51
|
Girardi E, López P, Pfeffer S. On the Importance of Host MicroRNAs During Viral Infection. Front Genet 2018; 9:439. [PMID: 30333857 PMCID: PMC6176045 DOI: 10.3389/fgene.2018.00439] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/14/2018] [Indexed: 12/21/2022] Open
Abstract
Every living organism has to constantly face threats from the environment and deal with a large number of pathogens against which it has to defend itself to survive. Among those, viruses represent a large class of obligatory intracellular parasites, which rely on their host machinery to multiply and propagate. As a result, viruses and their hosts have engaged in an ever-evolving arms race to be able to maintain their existence. The role played by micro (mi)RNAs in this ongoing battle has been extensively studied in the past 15 years and will be the subject of this review article. We will mainly focus on cellular miRNAs and their implication during viral infection in mammals. Thus, we will describe current techniques that can be used to identify miRNAs involved in the modulation of viral infection and to characterize their targets and mode of action. We will also present different reported examples of miRNA-mediated regulation of viruses, which can have a positive outcome either for the host or for the virus. In addition, the mode of action is also of a dual nature, depending on the target of the miRNA. Indeed, the regulatory small RNA can either directly guide an Argonaute protein on a viral transcript, or target a cellular mRNA involved in the host antiviral response. We will then see whether and how viruses respond to miRNA-mediated targeting. Finally, we will discuss how our knowledge of viral targeting by miRNA can be exploited for developing new antiviral therapeutic approaches.
Collapse
Affiliation(s)
- Erika Girardi
- Architecture and Reactivity of RNA, Institut de Biologie Moléculaire et Cellulaire du CNRS, Université de Strasbourg, Strasbourg, France
| | - Paula López
- Architecture and Reactivity of RNA, Institut de Biologie Moléculaire et Cellulaire du CNRS, Université de Strasbourg, Strasbourg, France
| | - Sébastien Pfeffer
- Architecture and Reactivity of RNA, Institut de Biologie Moléculaire et Cellulaire du CNRS, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
52
|
Zhao Q, Xiong Y, Xu J, Chen S, Li P, Huang Y, Wang Y, Chen WX, Wang B. Host MicroRNA hsa-miR-494-3p Promotes EV71 Replication by Directly Targeting PTEN. Front Cell Infect Microbiol 2018; 8:278. [PMID: 30234021 PMCID: PMC6130220 DOI: 10.3389/fcimb.2018.00278] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 07/25/2018] [Indexed: 11/13/2022] Open
Abstract
Many cellular processes are driven by spatially and temporally regulated microRNAs (miRNAs)-dependent signaling events. Substantial evidence collected over the years indicates that miRNAs are pivotal regulators that contribute to the initiation and development of EV71-related disorders. Importantly, so far, no clinical trial has been undertaken to address the effect of miRNAs on EV71-related diseases. In this study, we show that EV71 infection results in up-regulation of hsa-miR-494-3p levels, and that EV71-induced hsa-miR-494-3p impacts PI3K/Akt signaling pathway by targeting PTEN. However, very little is known about the relationship between hsa-miR-494-3p and EV71 infection. The overall goal of the study is to get a better insight into whether or not hsa-miR-494-3p is involved in the EV71 infection. We found that the EV71 infection induces cellular apoptosis, and that this process can be counteracted by the over-expression of hsa-miR-494-3p mimics. We also present evidence that cell lines deficient in hsa-miR-494-3p are more sensitive to EV71-induced cell death than the corresponding control cells. Collectively, these findings confirm and extend the pervious observation suggesting that disturbances in miRNAs expression can influence EV71 propagation. In addition, they lend strong support to the ideas that hsa-miR-494-3p-mediated signaling pathway plays an important role in the EV71 replication, and that this may have profound implications on our views on EV71-related diseases.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuan Xiong
- Department of Laboratory Medicine, Chongqing Health Center for Women and Children, Chongqing, China
| | - Jingru Xu
- Institute of Microbiology, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Shuang Chen
- Institute of Microbiology, Chongqing Center for Disease Control and Prevention, Chongqing, China
| | - Pu Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunying Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei-Xian Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bo Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
53
|
Li B, Zheng J. MicroR-9-5p suppresses EV71 replication through targeting NFκB of the RIG-I-mediated innate immune response. FEBS Open Bio 2018; 8:1457-1470. [PMID: 30186747 PMCID: PMC6120239 DOI: 10.1002/2211-5463.12490] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022] Open
Abstract
Accumulating evidence demonstrates that there is a causative link between hsa-microRNA-9-5p (miR-9) and pathophysiological processes. Enterovirus 71 (EV71) has been found to contribute to numerous severe clinical symptoms which result in death. The exact mechanism by which EV71 influences miR-9 expression is unknown, and the relationship between miR-9 and EV71 is still unclear. Here, miR-9 expression was found to be impaired upon EV71 infection in several cell lines and in an EV71 infection mouse model. Additionally, we confirmed that EV71 infection induces robust expression of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1) and interferons (IFN-α and IFN-β). Overexpression of miR-9 attenuated EV71 proliferation and reduced protein and gene expressions of virion protein 1 (VP1) of EV71. Furthermore, we observed that the inflammation caused by EV71 infection was restored to a moderate level via miR-9 overexpression. Nuclear factor kappa B (NFκB) in the retinoic acid-induced gene 1 (RIG-I) signaling pathway, but not interferon regulating factor 3 (IRF3), was significantly decreased and inactivated by ectopic miR-9 expression. Moreover, in mouse infection experiments, administration of miR-9 agomirs caused a significant decrease in VP1 levels and pro-inflammatory cytokine production after viral inoculation. Taken together, the present data demonstrate that miR-9 exerts an anti-EV71 effect in cells and a mouse model via mediating NFκB activity of the RIG-I signal pathway, thereby suggesting a new candidate for antiviral drug development.
Collapse
Affiliation(s)
- Bing Li
- Department of Pediatrics Jinan Maternity and Child Care Hospital China
| | - Junqing Zheng
- Department of Pediatrics Jinan Maternity and Child Care Hospital China
| |
Collapse
|
54
|
Abstract
During the last years, it has become evident that miRNAs are important players in almost all physiological and pathological processes, including viral infections. Enterovirus infections range from mild to severe acute infections concerning several organ systems and are also associated with chronic diseases. In this review, we summarize the findings on the impact of acute and persistent enterovirus infection on the expression of cellular miRNAs. Furthermore, the currently available data on the regulation of cellular or viral targets by the dysregulated miRNAs are reviewed. Finally, a translational perspective, namely the use of miRNAs as biomarkers of enterovirus infection and as antiviral strategy is discussed.
Collapse
Affiliation(s)
- Ilka Engelmann
- a Laboratoire de Virologie EA3610, Faculté de Médecine, CHU Lille, University of Lille , Lille , France
| | - Enagnon Kazali Alidjinou
- a Laboratoire de Virologie EA3610, Faculté de Médecine, CHU Lille, University of Lille , Lille , France
| | - Antoine Bertin
- a Laboratoire de Virologie EA3610, Faculté de Médecine, CHU Lille, University of Lille , Lille , France
| | - Famara Sane
- a Laboratoire de Virologie EA3610, Faculté de Médecine, CHU Lille, University of Lille , Lille , France
| | - Didier Hober
- a Laboratoire de Virologie EA3610, Faculté de Médecine, CHU Lille, University of Lille , Lille , France
| |
Collapse
|
55
|
Bourhill T, Mori Y, Rancourt DE, Shmulevitz M, Johnston RN. Going (Reo)Viral: Factors Promoting Successful Reoviral Oncolytic Infection. Viruses 2018; 10:E421. [PMID: 30103501 PMCID: PMC6116061 DOI: 10.3390/v10080421] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/09/2018] [Accepted: 08/09/2018] [Indexed: 02/06/2023] Open
Abstract
Oncolytic viruses show intriguing potential as cancer therapeutic agents. These viruses are capable of selectively targeting and killing cancerous cells while leaving healthy cells largely unaffected. The use of oncolytic viruses for cancer treatments in selected circumstances has recently been approved by the Food and Drug Administration (FDA) of the US and work is progressing on engineering viral vectors for enhanced selectivity, efficacy and safety. However, a better fundamental understanding of tumour and viral biology is essential for the continued advancement of the oncolytic field. This knowledge will not only help to engineer more potent and effective viruses but may also contribute to the identification of biomarkers that can determine which patients will benefit most from this treatment. A mechanistic understanding of the overlapping activity of viral and standard chemotherapeutics will enable the development of better combinational approaches to improve patient outcomes. In this review, we will examine each of the factors that contribute to productive viral infections in cancerous cells versus healthy cells. Special attention will be paid to reovirus as it is a well-studied virus and the only wild-type virus to have received orphan drug designation by the FDA. Although considerable insight into reoviral biology exists, there remain numerous deficiencies in our understanding of the factors regulating its successful oncolytic infection. Here we will discuss what is known to regulate infection as well as speculate about potential new mechanisms that may enhance successful replication. A joint appreciation of both tumour and viral biology will drive innovation for the next generation of reoviral mediated oncolytic therapy.
Collapse
Affiliation(s)
- Tarryn Bourhill
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
| | - Yoshinori Mori
- Department of Gastroenterology, Nagoya City West Medical Center, Kita-Ku, Nagoya 467-8601, Japan.
| | - Derrick E Rancourt
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
| | - Maya Shmulevitz
- Department of Medical Microbiology and Immunology, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB T6G 2E1, Canada.
| | - Randal N Johnston
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
56
|
Xia B, Lu J, Wang R, Yang Z, Zhou X, Huang P. miR-21-3p Regulates Influenza A Virus Replication by Targeting Histone Deacetylase-8. Front Cell Infect Microbiol 2018; 8:175. [PMID: 29888214 PMCID: PMC5981164 DOI: 10.3389/fcimb.2018.00175] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/08/2018] [Indexed: 12/20/2022] Open
Abstract
Influenza A virus (IAV) is responsible for severe morbidity and mortality in animals and humans worldwide. miRNAs are a class of small noncoding single-stranded RNA molecules that can negatively regulate gene expression and play important roles in virus-host interaction. However, the roles of miRNAs in IAV infection are still not fully understood. Here, we profiled the cellular miRNAs of A549 cells infected with A/goose/Jilin/hb/2003 (H5N1) and a comparison A/Beijing/501/2009 (H1N1). miRNA microarray and quantitative PCR analysis showed that several miRNAs were differentially expressed in A549 cells during IAV infection. Subsequently, we demonstrated that IAV replication was essential for the regulation of these miRNAs, and bioinformatic analysis revealed that the targets of these miRNAs affected biological processes relevant to IAV replication. Specifically, miR-21-3p was found to be down-regulated in IAV-infected A549 cells and selected for further detailed analysis. Target prediction and functional study illustrated that miR-21-3p repressed the expression of HDAC8 by targeting its 3′UTR. Furthermore, we confirmed miR-21-3p could promote virus replication, which was similar to the result of knocking down HDAC8, indicating that miR-21-3p promoted IAV replication by suppressing HDAC8 expression. Altogether, our results suggest a potential host defense against IAV through down-regulation of miR-21-3p.
Collapse
Affiliation(s)
- Binghui Xia
- Laboratory of Protein Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Jiansheng Lu
- Laboratory of Protein Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Rong Wang
- Laboratory of Protein Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Zhixin Yang
- Laboratory of Protein Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Xiaowei Zhou
- Laboratory of Protein Engineering, Beijing Institute of Biotechnology, Beijing, China
| | - Peitang Huang
- Laboratory of Protein Engineering, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
57
|
Abstract
Viruses infecting vertebrate hosts must overcome the interferon (IFN)-mediated antiviral response to replicate and propagate to new hosts. The complex regulation of the IFN response allows viruses to antagonize IFN at multiple levels. However, no single strategy appears to be the golden ticket, and viruses have adopted multiple means to dampen this host defense. This Review does not exhaustively cover all mechanisms of viral IFN antagonism. Rather it examines the ten most common strategies that viruses use to subvert the IFN response with examples from publications appearing in the last 10 years of Cell Host & Microbe. The virus-host interactions involved in induction and evasion of IFN represent a fertile area of research due to the significant large number of host and viral products that regulate this response, resulting in an intricate dance between hosts and their pathogens to achieve an optimal balance between virus replication, host disease, and survival.
Collapse
Affiliation(s)
- Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
58
|
Are microRNAs Important Players in HIV-1 Infection? An Update. Viruses 2018; 10:v10030110. [PMID: 29510515 PMCID: PMC5869503 DOI: 10.3390/v10030110] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 02/21/2018] [Accepted: 02/25/2018] [Indexed: 12/15/2022] Open
Abstract
HIV-1 has already claimed over 35 million human lives globally. No curative treatments are currently available, and the only treatment option for over 36 million people currently living with HIV/AIDS are antiretroviral drugs that disrupt the function of virus-encoded proteins. However, such virus-targeted therapeutic strategies are constrained by the ability of the virus to develop drug-resistance. Despite major advances in HIV/AIDS research over the years, substantial knowledge gaps exist in many aspects of HIV-1 replication, especially its interaction with the host. Hence, understanding the mechanistic details of virus–host interactions may lead to novel therapeutic strategies for the prevention and/or management of HIV/AIDS. Notably, unprecedented progress in deciphering host gene silencing processes mediated by several classes of cellular small non-coding RNAs (sncRNA) presents a promising and timely opportunity for developing non-traditional antiviral therapeutic strategies. Cellular microRNAs (miRNA) belong to one such important class of sncRNAs that regulate protein synthesis. Evidence is mounting that cellular miRNAs play important roles in viral replication, either usurped by the virus to promote its replication or employed by the host to control viral infection by directly targeting the viral genome or by targeting cellular proteins required for productive virus replication. In this review, we summarize the findings to date on the role of miRNAs in HIV-1 biology.
Collapse
|
59
|
Tang CZ, Li KR, Yu Q, Jiang Q, Yao J, Cao C. Activation of Nrf2 by Ginsenoside Rh3 protects retinal pigment epithelium cells and retinal ganglion cells from UV. Free Radic Biol Med 2018; 117:238-246. [PMID: 29427790 DOI: 10.1016/j.freeradbiomed.2018.02.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/11/2018] [Accepted: 02/02/2018] [Indexed: 12/20/2022]
Abstract
Excessive Ultra-violet (UV) radiation shall induce damages to resident retinal pigment epithelium (RPE) cells (RPEs) and retinal ganglion cells (RGCs). Here we tested the potential activity of Ginsenoside Rh3 ("Rh3") against the process. In cultured human RPEs and RGCs, pretreatment with Rh3 inhibited UV-induced reactive oxygen species (ROS) production and following apoptotic/non-apoptotic cell death. Rh3 treatment in retinal cells induced nuclear-factor-E2-related factor 2 (Nrf2) activation, which was evidenced by Nrf2 protein stabilization and its nuclear translocation, along with transcription of antioxidant responsive element (ARE)-dependent genes (HO1, NOQ1 and GCLC). Nrf2 knockdown by targeted-shRNA almost abolished Rh3-induced retinal cell protection against UV. Further studies found that Rh3 induced microRNA-141 ("miR-141") expression, causing downregulation of its targeted gene Keap1 in RPEs and RGCs. On the other hand, Rh3-induced Nrf2 activation and retinal cell protection were largely attenuated by the miR-141's inhibitor, antagomiR-141. In vivo, intravitreal injection of Rh3 inhibited retinal dysfunction by light damage in mice. Rh3 intravitreal injection also induced miR-141 expression, Keap1 downregulation and Nrf2 activation in mouse retinas. We conclude that Rh3 protects retinal cells from UV via activating Nrf2 signaling.
Collapse
Affiliation(s)
- Chun-Zhou Tang
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China; Ophthalmology Department, Jiangsu Jiangyin people's Hospital, Jiangyin, China
| | - Ke-Ran Li
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
| | - Qing Yu
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China
| | - Qin Jiang
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China.
| | - Jin Yao
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China.
| | - Cong Cao
- The Affiliated Eye Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China; Center of Translational Medicine, The First People Hospital of Zhangjiagang City, Soochow University, Suzhou, China; North District, The Municipal Hospital of Suzhou, Suzhou, China.
| |
Collapse
|
60
|
Cui L, Markou A, Stratton CW, Lianidou E. Diagnosis and Assessment of Microbial Infections with Host and Microbial MicroRNA Profiles. ADVANCED TECHNIQUES IN DIAGNOSTIC MICROBIOLOGY 2018. [PMCID: PMC7119978 DOI: 10.1007/978-3-319-95111-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) encoded by viral genome or host have been found participating in host-microbe interactions. Differential expression profiles of miRNAs were shown linking to specific disease pathologies which indicated its potency as diagnostic/prognostic biomarkers of infectious disease. This was emphasized by the discovery of circulating miRNAs which were found to be remarkably stable in mammalian biofluids. Standardized methods of miRNA quantification including RNA isolation should be established before they will be ready for use in clinical practice.
Collapse
|
61
|
Retraction: Xianzheng Gao, Shenglei Li, Wencai Li, Guannan Wang, Wugan Zhao, Jing Han, Changying Diao, Xiaohui Wang, and Mingzhi Zhang, MicroRNA-539 suppresses tumor cell growth by targeting the WNT8B gene in non-small cell lung cancer. J. Cell. Biochem. Accepted Article doi.org/10.1002/jcb.26634. J Cell Biochem 2017; 120:2687-2687. [PMID: 29266418 DOI: 10.1002/jcb.26634] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/19/2017] [Indexed: 12/20/2022]
|
62
|
Li XY, Wang SS, Han Z, Han F, Chang YP, Yang Y, Xue M, Sun B, Chen LM. Triptolide Restores Autophagy to Alleviate Diabetic Renal Fibrosis through the miR-141-3p/PTEN/Akt/mTOR Pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 9:48-56. [PMID: 29246323 PMCID: PMC5602517 DOI: 10.1016/j.omtn.2017.08.011] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 08/22/2017] [Accepted: 08/22/2017] [Indexed: 11/04/2022]
Abstract
Fibrosis is the major pathological feature of diabetic kidney disease (DKD). Autophagy, a process to maintain metabolic homeostasis, is obviously inhibited in DKD. Triptolide (TP) is a traditional Chinese medicine extract known for immune suppression and anti-inflammatory and anti-cancer activities. In this study, we investigated the effects of TP on autophagy and fibrosis in DKD. TP restored autophagy and alleviated fibrosis in DKD rats and high-glucose-incubated human mesangial cells. After we applied 3-methyladenine (an autophagy inhibitor) and autophagy-related gene 5-small interfering RNA (siRNA), we found that the improvement of fibrosis on TP was related to the restoration of autophagy. In addition, miR-141-3p levels were increased under high glucose but reduced after TP treatment. miR-141-3p overexpression aggravated the fibrosis and restrained the autophagy further, while miR-141-3p inhibition imitated the effects of TP. As an action target, phosphatase and tensin homolog (PTEN) showed corresponding opposite changes. After PTEN-siRNA transfection, the effects of TP on autophagy and fibrosis were inhibited. PTEN levels were downregulated, with downstream phosphorylated protein kinase B (Akt) and the mammalian target of rapamycin (mTOR) upregulated in high glucose, which were reversed by TP treatment. These findings indicate that TP alleviates fibrosis by restoring autophagy through the miR-141-3p/PTEN/Akt/mTOR pathway and is a novel therapeutic option for DKD.
Collapse
Affiliation(s)
- Xiao-Yu Li
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Shan-Shan Wang
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Zhe Han
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Fei Han
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Yun-Peng Chang
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Yang Yang
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Mei Xue
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China
| | - Bei Sun
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China.
| | - Li-Ming Chen
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
63
|
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
|
64
|
Dave P, George B, Sharma DK, Das S. Polypyrimidine tract-binding protein (PTB) and PTB-associated splicing factor in CVB3 infection: an ITAF for an ITAF. Nucleic Acids Res 2017. [PMID: 28633417 PMCID: PMC5587786 DOI: 10.1093/nar/gkx519] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The 5′ UTR of Coxsackievirus B3 (CVB3) contains internal ribosome entry site (IRES), which allows cap-independent translation of the viral RNA and a 5′-terminal cloverleaf structure that regulates viral replication, translation and stability. Here, we demonstrate that host protein PSF (PTB associated splicing factor) interacts with the cloverleaf RNA as well as the IRES element. PSF was found to be an important IRES trans acting factor (ITAF) for efficient translation of CVB3 RNA. Interestingly, cytoplasmic abundance of PSF protein increased during CVB3 infection and this is regulated by phosphorylation status at two different amino acid positions. Further, PSF protein was up-regulated in CVB3 infection. The expression of CVB3–2A protease alone could also induce increased PSF protein levels. Furthermore, we observed the presence of an IRES element in the 5′UTR of PSF mRNA, which is activated during CVB3 infection and might contribute to the elevated levels of PSF. It appears that PSF IRES is also positively regulated by PTB, which is known to regulate CVB3 IRES. Taken together, the results suggest for the first time a novel mechanism of regulations of ITAFs during viral infection, where an ITAF undergoes IRES mediated translation, sustaining its protein levels under condition of translation shut-off.
Collapse
Affiliation(s)
- Pratik Dave
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Biju George
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Divya Khandige Sharma
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Saumitra Das
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, Karnataka, India.,Centre for Infectious Disease Research, Indian Institute of Science, Bangalore 560012, Karnataka, India
| |
Collapse
|
65
|
Engelmann I, Alidjinou EK, Bertin A, Bossu J, Villenet C, Figeac M, Sane F, Hober D. Persistent coxsackievirus B4 infection induces microRNA dysregulation in human pancreatic cells. Cell Mol Life Sci 2017; 74:3851-3861. [PMID: 28601984 PMCID: PMC11107484 DOI: 10.1007/s00018-017-2567-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/16/2017] [Accepted: 06/06/2017] [Indexed: 12/15/2022]
Abstract
Enterovirus infections are implicated in the development of type 1 diabetes (T1D). MicroRNAs as regulators of gene expression are involved in many physiological and pathological processes. Given that viral infections dysregulate cellular microRNAs, we investigated the impact of persistent coxsackievirus B4 infection on microRNA expression of human pancreatic cells. Next-generation sequencing was used to determine microRNA expression in PANC-1 cells persistently infected (for several weeks) with coxsackievirus B4 and uninfected control cells. Target prediction restricted to T1D risk genes was performed with miRWalk2.0. Functional annotation analysis was performed with DAVID6.7. Expression of selected microRNAs and T1D risk genes was measured by quantitative reverse-transcription polymerase chain reaction. Eighty-one microRNAs were dysregulated in persistently infected PANC-1 cells. Forty-nine of the known fifty-five T1D risk genes were predicted as putative targets of at least one of the dysregulated microRNAs. Most functional annotation terms that were enriched in these 49 putative target genes were related to the immune response or autoimmunity. mRNA levels of AFF3, BACH2, and IL7R differed significantly between persistently infected cells and uninfected cells. This is the first characterization of the microRNA expression profile changes induced by persistent coxsackievirus B4 infection in pancreatic cells. The predicted targeting of genes involved in the immune response and autoimmunity by the dysregulated microRNAs as well as the dysregulated expression of diabetes risk genes shows that persistent coxsackievirus B4 infection profoundly impacts the host cell. These data support the hypothesis of a possible link between persistent coxsackievirus B4 infection and the development of T1D.
Collapse
Affiliation(s)
- Ilka Engelmann
- Univ Lille Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, F-59000, France
| | - Enagnon K Alidjinou
- Univ Lille Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, F-59000, France
| | - Antoine Bertin
- Univ Lille Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, F-59000, France
| | - Johann Bossu
- Univ Lille Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, F-59000, France
| | - Céline Villenet
- CHU Lille, Plate-forme de Génomique Fonctionnelle et Structurale, Lille, F-59000, France
| | - Martin Figeac
- CHU Lille, Plate-forme de Génomique Fonctionnelle et Structurale, Lille, F-59000, France
| | - Famara Sane
- Univ Lille Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, F-59000, France
| | - Didier Hober
- Univ Lille Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, F-59000, France.
| |
Collapse
|
66
|
Chen SG, Leu YL, Cheng ML, Ting SC, Liu CC, Wang SD, Yang CH, Hung CY, Sakurai H, Chen KH, Ho HY. Anti-enterovirus 71 activities of Melissa officinalis extract and its biologically active constituent rosmarinic acid. Sci Rep 2017; 7:12264. [PMID: 28947773 PMCID: PMC5613005 DOI: 10.1038/s41598-017-12388-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 09/07/2017] [Indexed: 01/22/2023] Open
Abstract
Enterovirus 71 (EV71) infection is endemic in the Asia-Pacific region. No specific antiviral drug has been available to treat EV71 infection. Melissa officinalis (MO) is a medicinal plant with long history of usage in the European and Middle East. We investigated whether an aqueous solution of concentrated methanolic extract (MOM) possesses antiviral activity. MOM inhibited plaque formation, cytopathic effect, and viral protein synthesis in EV71-infected cells. Using spectral techniques, we identified rosmarinic acid (RA) as a biologically active constituent of MOM. RA reduced viral attachment and entry; cleavage of eukaryotic translation initiation factor 4 G (eIF4G); reactive oxygen species (ROS) generation; and translocation of heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) from nucleus to cytoplasm. It alleviated EV71-induced hyperphosphorylation of p38 kinase and EPS15. RA is likely to suppress ROS-mediated p38 kinase activation, and such downstream molecular events as hnRNP A1 translocation and EPS15-regulated membrane trafficking in EV71-infected cells. These findings suggest that MO and its constituent RA possess anti-EV71 activities, and may serve as a candidate drug for therapeutic and prophylactic uses against EV71 infection.
Collapse
Affiliation(s)
- Sin-Guang Chen
- Graduate Institute of Biomedical Science, Chang Gung University, Guishan, Taoyuan, Taiwan
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital at Linkou, Guishan, Taoyuan, Taiwan
- Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Mei-Ling Cheng
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Guishan, Taoyuan, Taiwan
- Healthy Aging Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan
- Metabolomics Core Laboratory, Chang Gung University, Guishan, Taoyuan, Taiwan
- Clinical Phenome Center, Chang Gung Memorial Hospital at Linkou, Guishan, Taoyuan, Taiwan
| | - Siew Chin Ting
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Chuan Liu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Shulhn-Der Wang
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Cheng-Hung Yang
- Graduate Institute of Biomedical Science, Chang Gung University, Guishan, Taoyuan, Taiwan
| | - Cheng-Yu Hung
- Healthy Aging Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan
- Metabolomics Core Laboratory, Chang Gung University, Guishan, Taoyuan, Taiwan
| | - Hiroaki Sakurai
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Kuan-Hsing Chen
- Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Hung-Yao Ho
- Healthy Aging Research Center, Chang Gung University, Guishan, Taoyuan, Taiwan.
- Clinical Phenome Center, Chang Gung Memorial Hospital at Linkou, Guishan, Taoyuan, Taiwan.
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
67
|
miRNA-141 attenuates UV-induced oxidative stress via activating Keap1-Nrf2 signaling in human retinal pigment epithelium cells and retinal ganglion cells. Oncotarget 2017; 8:13186-13194. [PMID: 28061435 PMCID: PMC5355087 DOI: 10.18632/oncotarget.14489] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/07/2016] [Indexed: 12/30/2022] Open
Abstract
Activation of NF-E2-related factor 2 (Nrf2) signaling could protect cells from ultra violet (UV) radiation. We aim to provoke Nrf2 activation via downregulating its inhibitor Keap1 by microRNA-141 (“miR-141”). In both human retinal pigment epithelium cells (RPEs) and retinal ganglion cells (RGCs), forced-expression of miR-141 downregulated Keap1, causing Nrf2 stabilization, accumulation and nuclear translocation, which led to transcription of multiple antioxidant-responsive element (ARE) genes (HO1, NOQ1 and GCLC). Further, UV-induced reactive oxygen species (ROS) production and cell death were significantly attenuated in miR-141-expressing RPEs and RGCs. On the other hand, depletion of miR-141 via expressing its inhibitor antagomiR-141 led to Keap1 upregulation and Nrf2 degradation, which aggravated UV-induced death of RPEs and RGCs. Significantly, Nrf2 shRNA knockdown almost abolished miR-141-mediated cytoprotection against UV in RPEs. These results demonstrate that miR-141 targets Keap1 to activate Nrf2 signaling, which protects RPEs and RGCs from UV radiation.
Collapse
|
68
|
Antiviral activities of Schizonepeta tenuifolia Briq. against enterovirus 71 in vitro and in vivo. Sci Rep 2017; 7:935. [PMID: 28428548 PMCID: PMC5430552 DOI: 10.1038/s41598-017-01110-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/23/2017] [Indexed: 12/31/2022] Open
Abstract
No effective drug is currently available for treatment of enterovirus 71 (EV71) infection. Schizonepeta tenuifolia Briq. (ST) has been used as a herbal constituent of traditional Chinese medicine. We studied whether the aqueous extract of Schizonepeta tenuifolia Briq (STE) has antiviral activity. STE inhibited replication of EV71, as evident by its ability to diminish plaque formation and cytopathic effect induced by EV71, and to inhibit the synthesis of viral RNA and protein. Moreover, daily single-dose STE treatment significantly improved the survival of EV71-infected mice, and ameliorated the symptoms. Mechanistically, STE exerts multiple effects on enteroviral infection. Treatment with STE reduced viral attachment and entry; the cleavage of eukaryotic translation initiation factor 4 G (eIF4G) by EV71 protease, 2Apro; virus-induced reactive oxygen species (ROS) formation; and relocation of heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) from the nucleus to the cytoplasm. It was accompanied by a decline in EV71-associated hyperphosphorylation of p38 kinase and EPS15. It is plausible that STE may inhibit ROS-induced p38 kinase activation, and subsequent hnRNP A1 relocation and EPS15-mediated membrane trafficking in infected cells. These findings suggest that STE possesses anti-EV71 activities, and may serve as health food or candidate antiviral drug for protection against EV71.
Collapse
|
69
|
Pan X, Yang X, Zang J, Zhang S, Huang N, Guan X, Zhang J, Wang Z, Li X, Lei X. Downregulation of eIF4G by microRNA-503 enhances drug sensitivity of MCF-7/ADR cells through suppressing the expression of ABC transport proteins. Oncol Lett 2017; 13:4785-4793. [PMID: 28599480 PMCID: PMC5453003 DOI: 10.3892/ol.2017.6049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 02/23/2017] [Indexed: 11/16/2022] Open
Abstract
Overexpression of adenosine triphosphate-binding cassette (ABC) transport protein is emerging as a critical contributor to anticancer drug resistance. The eukaryotic translation initiation factor (eIF) 4F complex, the key modulator of mRNA translation, is regulated by the phosphoinositide 3-kinase-AKT-mammalian target of rapamycin pathway in anticancer drug-resistant tumors. The present study demonstrated the roles of ABC translation protein alterations in the acquisition of the Adriamycin (ADM)-resistant phenotype of MCF-7 human breast cells. Quantitative polymerase chain reaction and western blot analysis were applied to examine the differences in mRNA and protein levels, respectively. It was found that the expression of the ABC sub-family B member 1, ABC sub-family C member 1 and ABC sub-family G member 2 transport proteins were upregulated in MCF-7/ADR cells. An MTT assay was used to detect the cell viability, from the results MCF-7/ADR cells were less sensitive to ADM, tamoxifen (TAM) and taxol (TAX) treatment compared with MCF-7 cells. We predicted that the 3′-untranslated region of eukaryotic translation initiation factor 4-γ 1 (eIF4G) contains a potential miRNA binding site for microRNA (miR)-503 through using computational programs. These binding sites were confirmed by luciferase reporter assays. eIF4G mRNA degradation was accelerated in cells transfected with miR-503 mimics. Furthermore, it was demonstrated that eIF4G and ABC translation proteins were significantly downregulated in MCF-7/ADR cells after transfection with miR-503. It was found that miR-503 mimics could sensitize the cells to treatment with ADM, TAM and TAX. These findings demonstrated for the first time that eIF4G acted as a key factor in MCF-7/ADR cells, and may be an efficient agent for preventing and reversing multi-drug resistance in breast cancer.
Collapse
Affiliation(s)
- Xia Pan
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiaoyan Yang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jinglei Zang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Si Zhang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Nan Huang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xinxin Guan
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jianhua Zhang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zhihui Wang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xi Li
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiaoyong Lei
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
70
|
Yang X, Xie J, Jia L, Liu N, Liang Y, Wu F, Liang B, Li Y, Wang J, Sheng C, Li H, Liu H, Ma Q, Yang C, Du X, Qiu S, Song H. Analysis of miRNAs Involved in Mouse Brain Damage upon Enterovirus 71 Infection. Front Cell Infect Microbiol 2017; 7:133. [PMID: 28469998 PMCID: PMC5395563 DOI: 10.3389/fcimb.2017.00133] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 03/31/2017] [Indexed: 12/02/2022] Open
Abstract
Enterovirus 71 (EV71) infects the central nervous system (CNS) and causes brainstem encephalitis in children. MiRNAs have been found to play various functions in EV71 infection in human cell lines. To identify potential miRNAs involved in the inflammatory injury in CNS, our study, for the first time, performed a miRNA microarray assay in vivo using EV71 infected mice brains. Twenty differentially expressed miRNAs were identified (four up- and 16 down-regulated) and confirmed by qRT-PCR. The target genes of these miRNAs were analyzed using KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis, revealing that the miRNAs were mainly involved in the regulation of inflammation and neural system function. MiR-150-5p, -3082-5p, -3473a, -468-3p, -669n, -721, -709, and -5107-5p that regulate MAPK and chemokine signaling were all down-regulated, which might result in increased cytokine production. In addition, miR-3473a could also regulate focal adhesion and leukocyte trans-endothelial migration, suggesting a role in virus-induced blood-brain barrier disruption. The miRNAs and pathways identified in this study could help to understand the intricate interactions between EV71 and the brain injury, offering new insight for the future research of the molecular mechanism of EV71 induced brainstem encephalitis.
Collapse
Affiliation(s)
- Xiaoxia Yang
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Jing Xie
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Leili Jia
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Nan Liu
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Yuan Liang
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Fuli Wu
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Beibei Liang
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Yongrui Li
- The Key Laboratory of Pharmacology and Molecular Biology, Medical College, Henan University of Science and TechnologyLuoyang, China
| | - Jinyan Wang
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Chunyu Sheng
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Hao Li
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Hongbo Liu
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Qiuxia Ma
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Chaojie Yang
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Xinying Du
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Shaofu Qiu
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| | - Hongbin Song
- Center for Infectious Disease Control, Institute of Disease Control and Prevention, Academy of Military Medical SciencesBeijing, China
| |
Collapse
|
71
|
A MicroRNA Screen Identifies the Wnt Signaling Pathway as a Regulator of the Interferon Response during Flavivirus Infection. J Virol 2017; 91:JVI.02388-16. [PMID: 28148804 DOI: 10.1128/jvi.02388-16] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 01/25/2017] [Indexed: 02/07/2023] Open
Abstract
The impact of mosquito-borne flavivirus infections worldwide is significant, and many critical aspects of these viruses' biology, including virus-host interactions, host cell requirements for replication, and how virus-host interactions impact pathology, remain to be fully understood. The recent reemergence and spread of flaviviruses, including dengue virus (DENV), West Nile virus (WNV), and Zika virus (ZIKV), highlight the importance of performing basic research on this important group of pathogens. MicroRNAs (miRNAs) are small, noncoding RNAs that modulate gene expression posttranscriptionally and have been demonstrated to regulate a broad range of cellular processes. Our research is focused on identifying pro- and antiflaviviral miRNAs as a means of characterizing cellular pathways that support or limit viral replication. We have screened a library of known human miRNA mimics for their effect on the replication of three flaviviruses, DENV, WNV, and Japanese encephalitis virus (JEV), using a high-content immunofluorescence screen. Several families of miRNAs were identified as inhibiting multiple flaviviruses, including the miRNA miR-34, miR-15, and miR-517 families. Members of the miR-34 family, which have been extensively characterized for their ability to repress Wnt/β-catenin signaling, demonstrated strong antiflaviviral effects, and this inhibitory activity extended to other viruses, including ZIKV, alphaviruses, and herpesviruses. Previous research suggested a possible link between the Wnt and type I interferon (IFN) signaling pathways. Therefore, we investigated the role of type I IFN induction in the antiviral effects of the miR-34 family and confirmed that these miRNAs potentiate interferon regulatory factor 3 (IRF3) phosphorylation and translocation to the nucleus, the induction of IFN-responsive genes, and the release of type I IFN from transfected cells. We further demonstrate that the intersection between the Wnt and IFN signaling pathways occurs at the point of glycogen synthase kinase 3β (GSK3β)-TANK-binding kinase 1 (TBK1) binding, inducing TBK1 to phosphorylate IRF3 and initiate downstream IFN signaling. In this way, we have identified a novel cellular signaling network with a critical role in regulating the replication of multiple virus families. These findings highlight the opportunities for using miRNAs as tools to discover and characterize unique cellular factors involved in supporting or limiting virus replication, opening up new avenues for antiviral research.IMPORTANCE MicroRNAs are a class of small regulatory RNAs that modulate cellular processes through the posttranscriptional repression of multiple transcripts. We hypothesized that individual miRNAs may be capable of inhibiting viral replication through their effects on host proteins or pathways. To test this, we performed a high-content screen for miRNAs that inhibit the replication of three medically relevant members of the flavivirus family: West Nile virus, Japanese encephalitis virus, and dengue virus 2. The results of this screen identify multiple miRNAs that inhibit one or more of these viruses. Extensive follow-up on members of the miR-34 family of miRNAs, which are active against all three viruses as well as the closely related Zika virus, demonstrated that miR-34 functions through increasing the infected cell's ability to respond to infection through the interferon-based innate immune pathway. Our results not only add to the knowledge of how viruses interact with cellular pathways but also provide a basis for more extensive data mining by providing a comprehensive list of miRNAs capable of inhibiting flavivirus replication. Finally, the miRNAs themselves or cellular pathways identified as modulating virus infection may prove to be novel candidates for the development of therapeutic interventions.
Collapse
|
72
|
Alidjinou EK, Engelmann I, Bossu J, Villenet C, Figeac M, Romond MB, Sané F, Hober D. Persistence of Coxsackievirus B4 in pancreatic ductal-like cells results in cellular and viral changes. Virulence 2017; 8:1229-1244. [PMID: 28112573 DOI: 10.1080/21505594.2017.1284735] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Although known as cytolytic viruses, group B coxackieviruses (CVB) are able to establish a persistent infection in vitro and in vivo. Viral persistence has been reported as a key mechanism in the pathogenesis of CVB-associated chronic diseases such as type 1 diabetes (T1D). The impact of CVB4 persistence on human pancreas ductal-like cells was investigated. METHODS A persistent CVB4 infection was established in ductal-like cells. PDX-1 expression, resistance to CVB4-induced lysis and CAR expression were evaluated. The profile of cellular microRNAs (miRNAs) was investigated through miRNA-sequencing. Viral phenotypic changes were examined, and genomic modifications were assessed by sequencing of the viral genome. RESULTS The CVB4 persistence in ductal-like cells was productive, with continuous release of infectious particles. Persistently infected cells displayed a resistance to CVB4-induced lysis upon superinfection and expression of PDX-1 and CAR was decreased. These changes were maintained even after virus clearance. The patterns of cellular miRNA expression in mock-infected and in CVB4-persistently infected ductal-like cells were clearly different. The persistent infection-derived virus (PIDV) was still able to induce cytopathic effect but its plaques were smaller than the parental virus. Several mutations appeared in various PIDV genome regions, but amino acid substitutions did not affect the predicted site of interaction with CAR. CONCLUSION Cellular and viral changes occur during persistent infection of human pancreas ductal-like cells with CVB4. The persistence of cellular changes even after virus clearance supports the hypothesis of a long-lasting impact of persistent CVB infection on the cells.
Collapse
Affiliation(s)
- E K Alidjinou
- a Univ Lille, CHU Lille, Laboratoire de Virologie EA3610 , Lille , France
| | - I Engelmann
- a Univ Lille, CHU Lille, Laboratoire de Virologie EA3610 , Lille , France
| | - J Bossu
- a Univ Lille, CHU Lille, Laboratoire de Virologie EA3610 , Lille , France
| | - C Villenet
- b Plate-forme de Génomique Fonctionnelle et Structurale , CHU de Lille , France
| | - M Figeac
- b Plate-forme de Génomique Fonctionnelle et Structurale , CHU de Lille , France
| | - M-B Romond
- a Univ Lille, CHU Lille, Laboratoire de Virologie EA3610 , Lille , France
| | - F Sané
- a Univ Lille, CHU Lille, Laboratoire de Virologie EA3610 , Lille , France
| | - D Hober
- a Univ Lille, CHU Lille, Laboratoire de Virologie EA3610 , Lille , France
| |
Collapse
|
73
|
Trobaugh DW, Klimstra WB. MicroRNA Regulation of RNA Virus Replication and Pathogenesis. Trends Mol Med 2016; 23:80-93. [PMID: 27989642 PMCID: PMC5836316 DOI: 10.1016/j.molmed.2016.11.003] [Citation(s) in RCA: 286] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/30/2016] [Accepted: 11/12/2016] [Indexed: 01/08/2023]
Abstract
microRNAs (miRNAs) are non-coding RNAs that regulate many processes within a cell by manipulating protein levels through direct binding to mRNA and influencing translation efficiency, or mRNA abundance. Recent evidence demonstrates that miRNAs can also affect RNA virus replication and pathogenesis through direct binding to the RNA virus genome or through virus-mediated changes in the host transcriptome. Here, we review the current knowledge on the interaction between RNA viruses and cellular miRNAs. We also discuss how cell and tissue-specific expression of miRNAs can directly affect viral pathogenesis. Understanding the role of cellular miRNAs during viral infection may lead to the identification of novel mechanisms to block RNA virus replication or cell-specific regulation of viral vector targeting. Some RNA viruses possess miRNA-binding sites in a range of locations within the viral genome, including the 5′ and 3′ non-translated regions. Host cell miRNAs can bind to RNA virus genomes, enhancing genome stability, repressing translation of the viral genome, or altering free miRNA levels within the cell. miRNAs contribute to viral pathogenesis by promoting evasion of the host antiviral immune response, enhancing viral replication, or, potentially, altering miRNA-mediated host gene regulation. RNA virus infection can lead to widespread changes in the host transcriptome by modulating cell-specific miRNA levels.
Collapse
Affiliation(s)
- Derek W Trobaugh
- Center for Vaccine Research, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - William B Klimstra
- Center for Vaccine Research, Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
74
|
Isaacs SR, Wang J, Kim KW, Yin C, Zhou L, Mi QS, Craig ME. MicroRNAs in Type 1 Diabetes: Complex Interregulation of the Immune System, β Cell Function and Viral Infections. Curr Diab Rep 2016; 16:133. [PMID: 27844276 DOI: 10.1007/s11892-016-0819-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Since the discovery of the first mammalian microRNA (miRNA) more than two decades ago, a plethora of miRNAs has been identified in humans, now amounting to more than 2500. Essential for post-transcriptional regulation of gene networks integral for developmental pathways and immune response, it is not surprising that dysregulation of miRNAs is often associated with the aetiology of complex diseases including cancer, diabetes and autoimmune disorders. Despite massive expansion of small RNA studies and extensive investigation in diverse disease contexts, the role of miRNAs in type 1 diabetes has only recently been explored. Key studies using human islets have recently implicated virus-induced miRNA dysregulation as a pivotal mechanism of β cell destruction, while the interplay between miRNAs, the immune system and β cell survival has been illustrated in studies using animal and cellular models of disease. The role of specific miRNAs as major players in immune system homeostasis highlights their exciting potential as therapeutics and prognostic biomarkers of type 1 diabetes.
Collapse
Affiliation(s)
- Sonia R Isaacs
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- UNSW and POWH Virology Research Laboratory, Prince of Wales Hospital, Sydney, NSW, 2031, Australia
| | - Jie Wang
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Dermatology, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Ki Wook Kim
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- UNSW and POWH Virology Research Laboratory, Prince of Wales Hospital, Sydney, NSW, 2031, Australia
| | - Congcong Yin
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Dermatology, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Li Zhou
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Dermatology, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Qing Sheng Mi
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Dermatology, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Maria E Craig
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia.
- UNSW and POWH Virology Research Laboratory, Prince of Wales Hospital, Sydney, NSW, 2031, Australia.
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, NSW, 2145, Australia.
- Discipline of Child and Adolescent Health, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
75
|
Foo CH, Rootes CL, Cowley K, Marsh GA, Gould CM, Deffrasnes C, Cowled CJ, Klein R, Riddell SJ, Middleton D, Simpson KJ, Wang LF, Bean AGD, Stewart CR. Dual microRNA Screens Reveal That the Immune-Responsive miR-181 Promotes Henipavirus Entry and Cell-Cell Fusion. PLoS Pathog 2016; 12:e1005974. [PMID: 27783670 PMCID: PMC5082662 DOI: 10.1371/journal.ppat.1005974] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 10/03/2016] [Indexed: 12/24/2022] Open
Abstract
Hendra and Nipah viruses (family Paramyxoviridae, genus Henipavirus) are bat-borne viruses that cause fatal disease in humans and a range of other mammalian species. Gaining a deeper understanding of host pathways exploited by henipaviruses for infection may identify targets for new anti-viral therapies. Here we have performed genome-wide high-throughput agonist and antagonist screens at biosafety level 4 to identify host-encoded microRNAs (miRNAs) impacting henipavirus infection in human cells. Members of the miR-181 and miR-17~93 families strongly promoted Hendra virus infection. miR-181 also promoted Nipah virus infection, but did not affect infection by paramyxoviruses from other genera, indicating specificity in the virus-host interaction. Infection promotion was primarily mediated via the ability of miR-181 to significantly enhance henipavirus-induced membrane fusion. Cell signalling receptors of ephrins, namely EphA5 and EphA7, were identified as novel negative regulators of henipavirus fusion. The expression of these receptors, as well as EphB4, were suppressed by miR-181 overexpression, suggesting that simultaneous inhibition of several Ephs by the miRNA contributes to enhanced infection and fusion. Immune-responsive miR-181 levels was also up-regulated in the biofluids of ferrets and horses infected with Hendra virus, suggesting that the host innate immune response may promote henipavirus spread and exacerbate disease severity. This study is the first genome-wide screen of miRNAs influencing infection by a clinically significant mononegavirus and nominates select miRNAs as targets for future anti-viral therapy development.
Collapse
Affiliation(s)
- Chwan Hong Foo
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Christina L. Rootes
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Karla Cowley
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Glenn A. Marsh
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Cathryn M. Gould
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Celine Deffrasnes
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Christopher J. Cowled
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Reuben Klein
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Sarah J. Riddell
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Deborah Middleton
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Kaylene J. Simpson
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Lin-Fa Wang
- Program in Emerging Infectious Diseases, Duke-NUS Graduate Medical School, Singapore
| | - Andrew G. D. Bean
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
| | - Cameron R. Stewart
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria, Australia
- * E-mail:
| |
Collapse
|
76
|
Abstract
Viral myocarditis remains a prominent infectious-inflammatory disease for patients throughout the lifespan. The condition presents several challenges including varied modes of clinical presentation, a range of timepoints when patients come to attention, a diversity of approaches to diagnosis, a spectrum of clinical courses, and unsettled perspectives on therapeutics in different patient settings and in the face of different viral pathogens. In this review, we examine current knowledge about viral heart disease and especially provide information on evolving understanding of mechanisms of disease and efforts by investigators to identify and evaluate potential therapeutic avenues for intervention.
Collapse
Affiliation(s)
- Gabriel Fung
- From the Department of Pathology and Laboratory Medicine (G.F., H.L., Y.Q., D.Y., B.M.), Centre for Heart Lung Innovation (G.F., H.L., Y.Q., D.Y., B.M.), Centre of Excellence for Prevention of Organ Failure (PROOF Centre), and Institute for Heart + Lung Health, St. Paul's Hospital (B.M.), University of British Columbia, Vancouver, British Columbia, Canada
| | - Honglin Luo
- From the Department of Pathology and Laboratory Medicine (G.F., H.L., Y.Q., D.Y., B.M.), Centre for Heart Lung Innovation (G.F., H.L., Y.Q., D.Y., B.M.), Centre of Excellence for Prevention of Organ Failure (PROOF Centre), and Institute for Heart + Lung Health, St. Paul's Hospital (B.M.), University of British Columbia, Vancouver, British Columbia, Canada
| | - Ye Qiu
- From the Department of Pathology and Laboratory Medicine (G.F., H.L., Y.Q., D.Y., B.M.), Centre for Heart Lung Innovation (G.F., H.L., Y.Q., D.Y., B.M.), Centre of Excellence for Prevention of Organ Failure (PROOF Centre), and Institute for Heart + Lung Health, St. Paul's Hospital (B.M.), University of British Columbia, Vancouver, British Columbia, Canada
| | - Decheng Yang
- From the Department of Pathology and Laboratory Medicine (G.F., H.L., Y.Q., D.Y., B.M.), Centre for Heart Lung Innovation (G.F., H.L., Y.Q., D.Y., B.M.), Centre of Excellence for Prevention of Organ Failure (PROOF Centre), and Institute for Heart + Lung Health, St. Paul's Hospital (B.M.), University of British Columbia, Vancouver, British Columbia, Canada
| | - Bruce McManus
- From the Department of Pathology and Laboratory Medicine (G.F., H.L., Y.Q., D.Y., B.M.), Centre for Heart Lung Innovation (G.F., H.L., Y.Q., D.Y., B.M.), Centre of Excellence for Prevention of Organ Failure (PROOF Centre), and Institute for Heart + Lung Health, St. Paul's Hospital (B.M.), University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
77
|
Herbert KM, Nag A. A Tale of Two RNAs during Viral Infection: How Viruses Antagonize mRNAs and Small Non-Coding RNAs in The Host Cell. Viruses 2016; 8:E154. [PMID: 27271653 PMCID: PMC4926174 DOI: 10.3390/v8060154] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/12/2016] [Accepted: 05/20/2016] [Indexed: 02/06/2023] Open
Abstract
Viral infection initiates an array of changes in host gene expression. Many viruses dampen host protein expression and attempt to evade the host anti-viral defense machinery. Host gene expression is suppressed at several stages of host messenger RNA (mRNA) formation including selective degradation of translationally competent messenger RNAs. Besides mRNAs, host cells also express a variety of noncoding RNAs, including small RNAs, that may also be subject to inhibition upon viral infection. In this review we focused on different ways viruses antagonize coding and noncoding RNAs in the host cell to its advantage.
Collapse
Affiliation(s)
- Kristina M Herbert
- Department of Experimental Microbiology, Center for Scientific Research and Higher Education of Ensenada (CICESE), Ensenada, Baja California 22860, Mexico.
| | - Anita Nag
- Department of Chemistry, Florida A&M University, Tallahassee, FL 32307, USA.
| |
Collapse
|
78
|
Ho BC, Yang PC, Yu SL. MicroRNA and Pathogenesis of Enterovirus Infection. Viruses 2016; 8:v8010011. [PMID: 26751468 PMCID: PMC4728571 DOI: 10.3390/v8010011] [Citation(s) in RCA: 34] [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: 10/01/2015] [Revised: 12/04/2015] [Accepted: 12/18/2015] [Indexed: 12/15/2022] Open
Abstract
There are no currently available specific antiviral therapies for non-polio Enterovirus infections. Although several vaccines have entered clinical trials, the efficacy requires further evaluation, particularly for cross-strain protective activity. Curing patients with viral infections is a public health problem due to antigen alterations and drug resistance caused by the high genomic mutation rate. To conquer these limits in the development of anti-Enterovirus treatments, a comprehensive understanding of the interactions between Enterovirus and host cells is urgently needed. MicroRNA (miRNA) constitutes the biggest family of gene regulators in mammalian cells and regulates almost a half of all human genes. The roles of miRNAs in Enterovirus pathogenesis have recently begun to be noted. In this review, we shed light on recent advances in the understanding of Enterovirus infection-modulated miRNAs. The impacts of altered host miRNAs on cellular processes, including immune escape, apoptosis, signal transduction, shutdown of host protein synthesis and viral replication, are discussed. Finally, miRNA-based medication provides a promising strategy for the development of antiviral therapy.
Collapse
Affiliation(s)
- Bing-Ching Ho
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, No. 1 Chang-Te Street, Taipei 10048, Taiwan.
- Center of Genomic Medicine, National Taiwan University, Taipei 10048, Taiwan.
| | - Pan-Chyr Yang
- Center of Genomic Medicine, National Taiwan University, Taipei 10048, Taiwan.
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10048, Taiwan.
- Institute of Biomedical Sciences, Academia Sinica, Taipei 10048, Taiwan.
| | - Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, No. 1 Chang-Te Street, Taipei 10048, Taiwan.
- Center of Genomic Medicine, National Taiwan University, Taipei 10048, Taiwan.
- Center for Optoelectronic Biomedicine, College of Medicine, National Taiwan University, Taipei 10048, Taiwan.
- Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei 10048, Taiwan.
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei 10048, Taiwan.
| |
Collapse
|
79
|
Innate Immunity and Immune Evasion by Enterovirus 71. Viruses 2015; 7:6613-30. [PMID: 26694447 PMCID: PMC4690884 DOI: 10.3390/v7122961] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/16/2015] [Accepted: 12/09/2015] [Indexed: 12/23/2022] Open
Abstract
Enterovirus 71 (EV71) is a major infectious disease affecting millions of people worldwide and it is the main etiological agent for outbreaks of hand foot and mouth disease (HFMD). Infection is often associated with severe gastroenterological, pulmonary, and neurological diseases that are most prevalent in children. Currently, no effective vaccine or antiviral drugs exist against EV71 infection. A lack of knowledge on the molecular mechanisms of EV71 infection in the host and the virus-host interactions is a major constraint to developing specific antiviral strategies against this infection. Previous studies have identified and characterized the function of several viral proteins produced by EV71 that interact with the host innate immune proteins, including type I interferon signaling and microRNAs. These interactions eventually promote efficient viral replication and increased susceptibility to the disease. In this review we discuss the functions of EV71 viral proteins in the modulation of host innate immune responses to facilitate viral replication.
Collapse
|
80
|
Cleavage of DAP5 by coxsackievirus B3 2A protease facilitates viral replication and enhances apoptosis by altering translation of IRES-containing genes. Cell Death Differ 2015; 23:828-40. [PMID: 26586572 DOI: 10.1038/cdd.2015.145] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 09/17/2015] [Accepted: 09/25/2015] [Indexed: 12/22/2022] Open
Abstract
Cleavage of eukaryotic translation initiation factor 4G (eIF4G) by enterovirus proteases during infection leads to the shutoff of cellular cap-dependent translation, but does not affect the initiation of cap-independent translation of mRNAs containing an internal ribosome entry site (IRES). Death-associated protein 5 (DAP5), a structural homolog of eIF4G, is a translation initiation factor specific for IRES-containing mRNAs. Coxsackievirus B3 (CVB3) is a positive single-stranded RNA virus and a primary causal agent of human myocarditis. Its RNA genome harbors an IRES within the 5'-untranslated region and is translated by a cap-independent, IRES-driven mechanism. Previously, we have shown that DAP5 is cleaved during CVB3 infection. However, the protease responsible for cleavage, cleavage site and effects on the translation of target genes during CVB3 infection have not been investigated. In the present study, we demonstrated that viral protease 2A but not 3C is responsible for DAP5 cleavage, generating 45- and 52-kDa N- (DAP5-N) and C-terminal (DAP5-C) fragments, respectively. By site-directed mutagenesis, we found that DAP5 is cleaved at amino acid G434. Upon cleavage, DAP5-N largely translocated to the nucleus at the later time points of infection, whereas the DAP5-C largely remained in the cytoplasm. Overexpression of these DAP5 truncates demonstrated that DAP5-N retained the capability of initiating IRES-driven translation of apoptosis-associated p53, but not the prosurvival Bcl-2 (B-cell lymphoma 2) when compared with the full-length DAP5. Similarly, DAP5-N expression promoted CVB3 replication and progeny release; on the other hand, DAP5-C exerted a dominant-negative effect on cap-dependent translation. Taken together, viral protease 2A-mediated cleavage of DAP5 results in the production of two truncates that exert differential effects on protein translation of the IRES-containing genes, leading to enhanced host cell death.
Collapse
|
81
|
Host MicroRNA miR-197 Plays a Negative Regulatory Role in the Enterovirus 71 Infectious Cycle by Targeting the RAN Protein. J Virol 2015; 90:1424-38. [PMID: 26581983 DOI: 10.1128/jvi.02143-15] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 11/10/2015] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Enterovirus 71 (EV71), a member of Picornaviridae, is associated with severe central nervous system complications. In this study, we identified a cellular microRNA (miRNA), miR-197, whose expression was downregulated by viral infection in a time-dependent manner. In miR-197 mimic-transfected cells, EV71 replication was inhibited, whereas the internal ribosome entry site (IRES) activity was decreased in EV71 strains with or without predicted miR-197 target sites, indicating that miR-197 targets host proteins to modulate viral replication. We thus used a quantitative proteomics approach, aided by the TargetScan algorithm, to identify putative target genes of miR-197. Among them, RAN was selected and validated as a genuine target in a 3' untranslated region (UTR) reporter assay. Reduced production of RAN by RNA interference markedly reduced the synthesis of EV71-encoded viral proteins and virus titers. Furthermore, reintroduction of nondegradable RAN into these knockdown cells rescued viral protein synthesis. miR-197 levels were modulated by EV71 to maintain RAN mRNA translatability at late times postinfection since we demonstrated that cap-independent translation exerted by its intrinsic IRES activity was occurring at times when translation attenuation was induced by EV71. EV71-induced downregulation of miR-197 expression increased the expression of RAN, which supported the nuclear transport of the essential viral proteins 3D/3CD and host protein hnRNP K for viral replication. Our data suggest that downregulation of cellular miRNAs may constitute a newly identified mechanism that sustains the expression of host proteins to facilitate viral replication. IMPORTANCE Enterovirus 71 (EV71) is a picornavirus with a positive-sense single-stranded RNA that globally inhibits the cellular translational system, mainly by cleaving cellular eukaryotic translation initiation factor 4G (eIF4G) and poly(A)-binding protein (PABP), which inhibits the association of the ribosome with the host capped mRNA. We used a microRNA (miRNA) microarray chip to identify the host miRNA 197 (miR-197) that was downregulated by EV71. We also used quantitative mass spectrometry and a target site prediction tool to identify the miR-197 target genes. During viral infection, the expression of the target protein RAN was upregulated considerably, and there was a parallel downregulation of miR-197. The nuclear transport of viral 3D/3CD protein and of the host proteins involved in viral replication proceeded in an RAN-dependent manner. We have identified a new mechanism in picornavirus through which EV71-induced cellular miRNA downregulation can regulate host protein levels to facilitate viral replication.
Collapse
|
82
|
Wu J, Shen L, Chen J, Xu H, Mao L. The role of microRNAs in enteroviral infections. Braz J Infect Dis 2015; 19:510-6. [PMID: 26342975 PMCID: PMC9427576 DOI: 10.1016/j.bjid.2015.06.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 05/29/2015] [Accepted: 06/04/2015] [Indexed: 01/22/2023] Open
Abstract
The genus Enterovirus, a member of the Picornavirus family, are RNA viruses that can cause poliomyelitis, hand-food-mouth disease, viral meningitis or meningoencephalitis, viral myocarditis and so on. MicroRNAs are a class of highly conserved, small noncoding RNAs recognized as important regulators of gene expression. Recent studies found that MicroRNAs play a significant role in the infection of Enterovirus, such as enterovirus 71, coxsackievirus B3 and other Enterovirus. Enteroviral infection can alter the expression of cellular MicroRNAs, and cellular MicroRNAs can modulate viral pathogenesis and replication by regulating the expression level of viral or host's genes. Herein, this review summarizes the role of MicroRNAs in enteroviral infection.
Collapse
Affiliation(s)
- Jing Wu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Li Shen
- Department of Clinical Laboratory, Zhenjiang Center for Disease Control and Prevention, Zhenjiang, Jiangsu Province, China
| | - Jianguo Chen
- Department of Clinical Laboratory, Zhenjiang First People's Hospital, Jiangsu Province, China
| | - Huaxi Xu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Lingxiang Mao
- Department of Clinical Laboratory, Zhenjiang Center for Disease Control and Prevention, Zhenjiang, Jiangsu Province, China; School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province, China.
| |
Collapse
|
83
|
Garmaroudi FS, Marchant D, Hendry R, Luo H, Yang D, Ye X, Shi J, McManus BM. Coxsackievirus B3 replication and pathogenesis. Future Microbiol 2015; 10:629-53. [DOI: 10.2217/fmb.15.5] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
ABSTRACT Viruses such as coxsackievirus B3 (CVB3) are entirely host cell-dependent parasites. Indeed, they must cleverly exploit various compartments of host cells to complete their life cycle, and consequently launch disease. Evolution has equipped this pico-rna-virus, CVB3, to use different strategies, including CVB3-induced direct damage to host cells followed by a host inflammatory response to CVB3 infection, and cell death to super-additively promote target organ tissue injury, and dysfunction. In this update, the patho-stratagems of CVB3 are explored from molecular, and systems-level approaches. In summarizing recent developments in this field, we focus particularly on mechanisms by which CVB3 can harness different host cell processes including kinases, host cell-killing and cell-eating machineries, matrix metalloproteinases and miRNAs to promote disease.
Collapse
Affiliation(s)
- Farshid S Garmaroudi
- UBC James Hogg Research Centre, Institute for Heart & Lung Health, St. Paul's Hospital, University of British Columbia, Vancouver, BC, V6Z, Canada
| | - David Marchant
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Reid Hendry
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Honglin Luo
- UBC James Hogg Research Centre, Institute for Heart & Lung Health, St. Paul's Hospital, University of British Columbia, Vancouver, BC, V6Z, Canada
| | - Decheng Yang
- UBC James Hogg Research Centre, Institute for Heart & Lung Health, St. Paul's Hospital, University of British Columbia, Vancouver, BC, V6Z, Canada
| | - Xin Ye
- UBC James Hogg Research Centre, Institute for Heart & Lung Health, St. Paul's Hospital, University of British Columbia, Vancouver, BC, V6Z, Canada
| | - Junyan Shi
- UBC James Hogg Research Centre, Institute for Heart & Lung Health, St. Paul's Hospital, University of British Columbia, Vancouver, BC, V6Z, Canada
| | - Bruce M McManus
- UBC James Hogg Research Centre, Institute for Heart & Lung Health, St. Paul's Hospital, University of British Columbia, Vancouver, BC, V6Z, Canada
- Centre of Excellence for Prevention of Organ Failure, Vancouver, BC, Canada
| |
Collapse
|
84
|
Kuehl U, Lassner D, Gast M, Stroux A, Rohde M, Siegismund C, Wang X, Escher F, Gross M, Skurk C, Tschoepe C, Loebel M, Scheibenbogen C, Schultheiss HP, Poller W. Differential Cardiac MicroRNA Expression Predicts the Clinical Course in Human Enterovirus Cardiomyopathy. Circ Heart Fail 2015; 8:605-18. [PMID: 25761932 DOI: 10.1161/circheartfailure.114.001475] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 03/09/2015] [Indexed: 01/03/2023]
Abstract
BACKGROUND Investigation of disease pathogenesis confined to protein-coding regions of the genome may be incomplete because many noncoding variants are associated with disease. We aimed to identify novel predictive markers for the course of enterovirus (CVB3) cardiomyopathy by screening for noncoding elements influencing the grossly different antiviral capacity of individual patients. METHODS AND RESULTS Transcriptome mapping of CVB3 cardiomyopathy patients revealed distinctive cardiac microRNA (miR) patterns associated with spontaneous virus clearance and recovery (CVB3-ELIM) versus virus persistence and progressive clinical deterioration (CVB3-PERS). Profiling of protein-coding genes and 754 miRs in endomyocardial biopsies of test cohorts was performed at their initial presentation, and those spontaneously eliminating the virus were compared with those with virus persistence on follow-up. miR profiling revealed highly significant differences in cardiac levels of 16 miRs, but not of protein-coding genes. Evaluation of this primary distinctive miR pattern in validation cohorts, and multivariate receiver operating characteristic curve analysis, confirmed this pattern as highly predictive for disease course (area under the curve, 0.897±0.071; 95% confidence interval, 0.758-1.000). Eight miRs were strongly induced in CVB3-PERS (miRs 135b, 155, 190, 422a, 489, 590, 601, 1290), but undetectable in CVB3-ELIM or controls. They are predicted to target multiple immune response genes, and 2 of these were confirmed by antisense-mediated ablation of miRs 135b, 190, and 422a in the monocytic THP-1 cell line. CONCLUSIONS An immediate clinical application of the data is cardiac miR profiling to assess the risk of virus persistence and progressive clinical deterioration in CVB3 cardiomyopathy. Patients at risk are eligible for immediate antiviral therapy to minimize irreversible cardiac damage.
Collapse
Affiliation(s)
- Uwe Kuehl
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Dirk Lassner
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Martina Gast
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Andrea Stroux
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Maria Rohde
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Christine Siegismund
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Xiaomin Wang
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Felicitas Escher
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Michael Gross
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Carsten Skurk
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Carsten Tschoepe
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Madlen Loebel
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Carmen Scheibenbogen
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Heinz-Peter Schultheiss
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund)
| | - Wolfgang Poller
- From the Department of Cardiology and Pneumology (U.K., M.G., X.W., F.E., M.G., C. Skurk, C.T., H.-P.S., W.P.), Institute for Biometry and Clinical Epidemiology, Campus Benjamin Franklin (A.S.), Institute for Medical Immunology, Campus Virchow Klinikum (M.L., C. Scheibenbogen), Berlin Center for Regenerative Therapies (BCRT) (C.T., M.L., C. Scheibenbogen, W.P.), Charité-Universitätsmedizin Berlin, Berlin, Germany; and Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany (D.L., M.R., C. Siegismund).
| |
Collapse
|
85
|
Leong SY, Ong BKT, Chu JJH. The role of Misshapen NCK-related kinase (MINK), a novel Ste20 family kinase, in the IRES-mediated protein translation of human enterovirus 71. PLoS Pathog 2015; 11:e1004686. [PMID: 25747578 PMCID: PMC4352056 DOI: 10.1371/journal.ppat.1004686] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 01/16/2015] [Indexed: 11/18/2022] Open
Abstract
Human Enterovirus 71 (EV71) commonly causes Hand, Foot and Mouth Disease in young children, and occasional occurrences of neurological complications can be fatal. In this study, a high-throughput cell-based screening on the serine/threonine kinase siRNA library was performed to identify potential antiviral agents against EV71 replication. Among the hits, Misshapen/NIKs-related kinase (MINK) was selected for detailed analysis due to its strong inhibitory profile and novelty. In the investigation of the stage at which MINK is involved in EV71 replication, virus RNA transfection in MINK siRNA-treated cells continued to cause virus inhibition despite bypassing the normal entry pathway, suggesting its involvement at the post-entry stage. We have also shown that viral RNA and protein expression level was significantly reduced upon MINK silencing, suggesting its involvement in viral protein synthesis which feeds into viral RNA replication process. Through proteomic analysis and infection inhibition assay, we found that the activation of MINK was triggered by early replication events, instead of the binding and entry of the virus. Proteomic analysis on the activation profile of p38 Mitogen-activated Protein Kinase (MAPK) indicated that the phosphorylation of p38 MAPK was stimulated by EV71 infection upon MINK activation. Luciferase reporter assay further revealed that the translation efficiency of the EV71 internal ribosomal entry site (IRES) was reduced after blocking the MINK/p38 MAPK pathway. Further investigation on the effect of MINK silencing on heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) localisation demonstrated that cytoplasmic relocalisation of hnRNP A1 upon EV71 infection may be facilitated via the MINK/p38 MAPK pathway which then positively regulates the translation of viral RNA transcripts. These novel findings hence suggest that MINK plays a functional role in the IRES-mediated translation of EV71 viral RNA and may provide a potential target for the development of specific antiviral strategies against EV71 infection.
Collapse
Affiliation(s)
- Shi Yun Leong
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Bryan Kit Teck Ong
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Justin Jang Hann Chu
- Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
86
|
Song Y, Cheng X, Yang X, Zhao R, Wang P, Han Y, Luo Z, Cao Y, Zhu C, Xiong Y, Liu Y, Wu K, Wu J. Early growth response-1 facilitates enterovirus 71 replication by direct binding to the viral genome RNA. Int J Biochem Cell Biol 2015; 62:36-46. [PMID: 25724735 DOI: 10.1016/j.biocel.2015.02.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 02/05/2015] [Accepted: 02/17/2015] [Indexed: 01/14/2023]
Abstract
Enterovirus 71 (EV71) infections can cause hand, foot and mouth disease (HFMD), meningoencephalitis, neonatal sepsis, and even fatal encephalitis in children. Unfortunately, there is currently no effective treatment for EV71 infection due to the lack of understanding of viral replication and infection; and viral infections have emerged as an imperative global hazard. Thus, it is extremely important to understand the mechanism of EV71 replication in order to prevent and control the diseases associated with EV71 infections. Early growth response-1 (EGR1) is a multifunctional transcription factor that regulates diverse biological functions, including inflammation, apoptosis, differentiation, tumorigenesis, and even viral infection. Here, we provide new insight into the role of EV71 infection in regulating EGR1 production; and reveal a novel mechanism by which EGR1 facilitates EV71 replication. We demonstrate that EV71 activates EGR1 expression during infection by stimulating the protein kinase A/protein kinase Cɛ/phosphoinositide 3-kinase/Akt (PKA/PKCɛ/PI3K/Akt) cascade. We further reveal that EV71-activated EGR1, in turn, regulates the internal ribosomal entry site (IRES) of EV71 to enhance viral replication. In addition, EGR1 facilitates EV71 replication by binding directly to stem-loops I and IV of EV71 5'-untranslated region (5'UTR) with its first two zinc fingers. Moreover, EGR1 protein co-localizes with EV71 RNA in the cytoplasm of infected cells to facilitate viral replication. Our results reveal an important new role of EGR1 in viral infection, provide new insight into the novel mechanism underlying the regulation of EV71 replication, and suggest a potential application of EGR1 in the control of EV71 infection.
Collapse
Affiliation(s)
- Yu Song
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xin Cheng
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Xiaoxia Yang
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Rong Zhao
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Peili Wang
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yang Han
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zhen Luo
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yanhua Cao
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Chengliang Zhu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Ying Xiong
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yingle Liu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Kailang Wu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jianguo Wu
- State Key Laboratory of Virology and College of Life Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
87
|
Sharma N, Verma R, Kumawat KL, Basu A, Singh SK. miR-146a suppresses cellular immune response during Japanese encephalitis virus JaOArS982 strain infection in human microglial cells. J Neuroinflammation 2015; 12:30. [PMID: 25889446 PMCID: PMC4355369 DOI: 10.1186/s12974-015-0249-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 01/15/2015] [Indexed: 12/28/2022] Open
Abstract
Background Japanese encephalitis virus (JEV) is the causative agent of Japanese encephalitis which is more prevalent in South and Southeast Asia. JEV is a neurotropic virus which infiltrates into the brain through vascular endothelial cells. JEV infects neurons and microglial cells which causes neuronal damage and inflammation. However, JEV also evades the cellular immune response to survive in host cells. Viruses are known to modulate the expression of microRNAs, which in turn modulate cellular immune response by targeting expression of antiviral genes. The aim of this study is to understand the anti-inflammatory role of miR-146a during JEV infection, which facilitates immune evasion. Methods Human brain microglial cells (CHME3) were infected by JEV: JaOArS982 and P20778 strain, and expression of miR-146a were analyzed. Overexpression and knockdown studies of miR-146a were done to see the effect on NF-κB pathway and antiviral Jak-STAT pathway. Regulatory role of miR-146a on expression of interferon-stimulated genes was determined by real-time PCR and luciferase assays. Results JEV infection elevated the expression of miR-146a in JaOArS982 strain which caused downregulation of TRAF6, IRAK1, IRAK2, and STAT1 genes. Exogenous overexpression of miR-146a led to suppression of NF-κB activation and abrogation of Jak-STAT pathway upon JEV infection which led to downregulation of interferon-stimulated genes (IFIT-1 and IFIT-2) and facilitated viral replication. JEV infection initially upregulated cytokine production and activated STAT1 activity but STAT1 levels reduced at later time point, which led to the downregulation of interferon-stimulated genes. Conclusion Upregulation of miR-146a by JEV JaOArS982 strain leads to suppression of NF-κB activity and disruption of antiviral Jak-STAT signaling which helps the virus to evade the cellular immune response. This effect of JEV infection on miR-146a expression was found to be strain specific. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0249-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nikhil Sharma
- Laboratory of Neurovirology and Inflammation Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, 500007, Hyderabad, AP, India.
| | - Ruhi Verma
- Laboratory of Neurovirology and Inflammation Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, 500007, Hyderabad, AP, India.
| | | | - Anirban Basu
- National Brain Research Centre, Haryana-122050, Manesar, Haryana, India.
| | - Sunit K Singh
- Laboratory of Neurovirology and Inflammation Biology, CSIR-Centre for Cellular and Molecular Biology (CCMB), Uppal Road, 500007, Hyderabad, AP, India. .,Current Affiliation: Laboratory of Human Molecular Virology and Immunology, Molecular Biology Unit, Faculty of Medicine, Institute of Medical Sciences (IMS), Banaras Hindu University (BHU), 221005, Varanasi, India.
| |
Collapse
|
88
|
eIF4E as a control target for viruses. Viruses 2015; 7:739-50. [PMID: 25690796 PMCID: PMC4353914 DOI: 10.3390/v7020739] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/06/2015] [Accepted: 02/11/2015] [Indexed: 01/04/2023] Open
Abstract
Translation is a complex process involving diverse cellular proteins, including the translation initiation factor eIF4E, which has been shown to be a protein that is a point for translational regulation. Viruses require components from the host cell to complete their replication cycles. Various studies show how eIF4E and its regulatory cellular proteins are manipulated during viral infections. Interestingly, viral action mechanisms in eIF4E are diverse and have an impact not only on viral protein synthesis, but also on other aspects that are important for the replication cycle, such as the proliferation of infected cells and stimulation of viral reactivation. This review shows how some viruses use eIF4E and its regulatory proteins for their own benefit in order to spread themselves.
Collapse
|
89
|
Chang YL, Ho BC, Sher S, Yu SL, Yang PC. miR-146a and miR-370 coordinate enterovirus 71-induced cell apoptosis through targeting SOS1 and GADD45β. Cell Microbiol 2015; 17:802-18. [PMID: 25469565 DOI: 10.1111/cmi.12401] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 11/20/2014] [Accepted: 11/24/2014] [Indexed: 12/22/2022]
Abstract
Enterovirus 71 (EV71) is an emerging life-threatening pathogen particularly in the Asia-Pacific region. The major pathogenic feature in EV71-infected cells is apoptosis. However, which molecular mechanism mainly contributes to EV71-induced apoptosis is not investigated thoroughly. MicroRNAs (MiRNAs), the newly discovered molecules, govern a wide range of biological functions through post-transcriptional regulation including viral pathogenesis. Whether miRNAs and messenger RNAs (mRNAs) coordinate to trigger host cell apoptosis in EV71 infection was investigated in this study. We conducted an apoptosis-oriented approach using both mRNA and miRNA profiling and bioinformatic analysis. We identified two major apoptosis-associated signalling pathways, Bcl2 antagonist of cell death (BAD) phosphorylation and p53-dependent apoptosis pathways, in which Son of sevenless homolog 1 (SOS1) and Growth arrest and DNA damage-inducible protein 45β (GADD45β) were predicted as targets of miR-146a and miR-370 respectively. Luciferase reporter assays and Western blots demonstrated the negative regulation between miR-146a and SOS1 and between miR-370 and GADD45β. Silencing of miR-146a restored SOS1 expression and partially attenuated EV71 infection-induced apoptosis. Conversely, ectopic expression of miR-370 decreased virus infection-induced GADD45β expression and also diminished apoptosis. Finally, the transfection of antagomiR-146a and miR-370 contributed to attenuating EV71 infection-induced apoptosis. Herein we clearly demonstrate that EV71-induced cell apoptosis is partly governed by altered miRNAs.
Collapse
Affiliation(s)
- Ya-Ling Chang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bing-Ching Ho
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.,NTU Center for Genomic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Singh Sher
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.,NTU Center for Genomic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Department of Pathology and Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Center for Optoelectronic Biomedicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pan-Chyr Yang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,NTU Center for Genomic Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
90
|
Loveday EK, Diederich S, Pasick J, Jean F. Human microRNA-24 modulates highly pathogenic avian-origin H5N1 influenza A virus infection in A549 cells by targeting secretory pathway furin. J Gen Virol 2015; 96:30-39. [DOI: 10.1099/vir.0.068585-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Emma-Kate Loveday
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Sandra Diederich
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - John Pasick
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3M4, Canada
| | - François Jean
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
91
|
Tavanez JP, Quina AS, Cunha C. Virus and noncoding RNAs: stars in the host–virus interaction game. Future Virol 2014. [DOI: 10.2217/fvl.14.84] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
ABSTRACT: In the past few years, noncoding RNAs (ncRNAs) have emerged as key modulators of the transcriptional and post-transcriptional control of a variety of cellular processes such as development, signaling, homeostasis and oncogenesis. Like their host cells, many viruses produce ncRNAs. During viral infection, and in order to establish persistent life-long infection of the host, viruses express both protein-coding and noncoding genes, modulating the cellular environment to favor infection. Given their limited genomic capacity, viruses evolved or acquired ncRNAs only if advantageous, either by enhancing the viral life cycle or assisting the virus in immune evasion of the host's response to infection. With variable length, structure, number, abundance and protein-binding partners, viral ncRNAs show specificity and diversity with respect to time of expression during the different stages of the virus life cycle and viral infection. Here, we review our current knowledge on the RNA-based mechanisms that regulate host–virus interaction focusing on viral ncRNAs and cellular ncRNAs modulated by viruses upon infection.
Collapse
Affiliation(s)
- João Paulo Tavanez
- Unidade de Microbiologia Médica, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Ana Sofia Quina
- Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
- Centro de Estudos do Ambiente e do Mar, Aveiro, Portugal
| | - Celso Cunha
- Unidade de Microbiologia Médica, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisboa, Portugal
| |
Collapse
|
92
|
Prediction of signaling pathways involved in enterovirus 71 infection by algorithm analysis based on miRNA profiles and their target genes. Arch Virol 2014; 160:173-82. [PMID: 25287131 DOI: 10.1007/s00705-014-2249-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 09/29/2014] [Indexed: 12/12/2022]
Abstract
Enterovirus 71 (EV71) causes major outbreaks of hand, foot, and mouth disease. Host factors and signaling pathways exhibit important functions in the EV71 life cycle. We conducted algorithm analysis based on miRNA profiles and their target genes to identify the miRNAs and downstream signaling pathways involved in EV71 infection. The miRNA profiles of human rhabdomyosarcoma cells treated with interferon (IFN-)-α or IFN-γ were compared with those of cells infected with EV71. Genes targeted by differentially expressed miRNAs were identified and assigned to different signaling pathways according to public databases. The results showed that host miRNAs specifically responded to the viral infection and IFN treatment. Some miRNAs, including miR-124 and miR-491-3p, were regulated in opposite manners by the IFNs and EV71. Some signaling pathways regulated by both EV71 infection and IFN treatment were also predicted. These pathways included axon guidance, Wingless/Int1 (Wnt) signaling cascade, platelet-derived growth factor receptor (PDGFR)/PDGF, phosphatidylinositol 3-kinase (PI3K), Jun N-terminal kinase (JNK)/mitogen-activated protein kinase (MAPK), transforming growth factor-beta receptor (TGF-βR)/TGF-β, SMAD2/3, insulin/insulin-like growth factor (IGF), bone morphogenetic protein (BMP), CDC42, ERB1, hepatocyte growth factor receptor (c-Met), eukaryotic translation initiation factor 4E (eIF4E), protein kinase A (PKA), and IFN-γ pathways. The identified miRNA and downstream signaling pathways would help to elucidate the interaction between the virus and the host. The genomics method using algorithm analysis also provided a new way to investigate the host factors and signaling pathways critical for viral replication.
Collapse
|
93
|
Droplet digital PCR as a useful tool for the quantitative detection of Enterovirus 71. J Virol Methods 2014; 207:200-3. [DOI: 10.1016/j.jviromet.2014.07.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 12/15/2022]
|
94
|
miR-27a suppresses EV71 replication by directly targeting EGFR. Virus Genes 2014; 49:373-82. [PMID: 25212431 DOI: 10.1007/s11262-014-1114-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 09/04/2014] [Indexed: 12/11/2022]
Abstract
Enterovirus 71 (EV71), a major causative agent of hand, foot, and mouth disease, has broken out several times and was accompanied by neurological disease. microRNAs, a class of small non-coding RNAs that are approximately 20 nucleotides long, play important roles in the regulation of various biological processes, including antiviral defense. However, the roles of miRNAs in EV71 replication and pathogenesis are not well understood. In this study, we found that the expression of miR-27a was significantly decreased in EV71-infected cells. Interestingly, the over-expression of miR-27a could inhibit EV71 replication, as measured by virus titration, qPCR, and Western blotting. We identified EGFR mRNA is a bona fide target of miR-27a by computational analysis and luciferase reporter assays. Furthermore, miR-27a could decrease EGFR expression, as measured by qPCR and Western blotting. Moreover, the inhibition of EGFR expression by miR-27a decreased the phosphorylation of Akt and ERK, which facilitate EV71 replication. These results suggest that miR-27a may have antiviral activity against EV71 by inhibiting EGFR.
Collapse
|
95
|
Lui YLE, Tan TL, Woo WH, Timms P, Hafner LM, Tan KH, Tan EL. Enterovirus71 (EV71) utilise host microRNAs to mediate host immune system enhancing survival during infection. PLoS One 2014; 9:e102997. [PMID: 25047717 PMCID: PMC4105423 DOI: 10.1371/journal.pone.0102997] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 06/26/2014] [Indexed: 12/24/2022] Open
Abstract
Hand, Foot and Mouth Disease (HFMD) is a self-limiting viral disease that mainly affects infants and children. In contrast with other HFMD causing enteroviruses, Enterovirus71 (EV71) has commonly been associated with severe clinical manifestation leading to death. Currently, due to a lack in understanding of EV71 pathogenesis, there is no antiviral therapeutics for the treatment of HFMD patients. Therefore the need to better understand the mechanism of EV71 pathogenesis is warranted. We have previously reported a human colorectal adenocarcinoma cell line (HT29) based model to study the pathogenesis of EV71. Using this system, we showed that knockdown of DGCR8, an essential cofactor for microRNAs biogenesis resulted in a reduction of EV71 replication. We also demonstrated that there are miRNAs changes during EV71 pathogenesis and EV71 utilise host miRNAs to attenuate antiviral pathways during infection. Together, data from this study provide critical information on the role of miRNAs during EV71 infection.
Collapse
Affiliation(s)
- Yan Long Edmund Lui
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, Singapore
- Centre for Biomedical and Life Sciences, Singapore Polytechnic, Singapore, Singapore
- * E-mail: (YLEL) (YL); (LMH) (LH)
| | - Tuan Lin Tan
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, Singapore
| | - Wee Hong Woo
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, Singapore
| | - Peter Timms
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Louise Marie Hafner
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
- * E-mail: (YLEL) (YL); (LMH) (LH)
| | - Kian Hwa Tan
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, Singapore
| | - Eng Lee Tan
- Centre for Biomedical and Life Sciences, Singapore Polytechnic, Singapore, Singapore
- Department of Paediatrics, University Children’s Medical Institute, National University Hospital, Singapore, Singapore
| |
Collapse
|
96
|
Au HHT, Jan E. Novel viral translation strategies. WILEY INTERDISCIPLINARY REVIEWS-RNA 2014; 5:779-801. [PMID: 25045163 PMCID: PMC7169809 DOI: 10.1002/wrna.1246] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 05/03/2014] [Accepted: 05/08/2014] [Indexed: 01/06/2023]
Abstract
Viral genomes are compact and encode a limited number of proteins. Because they do not encode components of the translational machinery, viruses exhibit an absolute dependence on the host ribosome and factors for viral messenger RNA (mRNA) translation. In order to recruit the host ribosome, viruses have evolved unique strategies to either outcompete cellular transcripts that are efficiently translated by the canonical translation pathway or to reroute translation factors and ribosomes to the viral genome. Furthermore, viruses must evade host antiviral responses and escape immune surveillance. This review focuses on some recent major findings that have revealed unconventional strategies that viruses utilize, which include usurping the host translational machinery, modulating canonical translation initiation factors to specifically enhance or repress overall translation for the purpose of viral production, and increasing viral coding capacity. The discovery of these diverse viral strategies has provided insights into additional translational control mechanisms and into the viral host interactions that ensure viral protein synthesis and replication. WIREs RNA 2014, 5:779–801. doi: 10.1002/wrna.1246 This article is categorized under:
Translation > Translation Mechanisms Translation > Translation Regulation
Collapse
Affiliation(s)
- Hilda H T Au
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
97
|
Liu YC, Kuo RL, Lin JY, Huang PN, Huang Y, Liu H, Arnold JJ, Chen SJ, Wang RYL, Cameron CE, Shih SR. Cytoplasmic viral RNA-dependent RNA polymerase disrupts the intracellular splicing machinery by entering the nucleus and interfering with Prp8. PLoS Pathog 2014; 10:e1004199. [PMID: 24968230 PMCID: PMC4072778 DOI: 10.1371/journal.ppat.1004199] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 05/05/2014] [Indexed: 11/25/2022] Open
Abstract
The primary role of cytoplasmic viral RNA-dependent RNA polymerase (RdRp) is viral genome replication in the cellular cytoplasm. However, picornaviral RdRp denoted 3D polymerase (3Dpol) also enters the host nucleus, where its function remains unclear. In this study, we describe a novel mechanism of viral attack in which 3Dpol enters the nucleus through the nuclear localization signal (NLS) and targets the pre-mRNA processing factor 8 (Prp8) to block pre-mRNA splicing and mRNA synthesis. The fingers domain of 3Dpol associates with the C-terminal region of Prp8, which contains the Jab1/MPN domain, and interferes in the second catalytic step, resulting in the accumulation of the lariat form of the splicing intermediate. Endogenous pre-mRNAs trapped by the Prp8-3Dpol complex in enterovirus-infected cells were identified and classed into groups associated with cell growth, proliferation, and differentiation. Our results suggest that picornaviral RdRp disrupts pre-mRNA splicing processes, that differs from viral protease shutting off cellular transcription and translation which contributes to the pathogenesis of viral infection. RNA-dependent RNA polymerase (RdRp) is an enzyme that catalyzes the replication from an RNA template and is encoded in the genomes of all RNA viruses. RNA viruses in general replicate in cytoplasm and interfere host cellular gene expression by utilizing proteolytic destruction of cellular targets as the primary mechanism. However, several cytoplasmic RNA viral proteins have been found in the nucleus. What do they do in the nucleus? This study utilized picornaviral polymerase to probe the function of RdRp in the nucleus. Our findings reveal a novel mechanism of viruses attacking hosts whereby picornaviral 3D polymerase (3Dpol) enters the nucleus and targets the central pre-mRNA processing factor 8 (Prp8) to block pre-mRNA splicing and mRNA synthesis. The 3Dpol inhibits the second catalytic step of the splicing process, resulting in the accumulation of the lariat-form and the reduction of the mRNA. These results provide new insights into the strategy of a cytoplasmic RNA virus attacking host cell, that differs from viral shutting off cellular transcription and translation which contributes to the viral pathogenesis. To our knowledge, this study shows for the first time that a cytoplasmic RNA virus uses its polymerase to alter cellular gene expression by hijacking the splicing machinery.
Collapse
Affiliation(s)
- Yen-Chin Liu
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Rei-Lin Kuo
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Jing-Yi Lin
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, Taiwan
| | - Peng-Nien Huang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Yi Huang
- Molecular Medicine Research Center, Chang Gung University, Tao-Yuan, Taiwan
| | - Hsuan Liu
- Molecular Medicine Research Center, Chang Gung University, Tao-Yuan, Taiwan
| | - Jamine J. Arnold
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Shu-Jen Chen
- Molecular Medicine Research Center, Chang Gung University, Tao-Yuan, Taiwan
| | - Robert Yung-Liang Wang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Biomedical Sciences and Graduate Institutes of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Craig E. Cameron
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- Clinical Virology Laboratory, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
- * E-mail:
| |
Collapse
|
98
|
Abstract
microRNAs (miRNAs) are a subtype of short, endogenous, and non-coding RNAs, which post-transcriptionally regulate gene expression. The miRNA-mediated gene silencing mechanism is involved in a wide spectrum of biological processes, such as cellular proliferation, differentiation, and immune responses. Picornaviridae is a large family of RNA viruses, which includes a number of causative agents of many human and animal diseases viz., poliovirus, foot-and-mouth disease virus (FMDV), and coxsackievirus B3 (CVB3). Accumulated evidences have demonstrated that replication of picornaviruses can be regulated by miRNAs and picornaviral infections can alter the expression of cellular miRNAs. Herein, we outline the intricate interactions between miRNAs and picornaviral infections.
Collapse
Affiliation(s)
- Miao Wang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouse Disease Reference Laboratory, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, Gansu, China
| | - Zeqian Gao
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouse Disease Reference Laboratory, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, Gansu, China
| | - Li Pan
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouse Disease Reference Laboratory, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, Gansu, China
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouse Disease Reference Laboratory, Lanzhou Veterinary Research Institute, CAAS, Lanzhou, Gansu, China
| |
Collapse
|
99
|
Vela EM, Kasoji MD, Wendling MQ, Price JA, Knostman KAB, Bresler HS, Long JP. MicroRNA expression in mice infected with seasonal H1N1, swine H1N1 or highly pathogenic H5N1. J Med Microbiol 2014; 63:1131-1142. [PMID: 24913561 DOI: 10.1099/jmm.0.067959-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Influenza virus infections in humans remain a healthcare concern, and the need for vaccines, therapeutics and prophylactics remains a high priority. Understanding the molecular events associated with influenza-virus-induced pathology may lead to the identification of clinical disease biomarkers and novel antiviral targets. MicroRNAs (miRNAs) are well-conserved endogenous non-coding RNAs known to regulate post-transcriptional gene expression as well as play a major role in many biological processes and pathways. Animal studies have demonstrated that miRNAs are involved in viral disease and controlling inflammation. In this study, we examined the differences in the miRNA expression profiles associated with the lung in mice infected with influenza viruses that varied in virulence and pathogenicity. A statistical model was employed that utilized changes in miRNA expression to identify the virus that was used to infect the mice. This study identified a unique fingerprint of viral pathogenicity associated with seasonal H1N1, swine H1N1 and highly pathogenic H5N1 in the mouse model, and may lead to the identification of novel therapeutic and prophylactic targets.
Collapse
Affiliation(s)
- Eric M Vela
- Battelle, 505 King Avenue, Columbus, OH, USA
| | | | | | | | | | | | | |
Collapse
|
100
|
Replication of many human viruses is refractory to inhibition by endogenous cellular microRNAs. J Virol 2014; 88:8065-76. [PMID: 24807715 DOI: 10.1128/jvi.00985-14] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The issue of whether viruses are subject to restriction by endogenous microRNAs (miRNAs) and/or by virus-induced small interfering RNAs (siRNAs) in infected human somatic cells has been controversial. Here, we address this question in two ways. First, using deep sequencing, we demonstrate that infection of human cells by the RNA virus dengue virus (DENV) or West Nile virus (WNV) does not result in the production of any virus-derived siRNAs or viral miRNAs. Second, to more globally assess the potential of small regulatory RNAs to inhibit virus replication, we used gene editing to derive human cell lines that lack a functional Dicer enzyme and that therefore are unable to produce miRNAs or siRNAs. Infection of these cells with a wide range of viruses, including DENV, WNV, yellow fever virus, Sindbis virus, Venezuelan equine encephalitis virus, measles virus, influenza A virus, reovirus, vesicular stomatitis virus, human immunodeficiency virus type 1, or herpes simplex virus 1 (HSV-1), failed to reveal any enhancement in the replication of any of these viruses, although HSV-1, which encodes at least eight Dicer-dependent viral miRNAs, did replicate somewhat more slowly in the absence of Dicer. We conclude that most, and perhaps all, human viruses have evolved to be resistant to inhibition by endogenous human miRNAs during productive replication and that dependence on a cellular miRNA, as seen with hepatitis C virus, is rare. How viruses have evolved to avoid inhibition by endogenous cellular miRNAs, which are generally highly conserved during metazoan evolution, remains to be determined. Importance: Eukaryotic cells express a wide range of small regulatory RNAs, including miRNAs, that have the potential to inhibit the expression of mRNAs that show sequence complementarity. Indeed, previous work has suggested that endogenous miRNAs have the potential to inhibit viral gene expression and replication. Here, we demonstrate that the replication of a wide range of pathogenic viruses is not enhanced in human cells engineered to be unable to produce miRNAs, indicating that viruses have evolved to be resistant to inhibition by miRNAs. This result is important, as it implies that manipulation of miRNA levels is not likely to prove useful in inhibiting virus replication. It also focuses attention on the question of how viruses have evolved to resist inhibition by miRNAs and whether virus mutants that have lost this resistance might prove useful, for example, in the development of attenuated virus vaccines.
Collapse
|