Kasai H, Hori T, Miyasato S, Saito S, Yokoe H, Takahashi K, Tanaka T, Otoguro T, Maekawa S, Enomoto N, Tsubuki M, Moriishi K. Cinnamic acid derivatives inhibit hepatitis C virus replication via the induction of oxidative stress. Antiviral Res 2017; 145:123-130. [PMID: 28780423 DOI: 10.1016/j.antiviral.2017.07.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/02/2017] [Accepted: 07/30/2017] [Indexed: 02/08/2023]
Abstract
Several cinnamic acid derivatives have been reported to exhibit antiviral activity. In this study, we prepared 17 synthetic cinnamic acid derivatives and screened them to identify an effective antiviral compound against hepatitis C virus (HCV). Compound 6, one of two hit compounds, suppressed the viral replications of genotypes 1b, 2a, 3a, and 4a with EC50 values of 1.5-8.1 μM and SI values of 16.2-94.2. The effect of compound 6 on the phosphorylation of Tyr705 in signal transducer and activator of transcription 3 (STAT3) was investigated because a cinnamic acid derivative AG490 was reported to suppress HCV replication and the activity of Janus kinase (JAK) 2. Compound 6 potently suppressed HCV replication, but it did not inhibit the JAK1/2-dependent phosphorylation of STAT3 Tyr705 at the same concentration. Furthermore, a pan-JAK inhibitor tofacitinib potently impaired phosphorylation of STAT3 Tyr 705, but it did not inhibit HCV replication in the replicon cells and HCV-infected cells at the same concentration, supporting the notion that the phosphorylated state of STAT3 Tyr705 is not necessarily correlated with HCV replication. The production of reactive oxygen species (ROS) was induced by treatment with compound 6, whereas N-acetyl-cysteine restored HCV replication and impaired ROS production in the replicon cells treated with compound 6. These data suggest that compound 6 inhibits HCV replication via the induction of oxidative stress.
Collapse
Affiliation(s)
- Ryota Amano
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Atsuya Yamashita
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Hirotake Kasai
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Tomoka Hori
- Institute of Medical Chemistry, Hoshi University, Tokyo, Japan
| | - Sayoko Miyasato
- Institute of Medical Chemistry, Hoshi University, Tokyo, Japan
| | - Setsu Saito
- Institute of Medical Chemistry, Hoshi University, Tokyo, Japan
| | - Hiromasa Yokoe
- Institute of Medical Chemistry, Hoshi University, Tokyo, Japan
| | | | - Tomohisa Tanaka
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Teruhime Otoguro
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Shinya Maekawa
- The First Department of Internal Medicine, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | - Nobuyuki Enomoto
- The First Department of Internal Medicine, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan
| | | | - Kohji Moriishi
- Department of Microbiology, Faculty of Medicine, Graduate Faculty of Interdisciplinary Research, University of Yamanashi, Yamanashi, Japan.
| |
Collapse
|
30
|
Bosch-Barrera J, Queralt B, Menendez JA. Targeting STAT3 with silibinin to improve cancer therapeutics. Cancer Treat Rev 2017; 58:61-69. [DOI: 10.1016/j.ctrv.2017.06.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 06/15/2017] [Indexed: 02/08/2023]
|
31
|
Kim JH, Sung PS, Lee EB, Hur W, Park DJ, Shin EC, Windisch MP, Yoon SK. GRIM-19 Restricts HCV Replication by Attenuating Intracellular Lipid Accumulation. Front Microbiol 2017; 8:576. [PMID: 28443075 PMCID: PMC5387058 DOI: 10.3389/fmicb.2017.00576] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 03/20/2017] [Indexed: 12/15/2022] Open
Abstract
Gene-associated with retinoid-interferon-induced mortality 19 (GRIM-19) targets multiple signaling pathways involved in cell death and growth. However, the role of GRIM-19 in the pathogenesis of hepatitis virus infections remains unexplored. Here, we investigated the restrictive effects of GRIM-19 on the replication of hepatitis C virus (HCV). We found that GRIM-19 protein levels were reduced in HCV-infected Huh7 cells and Huh7 cells harboring HCV replicons. Moreover, ectopically expressed GRIM-19 caused a reduction in both intracellular viral RNA levels and secreted viruses in HCVcc-infected cell cultures. The restrictive effect on HCV replication was restored by treatment with siRNA against GRIM-19. Interestingly, GRIM-19 overexpression did not alter the level of phosphorylated STAT3 or its subcellular distribution. Strikingly, forced expression of GRIM-19 attenuated an increase in intracellular lipid droplets after oleic acid (OA) treatment or HCVcc infection. GRIM-19 overexpression abrogated fatty acid-induced upregulation of sterol regulatory element-binding transcription factor-1 (SREBP-1c), resulting in attenuated expression of its target genes such as fatty acid synthase (FAS) and acetyl CoA carboxylase (ACC). Treatment with OA or overexpression of SREBP-1c in GRIM-19-expressing, HCVcc-infected cells restored HCV replication. Our results suggest that GRIM-19 interferes with HCV replication by attenuating intracellular lipid accumulation and therefore is an anti-viral host factor that could be a promising target for HCV treatment.
Collapse
Affiliation(s)
- Jung-Hee Kim
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, The Catholic University of KoreaSeoul, South Korea
| | - Pil S Sung
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and TechnologyDaejeon, South Korea
| | - Eun B Lee
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, The Catholic University of KoreaSeoul, South Korea
| | - Wonhee Hur
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, The Catholic University of KoreaSeoul, South Korea
| | - Dong J Park
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, The Catholic University of KoreaSeoul, South Korea
| | - Eui-Cheol Shin
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and TechnologyDaejeon, South Korea
| | - Marc P Windisch
- Hepatitis Research Laboratory, Discovery Biology Department, Institut Pasteur Korea, Seongnam-siGyeonggi-do, South Korea
| | - Seung K Yoon
- The Catholic University Liver Research Center and WHO Collaborating Center of Viral Hepatitis, The Catholic University of KoreaSeoul, South Korea
| |
Collapse
|
32
|
Bathige SDNK, Thulasitha WS, Umasuthan N, Jayasinghe JDHE, Wan Q, Nam BH, Lee J. A homolog of teleostean signal transducer and activator of transcription 3 (STAT3) from rock bream, Oplegnathus fasciatus: Structural insights, transcriptional modulation, and subcellular localization. Vet Immunol Immunopathol 2017; 186:29-40. [PMID: 28413047 DOI: 10.1016/j.vetimm.2017.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 12/23/2016] [Accepted: 02/26/2017] [Indexed: 10/20/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is one of the crucial transcription factors in the Janus kinase (JAK)/STAT signaling pathway, and it was previously considered as acute phase response factor. A number of interleukins (ILs) such as IL-5, IL-6, IL-9, IL-10, IL-12, and IL-22 are known to be involved in activation of STAT3. In addition, various growth factors and pathogenic or oxidative stresses mediate the activation of a wide range of functions via STAT3. In this study, a STAT3 homolog was identified and functionally characterized from rock bream (RbSTAT3), Oplegnathus fasciatus. In silico characterization revealed that the RbSTAT3 amino acid sequence shares highly conserved common domain architectural features including N-terminal domain, coiled coil domain, DNA binding domain, linker domain, and Src homology 2 (SH2) domains. In addition, a fairly conserved transcriptional activation domain (TAD) was located at the C-terminus. Comparison of RbSTAT3 with other counterparts revealed higher identities (>90%) with fish orthologs. The genomic sequence of RbSTAT3 was obtained from a bacterial artificial chromosome (BAC) library, and was identified as a multi-exonic gene (24 exons), as found in other vertebrates. Genomic structural comparison and phylogenetic studies have showed that the evolutionary routes of teleostean and non-teleostean vertebrates were distinct. Quantitative real time PCR (qPCR) analysis revealed that the spatial distribution of RbSTAT3 mRNA expression was ubiquitous and highly detectable in blood, heart, and liver tissues. Transcriptional modulation of RbSTAT3 was examined in blood and liver tissues after challenges with bacteria (Edwardsiella tarda and Streptococcus iniae), rock bream irido virus (RBIV), and immune stimulants (LPS and poly (I:C)). Significant changes in RbSTAT3 transcription were also observed in response to tissue injury. In addition, the transcriptional up-regulation of RbSTAT3 was detected in rock bream heart cells upon recombinant rock bream IL-10 (rRbIL-10) treatment. Subcellular localization and nuclear translocation of rock bream STAT3 following poly (I:C) treatment were also demonstrated. Taken together, the results of the current study provide important evidence for potential roles of rock bream STAT3 in the immune system and wound healing processes.
Collapse
Affiliation(s)
- S D N K Bathige
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Fish Vaccine Research Center, Jeju National University, Jeju Special Self-Governing Province 63243, Republic of Korea
| | - William Shanthakumar Thulasitha
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Fish Vaccine Research Center, Jeju National University, Jeju Special Self-Governing Province 63243, Republic of Korea
| | - Navaneethaiyer Umasuthan
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Fish Vaccine Research Center, Jeju National University, Jeju Special Self-Governing Province 63243, Republic of Korea
| | - J D H E Jayasinghe
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Fish Vaccine Research Center, Jeju National University, Jeju Special Self-Governing Province 63243, Republic of Korea
| | - Qiang Wan
- Fish Vaccine Research Center, Jeju National University, Jeju Special Self-Governing Province 63243, Republic of Korea
| | - Bo-Hye Nam
- Biotechnology Research Division, National Institute of Fisheries science, 408-1 Sirang-ri, Gijang-up, Gijang-gun, Busan 46083, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Fish Vaccine Research Center, Jeju National University, Jeju Special Self-Governing Province 63243, Republic of Korea.
| |
Collapse
|
33
|
Saito F, Araki K, Yokobori T, Ishii N, Tsukagoshi M, Watanabe A, Kubo N, Altan B, Shirabe K, Kuwano H. High expression of karyopherin-α2 and stathmin 1 is associated with proliferation potency and transformation in the bile duct and gall bladder epithelia in the cases of pancreaticobiliary maljunction. J Surg Oncol 2016; 114:462-8. [DOI: 10.1002/jso.24330] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/05/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Fumiyoshi Saito
- Department of General Surgical Science; Graduate School of Medicine; Gunma University; Showamachi, Maebashi Gunma Japan
- Department of Hepatobiliary and Pancreatic Surgery; Graduate School of Medicine; Gunma University; Maebashi Gunma Japan
| | - Kenichiro Araki
- Department of General Surgical Science; Graduate School of Medicine; Gunma University; Showamachi, Maebashi Gunma Japan
- Department of Hepatobiliary and Pancreatic Surgery; Graduate School of Medicine; Gunma University; Maebashi Gunma Japan
| | - Takehiko Yokobori
- Department of Molecular Pharmacology and Oncology; Graduate School of Medicine; Gunma University; Maebashi Gunma Japan
| | - Norihiro Ishii
- Department of General Surgical Science; Graduate School of Medicine; Gunma University; Showamachi, Maebashi Gunma Japan
- Department of Hepatobiliary and Pancreatic Surgery; Graduate School of Medicine; Gunma University; Maebashi Gunma Japan
| | - Mariko Tsukagoshi
- Department of General Surgical Science; Graduate School of Medicine; Gunma University; Showamachi, Maebashi Gunma Japan
- Department of Hepatobiliary and Pancreatic Surgery; Graduate School of Medicine; Gunma University; Maebashi Gunma Japan
| | - Akira Watanabe
- Department of General Surgical Science; Graduate School of Medicine; Gunma University; Showamachi, Maebashi Gunma Japan
- Department of Hepatobiliary and Pancreatic Surgery; Graduate School of Medicine; Gunma University; Maebashi Gunma Japan
| | - Norio Kubo
- Department of General Surgical Science; Graduate School of Medicine; Gunma University; Showamachi, Maebashi Gunma Japan
- Department of Hepatobiliary and Pancreatic Surgery; Graduate School of Medicine; Gunma University; Maebashi Gunma Japan
| | - Bolag Altan
- Department of General Surgical Science; Graduate School of Medicine; Gunma University; Showamachi, Maebashi Gunma Japan
| | - Ken Shirabe
- Department of Hepatobiliary and Pancreatic Surgery; Graduate School of Medicine; Gunma University; Maebashi Gunma Japan
| | - Hiroyuki Kuwano
- Department of General Surgical Science; Graduate School of Medicine; Gunma University; Showamachi, Maebashi Gunma Japan
| |
Collapse
|
34
|
Hepatitis C virus and its protein NS4B activate the cancer-related STAT3 pathway via the endoplasmic reticulum overload response. Arch Virol 2016; 161:2149-59. [PMID: 27180099 DOI: 10.1007/s00705-016-2892-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 05/10/2016] [Indexed: 01/05/2023]
Abstract
Oxidative stress induces the activation of signal transducer and activator of transcription 3 (STAT3), which plays an important role in hepatocellular carcinoma (HCC). We have previously reported that hepatitis C virus (HCV) and its protein NS4B induce the production of reactive oxygen species (ROS) via the endoplasmic reticulum overload response (EOR) in human hepatocytes. Here, we found that NS4B and HCV induce STAT3 activation and stimulate the expression of cancer-related STAT3 target genes, including VEGF, c-myc, MMP-9 and Mcl-1, by EOR in human hepatocytes. Moreover, the cancer-related STAT3 pathway activated by NS4B and HCV via EOR were found to promote human hepatocyte viability. Taken together, these findings revealed that HCV NS4B might contribute to HCC by activating the EOR-mediated cancer-related STAT3 pathway, and this could provide novel insights into HCV-induced HCC.
Collapse
|
35
|
Inflammatory transcription factors as activation markers and functional readouts in immune-to-brain communication. Brain Behav Immun 2016; 54:1-14. [PMID: 26348582 DOI: 10.1016/j.bbi.2015.09.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 08/31/2015] [Accepted: 09/04/2015] [Indexed: 02/06/2023] Open
Abstract
Immune-to-brain communication pathways involve humoral mediators, including cytokines, central modulation by neuronal afferents and immune cell trafficking to the brain. During systemic inflammation these pathways contribute to mediating brain-controlled sickness symptoms including fever. Experimentally, activation of these signaling pathways can be mimicked and studied when injecting animals with pathogen associated molecular patterns (PAMPS). One central component of the brain inflammatory response, which leads, for example, to fever induction, is transcriptional activation of brain cells via cytokines and PAMPS. We and others have studied the spatiotemporal activation and the physiological significance of transcription factors for the induction of inflammation within the brain and the manifestation of fever. Evidence has revealed a role of nuclear factor (NF)κB in the initiation, signal transducer and activator of transcription (STAT)3 in the maintenance and NF-interleukin (IL)6 in the maintenance or even termination of brain-inflammation and fever. Moreover, psychological stressors, such as exposure to a novel environment, leads to increased body core temperature and genomic NF-IL6-activation, suggesting a potential use of NF-IL6-immunohistochemistry as a multimodal brain cell activation marker and a role for NF-IL6 for differential brain activity. In addition, the nutritional status, as reflected by circulating levels of the cytokine-like hormone leptin, influence immune-to-brain communication and age-dependent changes in LPS-induced fever. Overall, transcription factors remain therapeutically important targets for the treatment of brain-inflammation and fever induction during infectious/non-infectious inflammatory and psychological stress. However, the exact physiological role and significance of these transcription factors requires to be further investigated.
Collapse
|
36
|
HCV RNA traffic and association with NS5A in living cells. Virology 2016; 493:60-74. [PMID: 26999027 DOI: 10.1016/j.virol.2016.02.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/11/2016] [Accepted: 02/18/2016] [Indexed: 01/05/2023]
Abstract
The spatiotemporal dynamics of Hepatitis C Virus (HCV) RNA localisation are poorly understood. To address this we engineered HCV genomes harbouring MS2 bacteriophage RNA stem-loops within the 3'-untranslated region to allow tracking of HCV RNA via specific interaction with a MS2-Coat-mCherry fusion protein. Despite the impact of these insertions on viral fitness, live imaging revealed that replication of tagged-HCV genomes induced specific redistribution of the mCherry-tagged-MS2-Coat protein to motile and static foci. Further analysis showed that HCV RNA was associated with NS5A in both static and motile structures while a subset of motile NS5A structures was devoid of HCV RNA. Further investigation of viral RNA traffic with respect to lipid droplets (LDs) revealed HCV RNA-positive structures in close association with LDs. These studies provide new insights into the dynamics of HCV RNA traffic with NS5A and LDs and provide a platform for future investigations of HCV replication and assembly.
Collapse
|
37
|
Li M, Wang W, Jin R, Zhang T, Li N, Han Q, Wei P, Liu Z. Differential association of STAT3 and HK-II expression in hepatitis B virus- and hepatitis C virus-related hepatocellular carcinoma. J Med Virol 2016; 88:1552-9. [PMID: 26889748 DOI: 10.1002/jmv.24498] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2016] [Indexed: 12/14/2022]
Abstract
STAT3 and hexokinase II (HK-II) are involved in viral infection and carcinogenesis of various cancers including hepatocellular carcinoma (HCC). The roles of STAT3 and HK-II in hepatitis B virus (HBV)- and hepatitis C virus (HCV)-related HCC remain largely unclear. This study examined STAT3 and HK-II expression in HBV- and HCV-related HCC, HBV-related liver fibrosis, and normal control liver by using tissue microarray and immunohistochemical method. Results showed that STAT3 expression in HBV-related HCC, HCV-related HCC, and HBV-related liver fibrosis was significantly higher than in control liver (P < 0.001, P = 0.016, and P = 0.005, respectively) and had no significant differences between these three diseased liver tissues. The HK-II expression in HBV-related HCC was significantly higher than that in HCV-related HCC, HBV-related liver fibrosis, and control liver (P = 0.007, P = 0.029, and P = 0.008, respectively) but had no significant elevation in and no significant differences between HCV-related HCC, HBV-related liver fibrosis, and control liver. The HK-II expression was significantly correlated to STAT3 expression in HBV-related HCC (P = 0.022), but no correlation was observed in HCV-related HCC, HBV-related liver fibrosis, and control liver. In conclusion, STAT3 expression is upregulated in both HBV- and HCV-related HCC, while HK-II is predominantly upregulated and correlated to STAT3 in HBV-related HCC. These differential expression and association may suggest the distinct roles of STAT3 and HK-II in hepatocarcinogenesis of HBV and HCV infection. Studies are needed to confirm the relationship of STAT3 and HK-II and to examine the underlying mechanisms. J. Med. Virol. 88:1552-1559, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Man Li
- Department of Infectious Diseases, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.,Department of Internal Medicine, The Second Hospital of Xi'an, Xi'an, Shaanxi, People's Republic of China
| | - Weihua Wang
- Department of Pharmacogenomics, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Rui Jin
- Department of Radiology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Tieying Zhang
- Department of Internal Medicine, The Second Hospital of Xi'an, Xi'an, Shaanxi, People's Republic of China
| | - Na Li
- Department of Infectious Diseases, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Qunying Han
- Department of Infectious Diseases, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Ping Wei
- Department of Infectious Diseases, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Zhengwen Liu
- Department of Infectious Diseases, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
38
|
Interferon alpha antagonizes STAT3 and SOCS3 signaling triggered by hepatitis C virus. Cytokine 2016; 80:48-55. [PMID: 26945996 DOI: 10.1016/j.cyto.2015.08.264] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/26/2015] [Accepted: 08/27/2015] [Indexed: 12/24/2022]
Abstract
We aimed to investigate regulation of signal transducer and activator of transcription 3 (STAT3) and suppressor of cytokine signaling 3 (SOCS3) by interferon alpha (IFN-α) and to analyze the relationship between STAT3 and SOCS3 during hepatitis C virus (HCV) infection. Changes in STAT3 and SOCS3 were analyzed at both mRNA and protein levels in human hepatoma cells infected with HCV (J6/JFH1). At 72h of HCV infection, STAT3 expression was decreased with sustained phosphorylation, and IFN-α increased such decrease and phosphorylation. HCV increased SOCS3 expression, while IFN-α impaired such increase, indicating different regulation of STAT3 and SOCS3 by IFN-α. IFN-α-induced expression and phosphorylation of upstream kinases of the JAK/STAT pathway, Tyk2 and Jak1, were suppressed by HCV. Moreover, knockdown of STAT3 by RNA interference led to decreases in HCV RNA replication and viral protein expression, without affecting either the expression of Tyk2 and Jak1 or the SOCS3 induction in response to IFN-α. These results show that IFN-α antagonizes STAT3 and SOCS3 signaling triggered by HCV and that STAT3 regulation correlates inversely with SOCS3 induction by IFN-α, which may be important in better understanding the complex interplay between IFN-α and signal molecules during HCV infection.
Collapse
|
39
|
Host-Targeting Agents to Prevent and Cure Hepatitis C Virus Infection. Viruses 2015; 7:5659-85. [PMID: 26540069 PMCID: PMC4664971 DOI: 10.3390/v7112898] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 09/25/2015] [Accepted: 10/19/2015] [Indexed: 12/13/2022] Open
Abstract
Chronic hepatitis C virus (HCV) infection is a major cause of liver cirrhosis and hepatocellular carcinoma (HCC) which are leading indications of liver transplantation (LT). To date, there is no vaccine to prevent HCV infection and LT is invariably followed by infection of the liver graft. Within the past years, direct-acting antivirals (DAAs) have had a major impact on the management of chronic hepatitis C, which has become a curable disease in the majority of DAA-treated patients. In contrast to DAAs that target viral proteins, host-targeting agents (HTAs) interfere with cellular factors involved in the viral life cycle. By acting through a complementary mechanism of action and by exhibiting a generally higher barrier to resistance, HTAs offer a prospective option to prevent and treat viral resistance. Indeed, given their complementary mechanism of action, HTAs and DAAs can act in a synergistic manner to reduce viral loads. This review summarizes the different classes of HTAs against HCV infection that are in preclinical or clinical development and highlights their potential to prevent HCV infection, e.g., following LT, and to tailor combination treatments to cure chronic HCV infection.
Collapse
|
40
|
Narayana SK, Helbig KJ, McCartney EM, Eyre NS, Bull RA, Eltahla A, Lloyd AR, Beard MR. The Interferon-induced Transmembrane Proteins, IFITM1, IFITM2, and IFITM3 Inhibit Hepatitis C Virus Entry. J Biol Chem 2015; 290:25946-59. [PMID: 26354436 DOI: 10.1074/jbc.m115.657346] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Indexed: 01/15/2023] Open
Abstract
The interferon-induced transmembrane (IFITM) family of proteins have recently been identified as important host effector molecules of the type I interferon response against viruses. IFITM1 has been identified as a potent antiviral effector against hepatitis C virus (HCV), whereas the related family members IFITM2 and IFITM3 have been described to have antiviral effects against a broad range of RNA viruses. Here, we demonstrate that IFITM2 and IFITM3 play an integral role in the interferon response against HCV and act at the level of late entry stages of HCV infection. We have established that in hepatocytes, IFITM2 and IFITM3 localize to the late and early endosomes, respectively, as well as the lysosome. Furthermore, we have demonstrated that S-palmitoylation of all three IFITM proteins is essential for anti-HCV activity, whereas the conserved tyrosine residue in the N-terminal domain of IFITM2 and IFITM3 plays a significant role in protein localization. However, this tyrosine was found to be dispensable for anti-HCV activity, with mutation of the tyrosine resulting in an IFITM1-like phenotype with the retention of anti-HCV activity and co-localization of IFITM2 and IFITM3 with CD81. In conclusion, we propose that the IFITM proteins act in a coordinated manner to restrict HCV infection by targeting the endocytosed HCV virion for lysosomal degradation and demonstrate that the actions of the IFITM proteins are indeed virus and cell-type specific.
Collapse
Affiliation(s)
- Sumudu K Narayana
- From the School of Biological Sciences, and the Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia 5005, Australia, the Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, 5000, Australia, and
| | - Karla J Helbig
- From the School of Biological Sciences, and the Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia 5005, Australia, the Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, 5000, Australia, and
| | - Erin M McCartney
- From the School of Biological Sciences, and the Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia 5005, Australia, the Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, 5000, Australia, and
| | - Nicholas S Eyre
- From the School of Biological Sciences, and the Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia 5005, Australia, the Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, 5000, Australia, and
| | - Rowena A Bull
- the Inflammation and Infection Research Centre, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Auda Eltahla
- the Inflammation and Infection Research Centre, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Andrew R Lloyd
- the Inflammation and Infection Research Centre, School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Michael R Beard
- From the School of Biological Sciences, and the Research Centre for Infectious Diseases, University of Adelaide, Adelaide, South Australia 5005, Australia, the Centre for Cancer Biology, SA Pathology, Adelaide, South Australia, 5000, Australia, and
| |
Collapse
|
41
|
Xiong Y, Jia M, Yuan J, Zhang C, Zhu Y, Kuang X, Lan L, Wang X. STAT3‑regulated long non‑coding RNAs lnc‑7SK and lnc‑IGF2‑AS promote hepatitis C virus replication. Mol Med Rep 2015; 12:6738-44. [PMID: 26328522 PMCID: PMC4626162 DOI: 10.3892/mmr.2015.4278] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 07/31/2015] [Indexed: 12/16/2022] Open
Abstract
Long non‑coding RNAs (lncRNAs) are a class of RNAs that do not code protein but are important in diverse biological processes. In previous years, with the application of high‑throughput sequencing, a large number of lncRNAs associated with virus infections have been identified and intensively investigated, however, there are few studies examining the association between lncRNAs and HCV replication. Previous studies have demonstrated that signal transducer and activator of transcription 3 (STAT3) is activated by the hepatitis C virus (HCV) and in turn increases the replication of HCV. However, the detailed molecular mechanism is only partially understood. In the present study, using human lncRNA polymerase chain reaction (PCR) arrays, it was identified that lnc‑IGF2‑AS, lnc‑7SK, lnc‑SChLAP1 and lnc‑SRA1 are upregulated by STAT3. In addition, among these four lncRNAs, only lnc‑IGF2‑AS and lnc‑7SK were involved in HCV replication. Transfection of siRNA lnc‑7SK and siRNA lnc‑IGF2‑AS partially inhibited the replication of HCV in Huh7 cells. Data also indicated that when transfected with siRNA lnc‑7SK and siRNA lnc‑IGF2‑AS, the expression of phosphatidylinositol 4‑phosphate (PI4P), which was identified to be associated with HCV replication, was reduced. Thus, the present study identified two new types of lncRNAs, lnc‑IGF2‑AS and lnc‑7SK, which can be upregulated by STAT3 and are involved in HCV replication by regulating PI4P.
Collapse
Affiliation(s)
- Yulin Xiong
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Ming Jia
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Jing Yuan
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Changjiang Zhang
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Yan Zhu
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Xuemei Kuang
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Lin Lan
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Xiaohong Wang
- Key Laboratory of Infectious Disease Research, Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
42
|
Xiong Y, Yuan J, Zhang C, Zhu Y, Kuang X, Lan L, Wang X. The STAT3-regulated long non-coding RNA Lethe promote the HCV replication. Biomed Pharmacother 2015; 72:165-71. [PMID: 26054691 DOI: 10.1016/j.biopha.2015.04.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 04/27/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are a class of RNAs that do not code protein but play important roles in diverse biological processes. In recent years, with the application of high-throughput sequencing, a great deal of lncRNAs associated with virus infections have been discovered and intensively studied, but there are few studies about the relationship between lncRNAs and HCV replication. METHODS We identify that several lncRNAs can be upregulated and downregulated by phosphorylated STAT3 by using human PCR array method. And among these lncRNAs, lnc-Lethe was involved in the HCV replication. Transfection of siRNA Lethe partially blocked the replication of HCV in Huh7 cells. RESULTS In the present study, we have established that phosphorylated STAT3 can promote the HCV replication. Data also indicated that when transfected with siRNA Lethe, the expression levels of PKR, OAS and IRF1, which were all ISGs, were all up regulated. CONCLUSIONS Based on our findings from Lethe knockdown, we have identified that Lethe, which was upregulated by activated STAT3, may promoting the replication of HCV through a negative regulatory mechanism of type I IFN response.
Collapse
Affiliation(s)
- Yulin Xiong
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China
| | - Jing Yuan
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China
| | - Changjiang Zhang
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China
| | - Yan Zhu
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China
| | - Xuemei Kuang
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China
| | - Lin Lan
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China
| | - Xiaohong Wang
- Institute of Infectious Diseases of Chinese PLA, Southwest Hospital, Third Military Medical University, Gaotanyan Centre Street No. 30, Shapingba district, Chongqing 400038, China.
| |
Collapse
|
43
|
Niu Y, Si Y, Li Y, Chi X, Li X, Liu X, Li D, Cheng M, Fan J, Si S, Yang W. A novel small-molecule inhibitor of hepatitis C virus replication acts by suppressing signal transducer and activator of transcription 3. J Antimicrob Chemother 2015; 70:2013-23. [PMID: 25858355 DOI: 10.1093/jac/dkv077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 03/05/2015] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVES Hepatitis C virus (HCV) infects hepatocytes and causes liver damage. The aim of this study was to identify new classes of host-targeting anti-HCV compounds that may provide novel approaches for antiviral treatment regimens. METHODS Cell culture-derived HCV (HCVcc), replicons and pseudoparticles were used in combination with high-throughput screening, reporter gene assays and cytotoxicity and signalling pathway analyses. RESULTS A small-molecule inhibitor of HCV, N-(cyclopropyl(phenyl)methyl)thieno[2,3-d]pyrimidin-4-amine, designated IB-32, was identified by screening a compound library with a Jc1-luc HCVcc assay. By using various virus models, HCV replication was identified as the predominant step of IB-32's action. IB-32 inhibited HCVcc (genotype 2a) and HCV replicons (genotype 1b) at low nanomolar ranges (with IC50s of 40 ± 8 and 100 ± 15 nM, respectively). IB-32 was found to be non-toxic when tested against a panel of human cell lines in vitro at the effective antiviral dose. Mechanistically, IB-32 strongly inhibited STAT3 (Tyr705) phosphorylation, a necessary cellular factor for HCV replication and a pivotal therapeutic target for multiple cancers. Furthermore, the inhibition of HCV replication by IB-32 was augmented in cells with STAT3 knockdown. In contrast, the inhibitory effect of IB-32 was attenuated in cells overexpressing a constitutively active form of STAT3. CONCLUSION The results presented here identify a promising STAT3-targeting anti-HCV therapeutic candidate. This novel small molecule could be further optimized and developed for use as both an antiviral and an anti-cancer drug.
Collapse
Affiliation(s)
- Yuqiang Niu
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Youhui Si
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yan Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaojing Chi
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiang Li
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiuying Liu
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Duan Li
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Min Cheng
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jingjing Fan
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuyi Si
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Yang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Cellular stress responses in hepatitis C virus infection: Mastering a two-edged sword. Virus Res 2015; 209:100-17. [PMID: 25836277 DOI: 10.1016/j.virusres.2015.03.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 03/21/2015] [Accepted: 03/23/2015] [Indexed: 12/11/2022]
Abstract
Hepatitis C virus (HCV) infection affects chronically more than 150 million humans worldwide. Chronic HCV infection causes severe liver disease and hepatocellular carcinoma. While immune response-mediated events are major players in HCV pathogenesis, the impact that viral replication has on cellular homeostasis is increasingly recognized as a necessary contributor to pathological manifestations of HCV infection such as steatosis, insulin-resistance or liver cancer. In this review, we will briefly overview the different cellular stress pathways that are induced by hepatitis C virus infection, the response that the cell promotes to attempt regaining homeostasis or to induce dysfunctional cell death, and how the virus co-opts these response mechanisms to promote both viral replication and survival of the infected cell. We will review the role of unfolded protein and oxidative stress responses as well as the role of auto- and mitophagy in HCV infection. Finally, we will discuss the recent discovery of a cellular chaperone involved in stress responses, the sigma-1 receptor, as a cellular factor required at the onset of HCV infection and the potential molecular events underlying the proviral role of this cellular factor in HCV infection.
Collapse
|
45
|
Huang Y, Huang X, Yang Y, Wang W, Yu Y, Qin Q. Involvement of fish signal transducer and activator of transcription 3 (STAT3) in nodavirus infection induced cell death. FISH & SHELLFISH IMMUNOLOGY 2015; 43:241-248. [PMID: 25555814 DOI: 10.1016/j.fsi.2014.12.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 06/04/2023]
Abstract
The Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway is an important signaling pathway activated by interferons in response to virus infection. Fish STAT3 has been demonstrated to be involved in Singapore grouper iridovirus (SGIV) infection and virus induced paraptosis, but its effects on the replication of other fish viruses still remained uncertain. Here, the roles of grouper STAT3 (Ec-STAT3) in red spotted grouper nervous necrosis virus (RGNNV) infection were investigated. The present data showed that the distribution of phosphorylated Ec-STAT3 was altered in RGNNV infected fish cells, and the promoter activity of STAT3 was significantly increased during virus infection, suggesting that STAT3 activation was involved in RGNNV infection. Using STAT3 specific inhibitor, we found that inhibition of Ec-STAT3 in vitro did not affect the transcription and protein synthesis of RGNNV coat protein (CP), however, the severity of RGNNV induced vacuolation and autophagy was significantly increased. Meanwhile, at the late stage of virus infection, RGNNV induced necrotic cell death was significantly decreased after inhibition of Ec-STAT3. Further studies indicated that Ec-STAT3 inhibition significantly increased the transcript level of autophagy related genes, including UNC-51-like kinase 2 (ULK2) and microtubule-associated protein 1 light chain 3-II (LC3-II) induced by RGNNV infection. Moreover, the expression of several pro-inflammatory factors, including TNFα, IL-1β and IL-8 were mediated by Ec-STAT3 during RGNNV infection. Together, our results not only firstly revealed that STAT3 exerted novel roles in response to fish virus infection, but also provided new insights into understanding the roles of STAT3 in different forms of programmed cell death.
Collapse
Affiliation(s)
- Youhua Huang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China; University of Chinese Academy of Sciences, Beijing, PR China
| | - Xiaohong Huang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China; University of Chinese Academy of Sciences, Beijing, PR China
| | - Ying Yang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China; University of Chinese Academy of Sciences, Beijing, PR China
| | - Wei Wang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China; University of Chinese Academy of Sciences, Beijing, PR China
| | - Yepin Yu
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China; University of Chinese Academy of Sciences, Beijing, PR China
| | - Qiwei Qin
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China; University of Chinese Academy of Sciences, Beijing, PR China.
| |
Collapse
|
46
|
Jiang X, Kanda T, Nakamoto S, Saito K, Nakamura M, Wu S, Haga Y, Sasaki R, Sakamoto N, Shirasawa H, Okamoto H, Yokosuka O. The JAK2 inhibitor AZD1480 inhibits hepatitis A virus replication in Huh7 cells. Biochem Biophys Res Commun 2015; 458:908-12. [PMID: 25704089 DOI: 10.1016/j.bbrc.2015.02.058] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/11/2015] [Indexed: 12/18/2022]
Abstract
The JAK2 inhibitor AZD1480 has been reported to inhibit La protein expression. We previously demonstrated that the inhibition of La expression could inhibit hepatitis A virus (HAV) internal ribosomal entry-site (IRES)-mediated translation and HAV replication in vitro. In this study, we analyzed the effects of AZD1480 on HAV IRES-mediated translation and replication. HAV IRES-mediated translation in COS7-HAV-IRES cells was inhibited by 0.1-1 μM AZD1480, a dosage that did not affect cell viability. Results showed a significant reduction in intracellular HAV HA11-1299 genotype IIIA RNA levels in Huh7 cells treated with AZD1480. Furthermore, AZD1480 inhibited the expression of phosphorylated-(Tyr-705)-signal transducer and activator of transcription 3 (STAT3) and La in Huh7 cells. Therefore, we propose that AZD1480 can inhibit HAV IRES activity and HAV replication through the inhibition of the La protein.
Collapse
Affiliation(s)
- Xia Jiang
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Tatsuo Kanda
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.
| | - Shingo Nakamoto
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan; Department of Molecular Virology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Kengo Saito
- Department of Molecular Virology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Masato Nakamura
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Shuang Wu
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yuki Haga
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Reina Sasaki
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Hokkaido University, Sapporo, Japan
| | - Hiroshi Shirasawa
- Department of Molecular Virology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Hiroaki Okamoto
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, Shimotsuke, Japan
| | - Osamu Yokosuka
- Department of Gastroenterology and Nephrology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| |
Collapse
|
47
|
Yan B, Xie S, Liu Z, Luo Y, Zhou J, Li D, Liu M. STAT3 association with microtubules and its activation are independent of HDAC6 activity. DNA Cell Biol 2015; 34:290-5. [PMID: 25621430 DOI: 10.1089/dna.2014.2713] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is an important oncogenic transcription factor residing in the cytoplasm in the resting cells. Upon stimulation, STAT3 is activated and translocated to the nucleus to regulate target genes. Although the canonical transcriptional function of STAT3 has been intensively studied, less is known about its cytoplasmic localization. In this study, by immunoprecipitation, microtubule cosedimentation, and immunofluorescence assays, we present the first evidence that cytoplasmic STAT3 interacts with both tubulin and microtubules. By using small-molecule inhibitor approaches, we further demonstrate that the localization of STAT3 on microtubules and its activation are independent of histone deacetylase 6 (HDAC6) activity. In addition, disruption of microtubule dynamics does not alter the activation and nuclear translocation of STAT3 in response to interleukin-6 treatment. These findings reveal that cytoplasmic STAT3 is physically associated with microtubules, whereas its activation and nuclear translocation are independent of microtubule dynamics, implicating that the association of STAT3 with microtubules might be involved in the regulation of noncanonical functions of STAT3 in the cytoplasm.
Collapse
Affiliation(s)
- Bing Yan
- 1 State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University , Tianjin, China
| | | | | | | | | | | | | |
Collapse
|
48
|
Huang X, Huang Y, Yang Y, Wei S, Qin Q. Involvement of fish signal transducer and activator of transcription 3 (STAT3) in SGIV replication and virus induced paraptosis. FISH & SHELLFISH IMMUNOLOGY 2014; 41:308-316. [PMID: 25230133 DOI: 10.1016/j.fsi.2014.09.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/03/2014] [Accepted: 09/05/2014] [Indexed: 06/03/2023]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is an important transcription factor which plays crucial roles in immune regulation, inflammation, cell proliferation, transformation, and other physiological processes of the organism. In this study, a novel STAT3 gene from orange spotted grouper (Ec-STAT3) was cloned and characterized. Bioinformatic analysis revealed that full-length of Ec-STAT3 was 3105-bp long and contained a 280-bp 5'UTR, a 470-bp 3'UTR, and a 2355-bp open reading frame (ORF) that encoded a 784-amino acid peptide. The deduced protein of Ec-STAT3 showed 98% identity to that of turbot (Scophthalmus maximus). Amino acid alignment showed that Ec-STAT3 contained four conserved domains, including a protein interaction domain, a coiled coil domain, a DNA binding domain, and an SH2 domain. Quantitative real-time PCR analysis showed that the highest expression level was detected in the liver, followed by skin and spleen. After injection with Singapore grouper iridovirus (SGIV), the transcript of Ec-STAT3 in spleen was increased significantly. To further explore the function of Ec-STAT3, we investigated the roles of Ec-STAT3 in SGIV infection in vitro. Immune fluorescence analysis indicated that SGIV infection altered the distribution of phosphorylated Ec-STAT3 in nucleus, and a small part of phosphorylated Ec-STAT3 was associated with virus assembly sites, suggesting that Ec-STAT3 might be important for SGIV infection. Using STAT3 specific inhibitor, S3I-201, we found that inhibition of Ec-STAT3 activation decreased the SGIV replication significantly. Moreover, inhibition of Ec-STAT3 activation obviously altered SGIV infection induced cell cycle arrest and the expression of pro-survival genes, including Bcl-2, Bcl-xL and Bax inhibitor. Together, our results firstly demonstrated the critical roles of fish STAT3 in DNA virus replication and virus induced paraptosis, but also provided new insights into the mechanism of iridovirus pathogenesis.
Collapse
Affiliation(s)
- Xiaohong Huang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China; University of Chinese Academy of Sciences, Beijing, PR China
| | - Youhua Huang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China; University of Chinese Academy of Sciences, Beijing, PR China
| | - Ying Yang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China; University of Chinese Academy of Sciences, Beijing, PR China
| | - Shina Wei
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China; University of Chinese Academy of Sciences, Beijing, PR China
| | - Qiwei Qin
- Key Laboratory of Tropical Marine Bio-resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, PR China; University of Chinese Academy of Sciences, Beijing, PR China.
| |
Collapse
|
49
|
miR-223 inhibits dengue virus replication by negatively regulating the microtubule-destabilizing protein STMN1 in EAhy926 cells. Microbes Infect 2014; 16:911-22. [PMID: 25181337 PMCID: PMC7110837 DOI: 10.1016/j.micinf.2014.08.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 08/15/2014] [Accepted: 08/25/2014] [Indexed: 12/23/2022]
Abstract
The pathogenesis of dengue virus (DENV) infection is not completely understood. Endothelial cells may act as a target of the virus and be involved in disease pathogenesis. Therefore, the identification of host cell components involved in DENV replication would provide useful information for better understanding DENV infection. In this study, a significantly decreased level of miR-223 was found in DENV2-infected EAhy926 cells, a human endothelial-like cell line, whereas miR-223 overexpression inhibited DENV2 replication. Furthermore, we identified that miR-223 directly targeted the 3' untranslated region (3'UTR) of the messenger RNA (mRNA) for microtubule-destabilizing protein stathmin 1 (STMN1), thereby reducing its mRNA and protein levels. The depletion of miR-223 or overexpression of STMN1 enhanced DENV2 replication, whereas the opposite (increased miR-223 or decreased STMN1) suppressed DENV2 replication, indicating that miR-223 down-regulates STMN1 expression by targeting the 3'UTR of the STMN1 gene to inhibit DENV2 replication. Finally, we demonstrated that two transcription factors, C/EBPα and E2F1, are involved in the regulation of miR-223 levels after DENV2 infection in EAhy926 cells. Collectively, our results suggest that miR-223 may act as a novel antiviral factor, which may open an avenue to limit DENV infection.
Collapse
|
50
|
Human herpesvirus 8 viral interleukin-6 signaling through gp130 promotes virus replication in primary effusion lymphoma and endothelial cells. J Virol 2014; 88:12167-72. [PMID: 25078695 DOI: 10.1128/jvi.01751-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The contributions of human herpesvirus 8 (HHV-8) viral interleukin-6 (vIL-6) to virus biology remain unclear. Here we examined the role of vIL-6/gp130 signaling in HHV-8 productive replication in primary effusion lymphoma and endothelial cells. Depletion and depletion-complementation experiments revealed that endoplasmic reticulum-localized vIL-6 activity via gp130 and gp130-activated signal transducer and activator of transcription (STAT) signaling, but not extracellular signal-regulated kinase (ERK) activation, was critical for vIL-6 proreplication activity. Our data significantly extend current understanding of vIL-6 function and associated mechanisms in HHV-8 biology.
Collapse
|